Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Immunity ; 57(7): 1514-1532.e15, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38788712

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.


Subject(s)
Immunogenic Cell Death , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Humans , Animals , Mice , Proteolysis/drug effects , Cell Line, Tumor , Signal Transduction/drug effects , Immunogenic Cell Death/drug effects , Necroptosis/drug effects , Necroptosis/immunology , Neoplasms/immunology , Neoplasms/drug therapy , Mice, Inbred C57BL , Antineoplastic Agents/pharmacology , Immunotherapy/methods
2.
Int J Mol Sci ; 23(20)2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36293328

ABSTRACT

Malignant mesothelioma is an aggressive tumour of the pleura (MPM) or peritoneum with a clinical presentation at an advanced stage of the disease. Current therapies only marginally improve survival and there is an urgent need to identify new treatments. Carcinoma-associated fibroblasts (CAFs) represent the main component of a vast stroma within MPM and play an important role in the tumour microenvironment. The influence of CAFs on cancer progression, aggressiveness and metastasis is well understood; however, the role of CAF-derived extracellular vesicles (CAF-EVs) in the promotion of tumour development and invasiveness is underexplored. We purified CAF-EVs from MPM-associated cells and healthy dermal human fibroblasts and examined their effect on cell proliferation and motility. The data show that exposure of healthy mesothelial cells to EVs derived from CAFs, but not from normal dermal human fibroblasts (NDHF) resulted in activating pro-oncogenic signalling pathways and increased proliferation and motility. Consistent with its role in suppressing Yes-Associated Protein (YAP) activation (which in MPM is a result of Hippo pathway inactivation), treatment with Simvastatin ameliorated the pro-oncogenic effects instigated by CAF-EVs by mechanisms involving both a reduction in EV number and changes in EV cargo. Collectively, these data determine the significance of CAF-derived EVs in mesothelioma development and progression and suggest new targets in cancer therapy.


Subject(s)
Cancer-Associated Fibroblasts , Extracellular Vesicles , Mesothelioma, Malignant , Mesothelioma , Humans , Cancer-Associated Fibroblasts/metabolism , YAP-Signaling Proteins , Cell Line, Tumor , Mesothelioma/pathology , Extracellular Vesicles/metabolism , Carcinogenesis/metabolism , Simvastatin , Tumor Microenvironment
3.
Sci Adv ; 9(30): eadg2829, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37494451

ABSTRACT

Cell death coordinates repair programs following pathogen attack and tissue injury. However, aberrant cell death can interfere with such programs and cause organ failure. Cellular FLICE-like inhibitory protein (cFLIP) is a crucial regulator of cell death and a substrate of Caspase-8. However, the physiological role of cFLIP cleavage by Caspase-8 remains elusive. Here, we found an essential role for cFLIP cleavage in restraining cell death in different pathophysiological scenarios. Mice expressing a cleavage-resistant cFLIP mutant, CflipD377A, exhibited increased sensitivity to severe acute respiratory syndrome coronavirus (SARS-CoV)-induced lethality, impaired skin wound healing, and increased tissue damage caused by Sharpin deficiency. In vitro, abrogation of cFLIP cleavage sensitizes cells to tumor necrosis factor(TNF)-induced necroptosis and apoptosis by favoring complex-II formation. Mechanistically, the cell death-sensitizing effect of the D377A mutation depends on glutamine-469. These results reveal a crucial role for cFLIP cleavage in controlling the amplitude of cell death responses occurring upon tissue stress to ensure the execution of repair programs.


Subject(s)
Apoptosis , Virus Diseases , Animals , Mice , Caspase 8/genetics , Skin/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Nat Commun ; 14(1): 3292, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37369658

ABSTRACT

Age-associated B cells (ABC) accumulate with age and in individuals with different immunological disorders, including cancer patients treated with immune checkpoint blockade and those with inborn errors of immunity. Here, we investigate whether ABCs from different conditions are similar and how they impact the longitudinal level of the COVID-19 vaccine response. Single-cell RNA sequencing indicates that ABCs with distinct aetiologies have common transcriptional profiles and can be categorised according to their expression of immune genes, such as the autoimmune regulator (AIRE). Furthermore, higher baseline ABC frequency correlates with decreased levels of antigen-specific memory B cells and reduced neutralising capacity against SARS-CoV-2. ABCs express high levels of the inhibitory FcγRIIB receptor and are distinctive in their ability to bind immune complexes, which could contribute to diminish vaccine responses either directly, or indirectly via enhanced clearance of immune complexed-antigen. Expansion of ABCs may, therefore, serve as a biomarker identifying individuals at risk of suboptimal responses to vaccination.


Subject(s)
COVID-19 , Immunity, Humoral , Humans , Immune Checkpoint Inhibitors , COVID-19 Vaccines , COVID-19/prevention & control , SARS-CoV-2 , Vaccination , Antigen-Antibody Complex , Antibodies, Viral
5.
Immunol Rev ; 224: 183-200, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18759927

ABSTRACT

B cells are induced to enter the cell cycle by stimuli including ligation of the B-cell receptor (BCR) complex and Toll-like receptor (TLR) agonists. This review discusses the contribution of several molecules, which act at distinct steps in B-cell activation. The adapter molecule Bam32 (B-lymphocyte adapter of 32 kDa) helps promote BCR-induced cell cycle entry, while the secondary messenger superoxide has the opposite effect. Bam32 and superoxide may fine tune BCR-induced activation by competing for the same limited resources, namely Rac1 and the plasma membrane phospholipid PI(3,4)P(2). The co-receptor CD22 can inhibit BCR-induced proliferation by binding to novel CD22 ligands. Finally, regulators of B-cell survival and death also play roles in B-cell transit through the cell cycle. Caspase 6 negatively regulates CD40- and TLR-dependent G(1) entry, while acting later in the cell cycle to promote S-phase entry. Caspase 6 deficiency predisposes B cells to differentiate rather than proliferate after stimulation. Bim, a pro-apoptotic Bcl-2 family member, exerts a positive regulatory effect on cell cycle entry, which is opposed by Bcl-2. New insights into what regulates B-cell transit through the cell cycle may lead to thoughtful design of highly selective drugs that target pathogenic B cells.


Subject(s)
Apoptosis Regulatory Proteins/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Cycle/immunology , Receptors, Antigen, B-Cell/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , B-Lymphocytes/metabolism , Cell Proliferation , Cell Survival/immunology , Humans , Lymphocyte Activation/immunology , NADPH Oxidases/immunology , NADPH Oxidases/metabolism , Receptors, Antigen, B-Cell/immunology , Sialic Acid Binding Ig-like Lectin 2/metabolism , Signal Transduction/immunology
6.
J Exp Med ; 202(10): 1363-74, 2005 Nov 21.
Article in English | MEDLINE | ID: mdl-16301744

ABSTRACT

The B cell activating factor belonging to the tumor necrosis factor family (BAFF) is required for B cell survival and maturation. The mechanisms by which BAFF mediates B cell survival are less understood. We found that BAFF and a proliferation-inducing ligand (APRIL), which are related, block B cell antigen receptor (BCR)-induced apoptosis upstream of mitochondrial damage, which is consistent with a role for Bcl-2 family proteins. BCR ligation strongly increased expression of the proapoptotic Bcl-2 homology 3-only Bcl-2 protein Bim in both WEHI-231 and splenic B cells, and increases in Bim were reversed by BAFF or APRIL. Small interfering RNA vector-mediated suppression of Bim blocked BCR-induced apoptosis. BAFF also induced Bim phosphorylation and inhibited BCR-induced association of Bim with Bcl-2. BAFF induced delayed but sustained stimulation of extracellular signal-regulated kinase (ERK) and its activators, mitogen-activated protein kinase/ERK activating kinase (MEK) and c-Raf, and MEK inhibitors promoted accumulation and dephosphorylation of Bim. These results suggest that BAFF inhibits BCR-induced death by down-regulating Bim via sustained ERK activation, demonstrating that BAFF directly regulates Bim function. Although transitional immature type 1 (T1) B cell numbers are normal in Bim(-/-) mice, T2 and follicular mature B cells are elevated and marginal zone B cells are reduced. Our results suggest that mature B cell homeostasis is maintained by BAFF-mediated regulation of Bim.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Down-Regulation/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins/genetics , B-Cell Activating Factor , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Survival/physiology , Humans , MAP Kinase Kinase Kinases/physiology , Membrane Proteins/genetics , Mice , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins/genetics , Receptors, Antigen, B-Cell/physiology , Spleen/cytology , Tumor Necrosis Factor Ligand Superfamily Member 13
7.
Nat Commun ; 9(1): 3877, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250067

ABSTRACT

PAXX is a recently identified component of the nonhomologous end joining (NHEJ) DNA repair pathway. The molecular mechanisms of PAXX action remain largely unclear. Here we characterise the interactomes of PAXX and its paralogs, XLF and XRCC4, to show that these factors share the ability to interact with DNA polymerase λ (Pol λ), stimulate its activity and are required for recruitment of Pol λ to laser-induced DNA damage sites. Stimulation of Pol λ activity by XRCC4 paralogs requires a direct interaction between the SP/8 kDa domain of Pol λ and their N-terminal head domains to facilitate recognition of the 5' end of substrate gaps. Furthermore, PAXX and XLF collaborate with Pol λ to promote joining of incompatible DNA ends and are redundant in supporting Pol λ function in vivo. Our findings identify Pol λ as a novel downstream effector of PAXX function and show XRCC4 paralogs act in synergy to regulate polymerase activity in NHEJ.


Subject(s)
DNA End-Joining Repair/physiology , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , DNA Breaks, Double-Stranded/radiation effects , DNA Repair Enzymes/genetics , DNA Repair Enzymes/isolation & purification , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/isolation & purification , HEK293 Cells , Humans , Lasers/adverse effects , Mutagenesis, Site-Directed , Protein Binding/physiology , Protein Domains/physiology , Protein Interaction Mapping/methods , Protein Interaction Maps/physiology , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tandem Mass Spectrometry/methods
8.
Haematologica ; 91(6): 772-80, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16769579

ABSTRACT

BACKGROUND AND OBJECTIVES: We explored the expression of LCK and BAFF-R (B-cell activating factor receptor) both of which are known to play a role in signaling and apoptosis, in routine tissue biopsies. It was hypothesized that their expression patterns might yield information on apoptosis as it occurs in normal and reactive lymphoid cells, and also be of value for the detection of lymphoma subtypes. DESIGN AND METHODS: Both molecules were studied in paraffin-embedded tissue sections and cell lines by immunoperoxidase staining, and were also studied by western blotting. Human tonsillar B-cell subsets were analyzed by flow cytometry for LCK expression. RESULTS: LCK was detected for the first time in germinal centers and, at lower levels, in mantle zone B cells. The presence of LCK in B cells was confirmed by western blotting. Cross-linking surface IgM reduced LCK expression whereas cross-linking surface CD40 appeared to have the opposite effect. BAFF-R was present on mantle zone B cells but absent or weakly expressed in germinal center cells. Most lymphomas of germinal center origin (e.g. follicular lymphoma) and also many mantle cell lymphomas, chronic lymphocytic leukemia (CLL) and most T-cell neoplasms expressed LCK. In contrast, BAFF-R was expressed in a variety of B-cell lymphomas, but often absent in grade 3 follicular lymphomas and diffuse large B-cell lymphomas (DLBCL). Both LCK-positive and BAFF-R-positive DLBCL tended to be of germinal-center phenotype. INTERPRETATION AND CONCLUSIONS: The reciprocal expression pattern of LCK and BAFF-R in germinal center and mantle zone B cells may reflect their opposing roles in apoptosis. Their detection in lymphoma tissue biopsies may therefore be of clinical relevance in predicting response to treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphoma/genetics , Membrane Proteins/genetics , Receptors, Tumor Necrosis Factor/genetics , Apoptosis , B-Cell Activation Factor Receptor , Cell Line, Tumor , Humans , Immunohistochemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphoid Tissue/pathology , Lymphoma/pathology , Lymphoma, B-Cell/genetics , Lymphoma, Mantle-Cell/genetics , Lymphoma, T-Cell/genetics , Membrane Proteins/metabolism , Palatine Tonsil/immunology , Palatine Tonsil/pathology , Prognosis , Receptors, Tumor Necrosis Factor/metabolism
9.
Eur J Immunol ; 37(10): 2715-22, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17705137

ABSTRACT

BH3-only Bcl-2 homologs are key regulators of the intrinsic apoptotic pathway. In particular, Bim, is critical for mediating apoptosis of hematopoietic cells including B cells. While studies using Bcl-2 Tg mice have defined an important role for Bcl-2 in cell cycle control, the role of BH3-only proteins is less clear. Using Bim KO mice, we show that Bim is required for B cells to enter the cell cycle normally. Bim KO B cells had reduced cell division compared to WT B cells in response to BCR, TLR3 or TLR4 signaling, whereas Bim deficiency did not affect TLR9-induced B cell division. Cell cycle progression in BCR- and LPS-stimulated Bim KO B cells was blocked at the G0-G1 stage. BCR-induced p130 degradation and pRb hyperphosphorylation on Ser807/811, which are critical for G1 entry, were reduced in Bim KO compared to WT B cells. Likewise, BCR-induced p27(Kip1) degradation was decreased in Bim KO compared to WT B cells. These defects in BCR-induced cell cycle entry correlated with a proximal defect in BCR-mediated intracellular calcium release in Bim KO B cells. Our results suggest that the balance of pro- and anti-apoptotic Bcl-2 family proteins is critical for controlling both cell cycle progression and apoptosis in B cells.


Subject(s)
Apoptosis Regulatory Proteins/physiology , B-Lymphocytes/cytology , Cell Cycle/immunology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptors, Antigen, B-Cell/physiology , Animals , Apoptosis/immunology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , B-Lymphocytes/immunology , Bcl-2-Like Protein 11 , Cell Proliferation , Cell Survival/immunology , Cells, Cultured , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/physiology
10.
J Biol Chem ; 282(17): 12831-41, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17327223

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is selectively toxic to tumor compared with normal cells. Other members of the TNF family of death ligands (TNF, CD95L) engage their respective receptors (TNF-R1 and CD95), resulting in internalization of receptor and ligand and recruitment of adaptor proteins to the caspase activation platform known as the death-inducing signaling complex (DISC). Recently, TNF-R1 and CD95 have been shown to induce apoptosis with an absolute requirement for internalization of their corresponding receptors in the formation of a DISC. We show that TRAIL and its receptors are rapidly endocytosed in a time- and concentration-dependent manner. Blockade of receptor internalization with hyperosmotic sucrose did not inhibit TRAIL-induced apoptosis but, rather, amplified the apoptotic signaling of TRAIL. Plate-bound and soluble TRAIL induced similar levels of apoptosis. Together these results suggest that neither ligand nor receptor internalization is required for TRAIL-induced apoptosis. Internalization of TRAIL is mediated primarily by clathrin-dependent endocytosis and also by clathrin-independent pathways. Inhibition of clathrin-dependent internalization by overexpression of dominant negative forms of dynamin or AP180 did not inhibit TRAIL-induced apoptosis. Consistent with the finding that neither internalization of TRAIL nor its receptors is required for transmission of its apoptotic signal, recruitment of FADD (Fas-associated death domain) and procaspase-8 to form the TRAIL-associated DISC occurred at 4 degrees C, independent of endocytosis. Our findings demonstrate that TRAIL and TRAIL receptor 1/2, unlike TNF-TNF-R1 or CD95L-CD95, do not require internalization for formation of the DISC, activation of caspase-8, or transmission of an apoptotic signal in BJAB type I cells.


Subject(s)
Apoptosis/physiology , Endocytosis/physiology , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis/genetics , Caspase 8/metabolism , Cold Temperature , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Dose-Response Relationship, Drug , Dynamins/biosynthesis , Dynamins/genetics , Endocytosis/drug effects , Fas Ligand Protein/metabolism , Fas Ligand Protein/pharmacology , Fas-Associated Death Domain Protein/metabolism , HeLa Cells , Humans , Mutation , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/metabolism
11.
Eur J Immunol ; 37(4): 990-1000, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17357108

ABSTRACT

Microorganisms with pathogen-associated molecular patterns (PAMP) activate B cells directly by binding to TLR and also indirectly by inducing APC to release cytokines such as BAFF that promote B cell survival. We found that murine B cells activated concomitantly with LPS (TLR-4 ligand) and BAFF are protected from spontaneous apoptosis, but are more susceptible to Fas/CD95-mediated cell death. This increased susceptibility to Fas-induced apoptosis is associated with a dramatic coordinated up-regulation of Fas/CD95 and IRF-4 expression through a mechanism mediated, at least in part, by inhibition of the MEK/ERK pathway. Up-regulation of Fas/CD95 by BAFF is restricted to B cells activated through TLR-4, but not through TLR-9, BCR or CD40. TLR ligands differ in the BAFF family receptors (R) they induce on B cells: BAFF-R is increased by the TLR4 ligand, LPS, but not by the TLR9 ligand, CpG-containing oligodeoxynucleotides, which, in contrast, strongly up-regulates transmembrane activator and CAML interactor (TACI). This suggests the up-regulation of Fas by BAFF is mediated by BAFF-R and not by TACI. Consistently, APRIL, which binds to TACI and B cell maturation antigen but not BAFF-R, did not enhance Fas expression on LPS-activated B cells. Increased susceptibility to Fas-mediated killing of B cells activated with LPS and BAFF may be a fail-safe mechanism to avoid overexpansion of nonspecific or autoreactive B cells.


Subject(s)
Apoptosis/immunology , B-Cell Activating Factor/physiology , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Lipopolysaccharides/pharmacology , fas Receptor/physiology , Animals , B-Lymphocyte Subsets/metabolism , Cell Death/immunology , Cells, Cultured , Growth Inhibitors/biosynthesis , Growth Inhibitors/physiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Rats , Up-Regulation/immunology , fas Receptor/biosynthesis
12.
Immunity ; 18(6): 724-5, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12818154

ABSTRACT

In this issue of Immunity, Brodeur et al. show that C4b binding protein (C4BP), a regulator component of the classical complement (C) pathway, can bind to CD40 receptors on B cells and activate them. This suggests a novel way by which CD40 may function to bridge innate and adaptive immune responses.


Subject(s)
CD40 Antigens/immunology , Glycoproteins , Immunity, Innate , Animals , B-Lymphocytes/immunology , Complement Inactivator Proteins/metabolism , Humans , Receptors, Cell Surface/metabolism
13.
Immunol Rev ; 197: 129-46, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14962192

ABSTRACT

During their development, B-lineage cells are selected to mature, to die, to divide, or to survive and wait, ready to respond to external signals. The homeostatic balance between growth, death, and survival is mediated by signaling pathways through the B-cell antigen receptor (BCR) complex, cytokine and chemokine receptors or cell-cell coreceptor interactions. The BCR complex is a master regulator essential at key checkpoints during development. These checkpoints involve various processes, including negative selection (deletion), anergy, receptor editing, and positive selection. Without BCRs or downstream BCR-signaling components, B-lineage cells arrest during development. Removal of BCRs from mature B cells leads to their death. Here, we discuss signaling pathways in B cells that activate members of the caspase family of cysteine proteases. In some B-cell subsets, BCR signaling activates caspases, which in turn induce a program leading to cell death. However, in other contexts, caspases are involved in the proliferation of B cells. The outcome depends in part on the presence or absence of modifiers that affect signaling thresholds and on which caspases are activated. These mechanisms allow the coordinated regulation of proliferation and apoptosis that is essential for lymphoid homeostasis.


Subject(s)
B-Lymphocytes/enzymology , Caspases/metabolism , Animals , Apoptosis , B-Cell Activating Factor , B-Lymphocytes/cytology , Humans , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
14.
Blood ; 101(11): 4464-71, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12531790

ABSTRACT

Macrophages and dendritic cells play an important role in regulating B-cell responses, including proliferation to antigens such as trinitrophenyl (TNP)-Ficoll and TNP-Brucella abortus. However, the mechanisms and molecule(s) that regulate these processes are relatively undefined. In this report, we show that human macrophages generated in vitro strongly costimulate proliferation of dense human tonsillar B cells ligated via their B-cell antigen receptor (BCR) but not proliferation via CD40. Similarly, dendritic cells also markedly enhance BCR-activated B-cell proliferation. Soluble molecule(s) are required for human macrophages to costimulate proliferation of B cells triggered via their BCR. Importantly, a TACI (trans-membrane activator and CAML interactor)-Fc fusion protein inhibits both macrophage- and dendritic cell (DC)-dependent BCR-activated B-cell proliferation, indicating a requirement for at least one of the known TACI ligands, BAFF and/or APRIL. Consistent with a major role for BAFF, macrophages release BAFF at levels sufficient to potently costimulate BCR-induced B-cell proliferation. In addition, BAFF is more than 100-fold more potent than APRIL in enhancing BCR-mediated human B-cell proliferation. Furthermore, immunodepletion of APRIL under conditions that prevent APRIL-mediated B-cell costimulation does not block macrophage enhancement of B-cell proliferation. Finally, there is no correlation between the high levels of a proliferation-inducing ligand (APRIL) expressed by macrophages compared with DCs and the similar abilities of macrophages and DCs to enhance BCR-stimulated B-cell proliferation. In summary, our results suggest that macrophage- and DC-derived B-cell-activating factor belonging to the TNF family (BAFF) represents a key molecule by which macrophages and DCs directly regulate human B-cell proliferative responses to T-cell-independent stimuli.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Lymphocyte Activation , Macrophages/immunology , Membrane Proteins/physiology , Tumor Necrosis Factor-alpha/physiology , Adolescent , Adult , B-Cell Activating Factor , Cell Communication/immunology , Dendritic Cells/cytology , Female , Humans , Lymphocyte Activation/immunology , Macrophages/cytology , Male , Membrane Proteins/immunology , Middle Aged , Neuropeptides/immunology , Nuclear Proteins/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Tumor Necrosis Factor/immunology , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL