Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nature ; 597(7874): 92-96, 2021 09.
Article in English | MEDLINE | ID: mdl-34433968

ABSTRACT

Atherosclerotic cardiovascular disease causes heart attacks and strokes, which are the leading causes of mortality worldwide1. The formation of atherosclerotic plaques is initiated when low-density lipoproteins bind to heparan-sulfate proteoglycans (HSPGs)2 and become trapped in the subendothelial space of large and medium size arteries, which leads to chronic inflammation and remodelling of the artery wall2. A proliferation-inducing ligand (APRIL) is a cytokine that binds to HSPGs3, but the physiology of this interaction is largely unknown. Here we show that genetic ablation or antibody-mediated depletion of APRIL aggravates atherosclerosis in mice. Mechanistically, we demonstrate that APRIL confers atheroprotection by binding to heparan sulfate chains of heparan-sulfate proteoglycan 2 (HSPG2), which limits the retention of low-density lipoproteins, accumulation of macrophages and formation of necrotic cores. Indeed, antibody-mediated depletion of APRIL in mice expressing heparan sulfate-deficient HSPG2 had no effect on the development of atherosclerosis. Treatment with a specific anti-APRIL antibody that promotes the binding of APRIL to HSPGs reduced experimental atherosclerosis. Furthermore, the serum levels of a form of human APRIL protein that binds to HSPGs, which we termed non-canonical APRIL (nc-APRIL), are associated independently of traditional risk factors with long-term cardiovascular mortality in patients with atherosclerosis. Our data reveal properties of APRIL that have broad pathophysiological implications for vascular homeostasis.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Heparan Sulfate Proteoglycans/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , B-Cell Maturation Antigen/metabolism , Binding Sites , Cardiovascular Diseases/blood , Cardiovascular Diseases/mortality , Female , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/blood , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency
2.
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
3.
Antiviral Res ; 144: 130-137, 2017 08.
Article in English | MEDLINE | ID: mdl-28619678

ABSTRACT

There is significant need to develop a single-dose rabies vaccine to replace the current multi-dose rabies vaccine regimen and eliminate the requirement for rabies immune globulin in post-exposure settings. To accomplish this goal, rabies virus (RABV)-based vaccines must rapidly activate B cells to secrete antibodies which neutralize pathogenic RABV before it enters the CNS. Increased understanding of how B cells effectively respond to RABV-based vaccines may improve efforts to simplify post-exposure prophylaxis (PEP) regimens. Several studies have successfully employed the TNF family cytokine a proliferation-inducing ligand (APRIL) as a vaccine adjuvant. APRIL binds to the receptors TACI and B cell maturation antigen (BCMA)-expressed by B cells in various stages of maturation-with high affinity. We discovered that RABV-infected primary murine B cells upregulate APRIL ex vivo. Cytokines present at the time of antigen exposure affect the outcome of vaccination by influencing T and B cell activation and GC formation. Therefore, we hypothesized that the presence of APRIL at the time of RABV-based vaccine antigen exposure would support the generation of protective antibodies against RABV glycoprotein (G). In an effort to improve the response to RABV vaccination, we constructed and characterized a live recombinant RABV-based vaccine vector which expresses murine APRIL (rRABV-APRIL). Immunogenicity testing in mice demonstrated that expressing APRIL from the RABV genome does not impact the primary antibody response against RABV G compared to RABV alone. In order to evaluate the necessity of APRIL for the response to rabies vaccination, we compared the responses of APRIL-deficient and wild-type mice to immunization with rRABV. APRIL deficiency does not affect the primary antibody response to vaccination. Furthermore, APRIL expression by the vaccine did not improve the generation of long-lived antibody-secreting plasma cells (PCs) as serum antibody levels were equivalent in response to rRABV-APRIL and the vector eight weeks after immunization. Moreover, APRIL is dispensable for the long-lived antibody-secreting PC response to rRABV vaccination as anti-RABV G IgG levels were similar in APRIL-deficient and wild-type mice six months after vaccination. Mice lacking the APRIL receptor TACI demonstrated primary anti-RABV G antibody responses similar to wild-type mice following immunization with the vaccine vector indicating that this response is independent of TACI-mediated signals. Collectively, our findings demonstrate that APRIL and associated TACI signaling is dispensable for the immune response to RABV-based vaccination.


Subject(s)
Adjuvants, Immunologic/metabolism , Rabies Vaccines/immunology , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Antibodies, Viral/blood , Immunoglobulin G/blood , Mice , Mice, Knockout , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Vaccines, Attenuated/immunology
4.
J Immunol ; 191(3): 1154-63, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23797666

ABSTRACT

CD1d-restricted invariant NKT (iNKT) cells boost humoral immunity to T-dependent Ags that are coadministered with the CD1d-binding glycolipid Ag α-galactosylceramide (α-GC). Observations that mice lacking iNKT cells have decaying Ab responses following vaccination have led to the hypothesis that iNKT cells express plasma cell (PC) survival factors that sustain specific Ab titers. Bone marrow chimeric mice in which the entire hematopoietic compartment or iNKT cells selectively lacked BAFF, a proliferation-inducing ligand (APRIL), or both BAFF and APRIL were created and immunized with nitrophenol hapten-conjugated keyhole limpet hemocyanin adsorbed to Imject aluminum hydroxide-containing adjuvant or mixed with α-GC. In comparison with BAFF- or APRIL-sufficient bone marrow chimeras, absence of hematopoietic compartment- and iNKT-derived BAFF and APRIL was associated with rapidly decaying Ab titers and reduced PC numbers. The iNKT cell-derived BAFF or APRIL assumed a greater role in PC survival when α-GC was used as the adjuvant for immunization. These results show that iNKT cell-derived BAFF and APRIL each contribute to survival of PCs induced by immunization. This study sheds new light on the mechanisms through which iNKT cells impact humoral immunity and may inform design of vaccines that incorporate glycolipid adjuvants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies/blood , B-Cell Activating Factor/metabolism , Natural Killer T-Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/immunology , Animals , Antigens, CD1d/immunology , B-Cell Activating Factor/deficiency , B-Cell Activating Factor/genetics , Bone Marrow Cells , Female , Galactosylceramides/administration & dosage , Galactosylceramides/immunology , Hemocyanins/administration & dosage , Hemocyanins/immunology , Immunity, Humoral , Immunization , Mice , Mice, Knockout , Plasma Cells/metabolism , Transplantation Chimera , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Vaccination
6.
Arthritis Rheum ; 64(5): 1610-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22127792

ABSTRACT

OBJECTIVE: To determine the role of APRIL in the development of systemic lupus erythematosus (SLE) in mice. METHODS: Wild-type (WT) NZM 2328, NZM. April(-/-) , NZM.Baff(-/-) , and NZM.Baff(-/-) .April(-/-) mice were evaluated for lymphocyte phenotype by flow cytometry, for serum total IgG and IgG autoantibody levels by enzyme-linked immunosorbent assay, for glomerular deposition of IgG and C3 by immunofluorescence, for renal changes by histopathology, and for clinical disease by laboratory assessment (severe proteinuria). RESULTS: In comparison to WT mice, NZM.April(-/-) mice harbored increased spleen B cells, T cells, and plasma cells (PCs), increased serum levels of IgG antichromatin antibodies, and decreased numbers of bone marrow (BM) PCs. Glomerular deposition of IgG and C3 was similar in NZM.April(-/-) mice and WT mice, renal changes on histopathology tended to be more severe in NZM.April(-/-) mice than in WT mice, and development of clinical disease was identical in NZM.April(-/-) mice and WT mice. BM (but not spleen) PCs and serum IgG antichromatin and anti-double-stranded DNA antibody levels were lower in NZM.Baff(-/-) .April(-/-) mice than in NZM.Baff(-/-) mice, whereas renal immunopathology in each cohort was equally mild. CONCLUSION: APRIL is dispensable for the development of full-blown SLE in NZM mice. Moreover, the elimination of both APRIL and BAFF had no discernible effect on the development of renal immunopathology or clinical disease beyond that of elimination of BAFF alone. The reduction in BM PCs in hosts doubly deficient in APRIL and BAFF beyond that in hosts deficient only in BAFF raises concern that combined antagonism of APRIL and BAFF may lead to greater immunosuppression without a concomitant increase in therapeutic efficacy.


Subject(s)
B-Cell Activating Factor/deficiency , Lupus Erythematosus, Systemic/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , B-Cell Activating Factor/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Biomarkers/metabolism , Bone Marrow Cells , Complement C3/immunology , Complement C3/metabolism , Disease Models, Animal , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunosuppression Therapy , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred NZB , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism , Plasma Cells/pathology , Species Specificity , Spleen/immunology , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
7.
Eur J Immunol ; 38(12): 3450-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19016524

ABSTRACT

A proliferation-inducing ligand (APRIL or TNFSF13) shares receptors with B-cell activation factor of the TNF family (BAFF) on B and T cells. Although much is known about the function of APRIL in B cells, its role in T cells remains unclear. Blocking both BAFF and APRIL suggested that BAFF and/or APRIL contributed to collagen-induced arthritis (CIA); however, the role of APRIL alone in CIA remained unresolved. We show here that, in vitro, our newly generated APRIL(-/-) mice exhibited increased T-cell proliferation, enhanced Th2 cytokine production under non-polarizing conditions, and augmented IL-13 and IL-17 production under Th2 polarizing conditions. Upon immunization with OVA and aluminum potassium sulfate, APRIL(-/-) mice responded with an increased antigen-specific IgG1 response. We also show that in APRIL(-/-) mice, the incidence of CIA was significantly reduced compared with WT mice in parallel with diminished levels of antigen-specific IgG2a autoantibody and IL-17 production. Our data indicate that APRIL plays an important role in the regulation of cytokine production and that APRIL-triggered signals contribute to arthritis. Blockade of APRIL thus may be a valuable adjunct in the treatment of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Interleukin-17/biosynthesis , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Autoantibodies/immunology , Cell Proliferation , Collagen Type II/immunology , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
8.
Mol Immunol ; 45(11): 3050-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18471884

ABSTRACT

A proliferation inducing ligand (APRIL) is implicated in the regulation of class switch recombination to IgA in T-independent B cell responses. Since B cells play an important role in the immunity to influenza A virus and resistance against the virus is partly controlled by T-independent IgA B cell responses, we studied the role of APRIL during an influenza A infection in vivo. APRIL transgenic, wild-type and APRIL deficient mice were intranasally infected with a non-lethal dose of a mouse adapted strain of influenza A. Compared to wild-type mice, APRIL deficient mice showed a twofold reduction in the amount of macrophages in the lungs and a tendency towards decreased granulocyte influx in the early leukocyte recruitment phase. Although the T cell immune response against influenza was unaffected, APRIL Tg mice showed prolonged influenza-specific IgM production and differential class switching. Unexpectedly, the IgA B cell response was completely T helper cell dependent and also not affected by the absence or presence of APRIL. In addition, viral clearance and recovery from the infection was not influenced by APRIL. Combined these results indicate that APRIL affects specific aspects of the anti-influenza response, but plays a limited role in disease recovery.


Subject(s)
Antibody Formation/immunology , Cell Movement , Influenza A virus/immunology , Leukocytes/immunology , Leukocytes/virology , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , B-Lymphocytes/immunology , Body Weight , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Count , Chemokine CCL2/immunology , Interleukin-6/immunology , Lectins, C-Type , Leukocytes/cytology , Lung/cytology , Lung/immunology , Lung/virology , Macrophages/cytology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Up-Regulation , Viral Load
9.
J Immunol ; 180(6): 3655-9, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18322170

ABSTRACT

Memory B (B(MEM)) cells and long-lived bone marrow plasma cells (BM-PCs) persist within local environmental survival niches that afford cellular longevity. However, the factors supporting B(MEM) cell survival within the secondary lymphoid organs and allowing BM-PC persistence in the bone marrow remain poorly characterized. We report herein that long-lived B(MEM) cell survival and function are completely independent of BAFF (B cell-activating factor of the TNF family) or APRIL (a proliferation-inducing ligand). Thus, B(MEM) cells represent the only mature B2 lineage subset whose survival is independent of these ligands. We have previously shown that the TNFR family member receptor BCMA (B cell maturation Ag) is a critical survival receptor for BM-PC survival in vivo. We identify in this study the ligands critical for BM-PC survival and show that either BAFF or APRIL supports the survival of BM-PCs in vivo. These data define the BAFF/APRIL-dependent and -independent components of long-lived humoral immunity.


Subject(s)
B-Cell Activating Factor/physiology , B-Lymphocyte Subsets/immunology , Immunologic Memory , Tumor Necrosis Factor Ligand Superfamily Member 13/physiology , Animals , Antibody Formation/genetics , B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/biosynthesis , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/metabolism , B-Cell Maturation Antigen/biosynthesis , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/metabolism , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Survival/genetics , Cell Survival/immunology , Immunologic Memory/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Transmembrane Activator and CAML Interactor Protein/biosynthesis , Transmembrane Activator and CAML Interactor Protein/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
10.
Allergol Immunopathol (Madr) ; 34(6): 263-75, 2006.
Article in English | MEDLINE | ID: mdl-17173844

ABSTRACT

Common variable immunodeficiency (CVID) is a heterogeneous entity characterized by an impaired ability to produce antibodies. The failure is localized in partially mature B lymphocytes, though T lymphocyte abnormalities are occasionally present. This deficiency affects antibody synthesis and class switch from IgD and IgM, to IgG and IgA. CVID is related to selective IgA deficiency, and both abnormalities may coincide in one same family, and evolve from one to another in the same patient. The symptoms generally manifest in adults, but can occur at any age, even in infancy. Recurrent bacterial infections or pneumonias are frequent, and may be complicated by gastrointestinal problems, granulomas, autoimmune disorders or malignancies. A defect in memory B cells seems to condition the clinical severity. Recently, several mutations in genes encoding for molecules (CD19, TACI, ICOS) involved in B cell survival and isotype switch have been identified in patients with CVID. Nevertheless, genetic abnormalities have been found in less than 25 % of cases with CVID; the underlying mechanism thus remains unknown in the majority of CVID patients, and research in this field must continue.


Subject(s)
Common Variable Immunodeficiency , Adult , Aged , Animals , Antibody Formation , B-Cell Activating Factor/deficiency , B-Cell Activating Factor/immunology , B-Cell Activating Factor/physiology , B-Lymphocytes/pathology , CD40 Ligand/analysis , Cell Differentiation , Child , Chronic Disease , Cimetidine/therapeutic use , Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/drug therapy , Common Variable Immunodeficiency/epidemiology , Common Variable Immunodeficiency/etiology , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , Disease Models, Animal , Granuloma/etiology , Humans , Immunoglobulin Class Switching , Immunoglobulin G/therapeutic use , Immunoglobulins/biosynthesis , Incidence , Infections/complications , Lung Diseases/etiology , Mice , Mice, Knockout , Middle Aged , Transmembrane Activator and CAML Interactor Protein/deficiency , Transmembrane Activator and CAML Interactor Protein/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/deficiency , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...