Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2248: 43-61, 2021.
Article in English | MEDLINE | ID: mdl-33185866

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) are major constituents of the extracellular matrix and well-established obstacles to regeneration in the central nervous system. As such, they are promising targets for therapy in neurological pathologies where repair is needed, such as spinal cord injuries, and multiple sclerosis. Since CSPGs mediate their inhibitory functions by interacting with signaling protein partners through their variably sulfated chondroitin sulfate glycosaminoglycan (CS-GAG) chains, blocking these epitopes presents a path to promoting repair. A member of the tumor necrosis factor (TNF) superfamily, a proliferation-inducing ligand (APRIL) has been shown to bind to CSPGs. Here we describe in vitro methods to evaluate APRIL's ability to block CSPGs from interacting with their partner proteins and promote neuronal growth.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Blotting, Western , Brain , Cell Line , Chondroitin Sulfate Proteoglycans/antagonists & inhibitors , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Gene Expression , Gene Library , Humans , Mice , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
2.
Ann Neurol ; 85(3): 406-420, 2019 03.
Article in English | MEDLINE | ID: mdl-30635946

ABSTRACT

OBJECTIVE: The two related tumor necrosis factor members a proliferation-inducing ligand (APRIL) and B-cell activation factor (BAFF) are currently targeted in autoimmune diseases as B-cell regulators. In multiple sclerosis (MS), combined APRIL/BAFF blockade led to unexpected exacerbated inflammation in the central nervous system (CNS) of patients. Here, we investigate the role of the APRIL/BAFF axis in the CNS. METHODS: APRIL expression was analyzed in MS lesions by immunohistochemistry. The in vivo role of APRIL was assessed in the murine MS model, experimental autoimmune encephalitis (EAE). Functional in vitro studies were performed with human and mouse astrocytes. RESULTS: APRIL was expressed in lesions from EAE. In its absence, the disease was worst. Lesions from MS patients also showed APRIL expression upon infiltration of macrophages. Notably, all the APRIL secreted by these macrophages specifically targeted astrocytes. The upregulation of chondroitin sulfate proteoglycan, sometimes bearing chondroitin sulfate of type E sugar moieties, binding APRIL, in reactive astrocytes explained the latter selectivity. Astrocytes responded to APRIL by producing a sufficient amount of IL-10 to dampen antigen-specific T-cell proliferation and pathogenic cytokine secretion. Finally, an intraspinal delivery of recombinant APRIL before disease onset, shortly reduced EAE symptoms. Repeated intravenous injections of recombinant APRIL before and even at disease onset also had an effect. INTERPRETATION: Our data show that APRIL mediates an anti-inflammatory response from astrocytes in MS lesions. This protective activity is not shared with BAFF. ANN NEUROL 2019;85:406-420.


Subject(s)
Astrocytes/metabolism , B-Cell Activating Factor/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Adult , Aged , Animals , Astrocytes/immunology , Astrocytes/pathology , Cell Proliferation , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/metabolism , Cytokines/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunohistochemistry , Interleukin-10/immunology , Macrophages/pathology , Male , Mice , Mice, Knockout , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
3.
Exp Neurol ; 298(Pt A): 97-103, 2017 12.
Article in English | MEDLINE | ID: mdl-28911883

ABSTRACT

We have studied the role of the tumor necrosis factor superfamily member APRIL in the development of embryonic mouse midbrain dopaminergic neurons in vitro and in vivo. In culture, soluble APRIL enhanced axon growth during a window of development between E12 and E14 when nigrostriatal axons are growing to their targets in the striatum in vivo. April transcripts were detected in both the striatum and midbrain during this period and at later stages. The axon growth-enhancing effect of APRIL was similar to that of glial cell-derived neurotrophic factor (GDNF), but in contrast to GDNF, APRIL did not promote the survival of midbrain dopaminergic neurons. The effect of APRIL on axon growth was prevented by function-blocking antibodies to one of its receptors, BCMA (TNFRSF13A), but not by function-blocking antibodies to the other APRIL receptor, TACI (TNFRSF13B), suggesting that the effects of APRIL on axon growth are mediated by BCMA. In vivo, there was a significant reduction in the density of midbrain dopaminergic projections to the striatum in April-/- embryos compared with wild type littermates at E14. These findings demonstrate that APRIL is a physiologically relevant factor for the nigrostriatal projection. Given the importance of the degeneration of dopaminergic nigrostriatal connections in the pathogenesis and progression of Parkinson's disease, our findings contribute to our understanding of the factors that establish nigrostriatal integrity.


Subject(s)
Axons/physiology , Corpus Striatum/physiology , Dopaminergic Neurons/physiology , Mesencephalon/physiology , Substantia Nigra/physiology , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Animals , Axons/drug effects , Cells, Cultured , Corpus Striatum/drug effects , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Mesencephalon/drug effects , Mice , Mice, Knockout , Substantia Nigra/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency
4.
Eur J Immunol ; 47(7): 1211-1219, 2017 07.
Article in English | MEDLINE | ID: mdl-28463395

ABSTRACT

Disturbances of plasma cell homeostasis and auto-antibody production are hallmarks of systemic lupus erythematosus. The aim of this study was to explore the presence of circulating anti-ENA and anti-dsDNA antibody-secreting cells, to determine their dependence on plasma cell-niche cytokines and to analyze their clinical value. The study was performed in SLE patients with serum anti-ENA and/or anti-dsDNA antibodies (n = 57). Enriched B-cell fractions and sorted antibody-secreting cells (CD19low CD38high ) were obtained from blood. dsDNA- and ENA-specific antibody-secreting cells were identified as cells capable of active auto-antibody production in culture. The addition of a combination of IL-6, IL-21, BAFF, APRIL, and CXCL12 to the cultures significantly augmented auto-antibody production and antibody-secreting cell proliferation, whereas it diminished apoptosis. The effect on auto-antibody production was dependent on STAT-3 activation as it was abrogated in the presence of the JAK/STAT-3 pathway inhibitors ruxolitinib and stattic. Among patients with serum anti-dsDNA antibodies, the detection of circulating anti-dsDNA-antibody-secreting cells was associated with higher disease activity markers. In conclusion, auto-antibody production in response to plasma cell-niche cytokines that are usually at high levels in SLE patients is dependent on JAK/STAT-3 activation. Thus, patients with circulating anti-dsDNA antibody-secreting cells and active disease could potentially benefit from therapies targeting the JAK/STAT3 pathway.


Subject(s)
Antibodies, Antinuclear/blood , Antibody-Producing Cells/immunology , DNA/immunology , Lupus Erythematosus, Systemic/immunology , STAT3 Transcription Factor/metabolism , Adolescent , Adult , Aged , Antibodies, Antinuclear/immunology , Antibody-Producing Cells/drug effects , Apoptosis/drug effects , B-Cell Activating Factor/pharmacology , Cell Proliferation , Chemokine CXCL2/pharmacology , Cyclic S-Oxides/pharmacology , DNA/blood , Female , Humans , Interleukin-6/pharmacology , Interleukins/pharmacology , Lupus Erythematosus, Systemic/blood , Male , Middle Aged , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Young Adult
5.
PLoS One ; 12(1): e0171171, 2017.
Article in English | MEDLINE | ID: mdl-28125737

ABSTRACT

Immunological prevention of infectious disease, especially viral, is based on antigen-specific long-lived memory B cells. To test for cellular proliferation and differentiation factors in swine, an outbred model for humans, CD21+ B cells were activated in vitro with CD40L and stimulated with purported stimulatory cytokines to characterize functional responses. IL-21 induced a 3-fold expansion in total cell numbers with roughly 15% of all B cells differentiating to IgM or IgG antibody secreting cells (ASCs.) However, even with robust proliferation, cellular viability rapidly deteriorated. Therefore, a proliferation inducing ligand (APRIL) and B cell activating factor (BAFF) were evaluated as survival and maintenance factors. BAFF was effective at enhancing the viability of mature B cells as well as ASCs, while APRIL was only effective for ASCs. Both cytokines increased approximately two-fold the amount of IgM and IgG which was secreted by IL-21 differentiated ASCs. Mature B cells from porcine reproductive and respiratory virus (PRRSV) immune and naïve age-matched pigs were activated and treated with IL-21 and then tested for memory cell differentiation using a PRRSV non-structural protein 7 ELISPOT and ELISA. PRRSV immune pigs were positive on both ELISPOT and ELISA while naïve animals were negative on both assays. These results highlight the IL-21-driven expansion and differentiation of memory B cells in vitro without stimulation of the surface immunoglobulin receptor complex, as well as the establishment of a defined memory B cell culture system for characterization of vaccine responses in outbred animals.


Subject(s)
Antibody-Producing Cells/cytology , B-Lymphocytes/cytology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Interleukins/pharmacology , Animals , Antibody-Producing Cells/immunology , B-Cell Activating Factor/pharmacology , B-Lymphocytes/immunology , Cell Survival/drug effects , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Swine , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
6.
FEBS J ; 284(1): 97-113, 2017 01.
Article in English | MEDLINE | ID: mdl-27981746

ABSTRACT

HIV-1-infected macrophages are a key contributor to the formation of a viral reservoir and new treatment strategies focus on eliminating this pool of virus. Galectin-3 is a potent apoptosis-inducing protein that regulates diverse cellular activities. In the present study, we investigated whether galectin-3 could induce cell death in HIV-1-infected macrophages using HIV-1-infected THP1 monocytes (THP1-MNs) and THP1-derived macrophages (THP1-MΦs) as in vitro cellular models. We found that THP1-MΦs were more resistant than the THP1-MNs to HIV-1 infection-induced death, and that HIV-1 infection of the THP1-MΦs increased expression of the anti-apoptotic proteins Mcl-1, Bcl-2 and Bcl-xL. Additionally, galectin-3 but not FasL, tumor necrosis factor (TNF)-related apoptosis-inducing ligand or TNF-α, could induce cell death in HIV-1-infected THP1-MΦs. A similar result was shown for primary human monocyte-derived macrophages. Galectin-3-induced cell death was also significantly increased in macrophages obtained from SIVmac251-infected macaques compared to that of macrophages from healthy macaques. Furthermore, galectin-3-induced cell death in HIV-1-infected THP1-MΦs was caspase independent. Interestingly, endonuclease G (Endo G) was increased in the nucleus and decreased in the cytoplasm of galectin-3-treated cells; thus, galectin-3-induced cell death in HIV-1-infected THP1-MΦs is most likely related to the translocation of Endo G from the cytoplasm to the nucleus. These findings suggest that galectin-3 may potentially aid in the eradication of HIV-1/SIV-infected macrophages.


Subject(s)
Endodeoxyribonucleases/genetics , Galectin 3/pharmacology , HIV-1/drug effects , Macrophages/drug effects , Monocytes/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Cell Death/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Endodeoxyribonucleases/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression Regulation , HIV-1/growth & development , Humans , Macaca , Macrophages/pathology , Macrophages/virology , Monocytes/pathology , Monocytes/virology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/growth & development , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , bcl-X Protein/genetics , bcl-X Protein/metabolism
7.
Anim Biotechnol ; 27(1): 1-8, 2016.
Article in English | MEDLINE | ID: mdl-26485397

ABSTRACT

A proliferation-inducing ligand (APRIL) is a critical member of the tumor necrosis factor (TNF) superfamily, which is involved in immune regulation. In the present study, the cDNA of cat APRIL (cAPRIL) was successfully amplified. Sequence analysis showed that the open reading frame (ORF) of cAPRIL contains a putative furin protease cleavage site (R-R-K-R), a conserved putative N-glycosylation site (Asn(124)), and two conservative cysteine residues (Cys(196) and Cys(211)). Real-time quantitative PCR (qPCR) analysis revealed that cAPRIL could be detected in various tissues. The phylogenetic analysis and predicted three dimensional (3D) structure revealed that it is similar to its counterparts. The extracellular soluble domain of the cAPRIL (csAPRIL) fragment was cloned into the expression vector pET43.1a. SDS-PAGE and Western blotting analysis indicated a high-level expression of csAPRIL protein in Escherichia coli BL21 (DE3). MTT assays revealed that purified recombinant csAPRIL protein was able to stimulate proliferation of mouse B-cells. These findings indicate that cAPRIL plays an important role in proliferation of B-cells and provide the basis for investigation on the roles of APRIL in this important domestic species.


Subject(s)
Cats/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Amino Acid Sequence , Animals , B-Lymphocytes/drug effects , Base Sequence , Cell Proliferation/drug effects , Cloning, Molecular , Escherichia coli/genetics , Male , Mice , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 13/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
8.
Cell Immunol ; 295(2): 127-36, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25880104

ABSTRACT

Long-lived plasma cells (PCs) contribute to humoral immunity through an undefined mechanism. Memory B cells, but not human naïve B cells, can be induced to differentiate into long-lived PCs in vitro. Because evidence links a proliferation-inducing ligand (APRIL), a tumor necrosis factor family member, to the ability of bone marrow to mediate long-term PC survival, we reasoned that APRIL influences the proliferation and differentiation of naïve B cells. We describe here the development of a simple cell culture system that allowed us to show that APRIL sustained the proliferation of naïve human B cells and induced them to differentiate into long-lived PCs. Blocking the transmembrane activator and calcium modulator and cyclophilin ligand interactor or B-cell mature antigen shows they were required for the differentiation of naïve B cells into long-lived PCs in vitro. Our in vitro culture system will reveal new insights into the biology of long-lived PCs.


Subject(s)
B-Cell Maturation Antigen/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Transmembrane Activator and CAML Interactor Protein/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Adult , Antibodies, Monoclonal/pharmacology , B-Cell Maturation Antigen/antagonists & inhibitors , Cell Culture Techniques , Cell Differentiation/drug effects , Female , Flow Cytometry , Humans , Male , Middle Aged , Transmembrane Activator and CAML Interactor Protein/antagonists & inhibitors , Young Adult
9.
Mol Immunol ; 64(1): 90-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25466610

ABSTRACT

Fibroblast-like synoviocyte (FLS) is the ultimate effectual cells in the pathogenesis of rheumatoid arthritis (RA). The current study was undertaken to investigate whether a proliferation-inducing ligand (APRIL) mediates the function of FLS in an animal model of RA. Rat adjuvant-induced arthritis (AA) was induced by intradermal injection of 0.1 ml complete Freund's adjuvant. Synovium APRIL expression was detected by immunohistochemical analysis. The level of APRIL and matrix metalloproteinase (MMP)-9 were assayed by enzyme-linked immunosorbent assay. The expression of APRIL and its receptors (TACI, BCMA and BAFF-R) were assessed by immunofluorescence staining, flow cytometry, reverse transcription-polymerase chain reaction (RT-PCR) and real-time RT-PCR. The effects of APRIL on the function of FLS were investigated by MTT, Quantibody Rat Inflammation Array 1 and transwell assays, respectively. A higher concentration of APRIL was detected in AA synovium homogenate compared with normal group. AA FLS expressed APRIL, TACI, BAFF-R and BCMA at the mRNA levels, whereas only APRIL and BCMA were confirmed to be expressed on membrane by flow cytometry. APRIL stimulated AA FLS proliferation, migration and invasion and the secretion of proinflammatory factors. In addition, FLS cocultured with APRIL-treated B cells or T cells had a significantly greater proliferation than FLS cultured alone. Neutralization of APRIL by the TACI-Ig fusion protein attenuated these stimulating effects of APRIL on FLS. Our data indicate that APRIL may act as an important mediator for facilitating the function of FLS. Blockade of APRIL thus may be a valuable adjunct in the treatment of RA.


Subject(s)
Arthritis, Experimental/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Synovial Membrane/pathology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/genetics , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/metabolism , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Fibroblasts/enzymology , Inflammation Mediators/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Rats, Sprague-Dawley , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
10.
Leukemia ; 28(8): 1647-56, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24504026

ABSTRACT

The recent understanding of plasma cell (PC) biology has been obtained mainly from murine models. The current concept is that plasmablasts home to the BM and further differentiate into long-lived PCs (LLPCs). These LLPCs survive for months in contact with a complex niche comprising stromal cells (SCs) and hematopoietic cells, both producing recruitment and survival factors. Using a multi-step culture system, we show here the possibility to differentiate human memory B cells into LLPCs surviving for at least 4 months in vitro and producing immunoglobulins continuously. A remarkable feature is that IL-6 is mandatory to generate LLPCs in vitro together with either APRIL or soluble factors produced by SCs, unrelated to APRIL/BAFF, SDF-1, or IGF-1. These LLPCs are out of the cell cycle, express highly PC transcription factors and surface markers. This model shows a remarkable robustness of human LLPCs, which can survive and produce highly immunoglobulins for months in vitro without the contact with niche cells, providing the presence of a minimal cocktail of growth factors and nutrients. This model should be useful to understand further normal PC biology and its deregulation in premalignant or malignant PC disorders.


Subject(s)
Chemokine CXCL12/pharmacology , Interleukin-6/pharmacology , Plasma Cells/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , B-Cell Activation Factor Receptor/pharmacology , Cell Survival , Cells, Cultured , Humans , NF-kappa B/physiology , Plasma Cells/physiology , Transcriptome
11.
Protein Pept Lett ; 19(3): 315-25, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21933134

ABSTRACT

TACI is a member of the tumor necrosis factor receptor superfamily and serves as a key regulator of B cell function. The extracellular domain of a typical TNFR contains multiple copies of CRD, which bind in the monomermonomer interfaces of a trimeric ligand. TACI binds to two ligands, APRIL and BAFF, with high affinity and contains two CRD in its extracellular regions, while BCMA and BR3, contain a single or partial CRD for binding the two ligands. However, TACI can be classified as a single CRD receptor because the amino-terminal CRD1 doesn't contribute to ligand binding. To obtain a new variant of TACI possessing higher affinities for binding, we fused a repeat sequence of CRD2 to the N-terminus of the short form of TACI. The new APRIL antagonist peptide, CRD2-shortTACI-Fc, was designed based on the modeling 3-D complex structure of TACI and APRIL. As expected, the purified recombinant CRD2-shortTACI-Fc fusion protein could bind to APRIL in vitro and demonstrated dose-dependent inhibition of APRIL-induced proliferative activity in Raji cells. We found that CRD2-shortTACI-Fc, has a higher affinity for binding to ligands than short-TACI-Fc, which contains a single CRD2.


Subject(s)
Genetic Engineering/methods , Pichia/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism , Amino Acid Sequence , Artificial Gene Fusion , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression , Humans , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Signal Transduction , Transmembrane Activator and CAML Interactor Protein/chemistry , Transmembrane Activator and CAML Interactor Protein/isolation & purification , Tumor Necrosis Factor Ligand Superfamily Member 13/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
12.
Blood ; 117(3): 890-901, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20709908

ABSTRACT

A proliferation-inducing ligand (APRIL) promotes survival and drug resistance in multiple myeloma (MM) cell lines. We studied the effect of APRIL on cell-cycle behavior in primary MM cells and correlated our findings with D-type cyclin expression by immunohistochemistry and/or Western blotting. In MM cases, expressing cyclin D2 APRIL significantly increased the percentage of CD138(+) cells in S + G(2)/M phase (from 8.4% ± 1.9% to 14.3% ± 2.6%, n = 15, P < .01), whereas a lesser effect was seen in cases expressing cyclin D1 (n = 18). Cell-cycle response to APRIL was most marked for cyclin D2-expressing cases with IgH translocations (P < .01) and was accompanied by increased expression of cyclin D2, CDK4, CDK6, and phospho-retinoblastoma protein. Cell-cycle proteins in cyclin D1(+) cells were not modulated by APRIL. Surface expression of B-cell maturation antigen and transmembrane activator and calcium-modulating cyclophilin ligand interactor was not significantly different between cyclin D1(+) and D2(+) MM cells. We observed activation of nuclear factor-κB and PI3-kinase pathways in response to APRIL in both cyclin D1(+) and D2(+) MM cells. In conclusion, APRIL stimulates G(1)/S progression in cyclin D2(+) MM cells bearing IgH translocations but has minimal effect on cyclin D1(+) cells, suggesting MM cells from different cyclin D/translocation classes rely on different mechanisms for cell-cycle re-entry.


Subject(s)
Cell Cycle/drug effects , Cyclin D/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Blotting, Western , Bone Marrow Cells/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Cyclin D2/metabolism , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Protein Transport/drug effects , Syndecan-1/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
13.
Acta Biochim Pol ; 56(4): 703-10, 2009.
Article in English | MEDLINE | ID: mdl-19997650

ABSTRACT

Soluble APRIL (sAPRIL), the active form of a proliferation-inducing ligand (APRIL), is implicated in the proliferation of tumor cells. Suppressing APRIL function has been considered as a potential strategy for the therapy of APRIL-associated tumors. In the present study, we generated human sAPRIL and its two mutants, Gln187-D-sAPRIL (Gln187 deleted) and Gly187-sAPRIL (Gln187 replaced by Gly). In vitro experiments showed that the two mutants had similar specific binding capacity to lung carcinoma A549 cells compared to the wild-type sAPRIL, and both, especially Gly187-sAPRIL, exhibited significant antagonistic effect on sAPRIL-induced tumor cell proliferation in a dose-dependent manner, which might be predominantly mediated by blocking sAPRIL-induced MEK and ERK phosphorylation but not p38MAPK or JNK signaling. In vivo experiments with nude mice bearing A549 cell-derived xenograft tumor showed that only the Gly187-sAPRIL mutant could significantly suppress the tumor growth. These results suggest that Gln187 may be a crucial amino acid in APRIL-mediated tumor cell proliferation via the MEK-ERK signaling pathway and that the sAPRIL mutants may serve as novel potential antagonists of APRIL for the therapy of APRIL-associated cancers.


Subject(s)
Cell Proliferation/drug effects , Lung Neoplasms/pathology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Glutamine/genetics , Humans , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/physiology , Male , Mice , Sequence Alignment , Signal Transduction/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Xenograft Model Antitumor Assays
14.
J Allergy Clin Immunol ; 123(6): 1277-86.e5, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19427685

ABSTRACT

BACKGROUND: Transmembrane activator, calcium modulator, and cyclophilin ligand interactor (TACI) expression on B cells is upregulated by Toll-like receptor (TLR) 4. OBJECTIVE: We sought to examine whether TACI synergizes with TLR4 in driving immunoglobulin production by B cells and to examine the mechanism of this synergy. METHODS: Purified mouse naive B cells were stimulated with the TACI ligand a proliferation-inducing ligand (APRIL) and with suboptimal concentrations of the TLR4 ligand LPS in the presence or absence of IL-4. Immunoglobulin secretion was measured by means of ELISA. Surface IgG1-positive B cells and CD138+ plasmacytoid cells were enumerated by means of FACS. Expression of gamma1 and epsilon germline transcripts, activation-induced cytidine deaminase, and gamma1 and epsilon mature transcripts was measured by means of RT-PCR. RESULTS: APRIL synergized with LPS in driving B-cell proliferation and IgM, IgG1, IgG3, IgE, and IgA production. This was mediated by TACI because it was preserved in B-cell maturation antigen-/-, but not TACI-/-, B cells. APRIL and LPS synergized to promote isotype switching, as evidenced by increased expression of activation-induced cytidine deaminase and gamma1 and epsilon mature transcripts and generation of surface IgG1-positive cells. More importantly, APRIL and LPS strongly synergized to drive the plasma cell differentiation program, as evidenced by an increase in CD138+ cells and expression of B lymphocyte induced maturation protein-1 (Blimp-1), interferon regulatory factor-4 (IRF-4), and the spliced form of X-box binding protein-1 (XBP-1). TACI-/- mice had impaired IgM and IgG1 antibody responses to immunization, with a suboptimal dose of the type I T cell-independent antigen 2, 4, 6- Trinitrophenol (TNP)-LPS. CONCLUSIONS: These observations suggest that TACI cooperates with TLR4 to drive B-cell differentiation and immunoglobulin production in vitro and in vivo.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Plasma Cells/immunology , Toll-Like Receptor 4/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology , Animals , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/immunology , B-Cell Maturation Antigen/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Immunoglobulins/immunology , Immunoglobulins/metabolism , Interleukin-4/pharmacology , Ligands , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Plasma Cells/drug effects , Plasma Cells/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics
15.
Blood ; 113(19): 4614-26, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19228610

ABSTRACT

A plethora of myeloma growth factors (MGFs) has been identified, but their relative importance and cooperation have not been determined. We investigated 5 MGFs (interleukin-6 [IL-6], insulin-like growth factor type 1 [IGF-1], hepatocyte growth factor [HGF], HB-epidermal growth factor [HB-EGF], and a proliferation-inducing ligand [APRIL]) in serum-free cultures of human myeloma cell lines (HMCLs). In CD45(-) HMCLs, an autocrine IGF-1 loop promoted autonomous survival whereas CD45(+) HMCLs could not survive without addition of MGFs, mainly IGF-1 and IL-6. IGF-1 was the major one: its activity was abrogated by an IGF-1R inhibitor only, whereas IL-6, HGF, or HB-EGF activity was inhibited by both IGF-1R- and receptor-specific inhibition. APRIL activity was inhibited by its specific inhibitor only. Of the investigated MGFs and their receptors, only expressions of IGF-1R and IL-6R in multiple myeloma cells (MMCs) of patients delineate a group with adverse prognosis. This is mainly explained by a strong association of IGF-1R and IL-6R expression and t(4;14) translocation, but IGF-1R expression without t(4;14) can also have a poor prognosis. Thus, IGF-1-targeted therapy, eventually in combination with anti-IL-6 therapy, could be promising in a subset of patients with MMCs expressing IGF-1R.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Multiple Myeloma/pathology , Receptor, IGF Type 1/metabolism , Blotting, Western , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , Culture Media, Serum-Free , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Heparin-binding EGF-like Growth Factor , Hepatocyte Growth Factor/pharmacology , Humans , Immunoblotting , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Interleukin-6/pharmacology , Leukocyte Common Antigens/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
16.
J Allergy Clin Immunol ; 120(4): 885-91, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17689597

ABSTRACT

BACKGROUND: Transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) is a receptor used by B cell-activating factor of the TNF family and a proliferation-inducing ligand (APRIL) to induce isotype switching independently of CD40 and is mutated in patients with common variable immunodeficiency. OBJECTIVE: We sought to determine whether TACI and CD40 cooperate in inducing class switch recombination and immunoglobulin production. METHODS: Naive mouse B cells were stimulated with suboptimal concentrations of anti-CD40 plus IL-4 in the presence or absence of APRIL or anti-TACI. IgG(1) and IgE production was measured by means of ELISA. mRNA for Cgamma(1) and Cepsilon germ-line transcripts, activation-induced cytidine deaminase, and mature gamma(1) and epsilon transcripts were measured by means of RT-PCR. Plasmablasts were enumerated by using syndecan-1/CD138 staining. Interferon regulatory factor 4, B lymphocyte-induced maturation protein 1, and IL6 mRNA expression was measured by using quantitative PCR. RESULTS: TACI ligation enhanced IgG(1) and IgE secretion by naive murine B cells stimulated by anti-CD40 plus IL-4, with little effect on B-cell proliferation or class switch recombination. In contrast, TACI ligation of anti-CD40 plus IL-4-stimulated B cells induced a significant increase in syndecans-1/CD138-positive cells. TACI ligation caused a modest but significant increase in the expression of interferon regulatory factor 4, with no detectable change in B lymphocyte-induced maturation protein 1 expression. CONCLUSION: TACI and CD40 signaling converge to promote B-cell differentiation into plasmablasts. CLINICAL IMPLICATIONS: Our data suggest that TACI dysfunction could contribute to the impaired antibody response to T-dependent antigens in common variable immunodeficiency.


Subject(s)
CD40 Antigens/physiology , Plasma Cells/cytology , Transmembrane Activator and CAML Interactor Protein/physiology , Animals , B-Lymphocytes/physiology , Cell Differentiation , Cytidine Deaminase/genetics , Immunoglobulin E/biosynthesis , Interferon Regulatory Factors/genetics , Interleukin-4/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
17.
Blood ; 109(2): 729-39, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16960154

ABSTRACT

Hodgkin lymphoma (HL) originates from the clonal expansion of malignant Hodgkin and Reed-Sternberg (HRS) cells. These B-cell-derived elements constitute less than 10% of the tumoral mass. The remaining tissue is comprised of an inflammatory infiltrate that includes myeloid cells. Myeloid cells activate B cells by producing BAFF and APRIL, which engage TACI, BCMA, and BAFF-R receptors on the B cells. Here, we studied the role of BAFF and APRIL in HL. Inflammatory and HRS cells from HL tumors expressed BAFF and APRIL. Unlike their putative germinal center B-cell precursors, HRS cells lacked BAFF-R, but expressed TACI and BCMA, a phenotype similar to that of plasmacytoid B cells. BAFF and APRIL enhanced HRS cell survival and proliferation by delivering nonredundant signals via TACI and BCMA receptors through both autocrine and paracrine pathways. These signals caused NF-kappaB activation; Bcl-2, Bcl-xL, and c-Myc up-regulation; and Bax down-regulation, and were amplified by APRIL-binding proteoglycans on HRS cells. Interruption of BAFF and APRIL signaling by TACI-Ig decoy receptor, which binds to and neutralizes BAFF and APRIL, or by small-interfering RNAs targeting BAFF, APRIL, TACI, and BCMA inhibited HRS cell accumulation in vitro and might attenuate HL expansion in vivo.


Subject(s)
B-Cell Activating Factor/metabolism , B-Cell Maturation Antigen/biosynthesis , B-Lymphocytes/immunology , Hodgkin Disease/metabolism , Transmembrane Activator and CAML Interactor Protein/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , B-Cell Activating Factor/pharmacology , B-Cell Maturation Antigen/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Profiling , Hodgkin Disease/diagnosis , Hodgkin Disease/pathology , Humans , Immunophenotyping , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction , Transmembrane Activator and CAML Interactor Protein/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
18.
Blood ; 109(7): 2961-7, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17119122

ABSTRACT

Transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) is a member of the tumor necrosis factor (TNF) receptor family that serves as a receptor for B-cell activating factor of the TNF family (BAFF) and as a proliferation-inducing ligand (APRIL). Although TACI is reported to function as a positive or negative regulator for B-cell responses, its roles remain elusive. Experiments using TACI siRNA into B cells indicated that TACI positively regulated APRIL-induced IgA production in collaboration with heparan sulfate proteoglycans (HSPG). Furthermore, TACI negatively regulated BAFF-induced B-cell proliferation and production of IgA and IgG. In addition, B cells treated with heparitinase to denature HSPG showed that HSPG is essential for APRIL-induced B-cell responses such as B-cell proliferation, IgG and IgA production, induction of activation-induced cytidine deaminase (AID), and noncanonical NF-kappaB2. In contrast, phosphorylation of physiological AID kinase, protein kinase A (PKA), was dependent on TACI. Importantly, coligation of TACI and HSPG by specific antibodies, but not by TACI or HSPG ligation itself, could induce the phosphorylation of PKA and IgA production instead of APRIL. Our findings indicate that simultaneous binding of TACI and HSPG on B cells with APRIL is crucial for IgA production.


Subject(s)
Heparan Sulfate Proteoglycans/immunology , Immunoglobulin A/biosynthesis , Transmembrane Activator and CAML Interactor Protein/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , B-Cell Activating Factor/immunology , B-Cell Activating Factor/pharmacology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Heparan Sulfate Proteoglycans/chemistry , Humans , In Vitro Techniques , Polysaccharide-Lyases , Protein Binding , Protein Denaturation , RNA, Small Interfering/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Transmembrane Activator and CAML Interactor Protein/antagonists & inhibitors , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...