Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Immunity ; 50(2): 477-492.e8, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30737146

ABSTRACT

Resistance to checkpoint-blockade treatments is a challenge in the clinic. We found that although treatment with combined anti-CTLA-4 and anti-PD-1 improved control of established tumors, this combination compromised anti-tumor immunity in the low tumor burden (LTB) state in pre-clinical models as well as in melanoma patients. Activated tumor-specific T cells expressed higher amounts of interferon-γ (IFN-γ) receptor and were more susceptible to apoptosis than naive T cells. Combination treatment induced deletion of tumor-specific T cells and altered the T cell repertoire landscape, skewing the distribution of T cells toward lower-frequency clonotypes. Additionally, combination therapy induced higher IFN-γ production in the LTB state than in the high tumor burden (HTB) state on a per-cell basis, reflecting a less exhausted immune status in the LTB state. Thus, elevated IFN-γ secretion in the LTB state contributes to the development of an immune-intrinsic mechanism of resistance to combination checkpoint blockade, highlighting the importance of achieving the optimal magnitude of immune stimulation for successful combination immunotherapy strategies.


Subject(s)
Antibodies, Monoclonal/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Interferon-gamma/pharmacology , Neoplasms, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , Animals , Antibodies, Monoclonal/immunology , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Clonal Deletion/drug effects , Clonal Deletion/immunology , Drug Resistance, Neoplasm/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology
2.
J Immunol ; 203(1): 58-75, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31109957

ABSTRACT

CD40 is a costimulatory receptor on APCs that is critical for the induction and maintenance of humoral and cell-mediated immunity. Accordingly, CD40 and its ligand, CD40L, have long been considered targets for the treatment of autoimmune diseases. We developed a rat/mouse chimeric anti-mouse CD40 antagonist mAb, 201A3, and evaluated its ability to alleviate murine lupus. Treatment of NZB/W-F1 mice with 201A3 after the onset of severe proteinuria rapidly reversed established severe proteinuria and nephritis and largely restored normal glomerular and tubular morphology. This coincided with a normalization of the expression of genes associated with proteinuria and injury by kidney parenchymal cells. Anti-CD40 treatment also prevented and reversed loss of saliva production and sialadenitis. These effects on kidney and salivary gland function were confirmed using mice of a second strain, MRL/Mp-lpr/lpr, and extended to alleviating joint inflammation. Immunologically, anti-CD40 treatment disrupted multiple processes that contribute to the pathogenesis of systemic lupus erythematosus (SLE), including autoreactive B cell activation, T effector cell function in target tissues, and type I IFN production. This ability to disrupt disease-critical immunological mechanisms, to reverse glomerular and tubular injury at the cellular and gene expression levels, and to confer exceptional therapeutic efficacy suggests that CD40 is a central disease pathway in murine SLE. Thus, a CD40 antagonist Ab could be an effective therapeutic in the treatment of SLE.


Subject(s)
Antibodies, Blocking/therapeutic use , B-Lymphocytes/immunology , CD40 Antigens/immunology , Immunotherapy/methods , Kidney Glomerulus/pathology , Lupus Erythematosus, Systemic/therapy , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/immunology , Animals , Autoantigens/immunology , Cells, Cultured , Disease Models, Animal , Humans , Interferon Type I/metabolism , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred MRL lpr , Mice, Inbred NZB , Proteinuria , Rats , Salivary Elimination
3.
Eur J Immunol ; 49(1): 192-194, 2019 01.
Article in English | MEDLINE | ID: mdl-30359469

ABSTRACT

The germinal center reaction is essential for efficient humoral immunity, but it can also give rise to B cell lymphomas. Cre/loxP-mediated conditional gene knock-out or knock-in can be used for the genetic manipulation of germinal center B cells in vivo. Here we present a novel allele, Cγ1-CreERT2, that allows for timed activation of Cre recombinase in a small fraction of germinal center B cells. This allele will be useful to study normal and malignant germinal center B cell development in vivo.


Subject(s)
B-Lymphocytes/physiology , Gene Knock-In Techniques/methods , Gene Knockout Techniques/methods , Germinal Center/immunology , Integrases/genetics , Alleles , Animals , Cell Differentiation , Humans , Mice
4.
Exp Dermatol ; 28(2): 113-120, 2019 02.
Article in English | MEDLINE | ID: mdl-30417427

ABSTRACT

Psoriasis vulgaris (PV) results from activation of IL-23/Th17 immune pathway and is further amplified by cytokines/chemokines from skin cells. Among skin-derived pro-inflammatory cytokines, IL-36 family members are highly upregulated in PV patients and play a critical role in general pustular psoriasis. However, there is limited data showing crosstalk between the IL-23 and IL-36 pathways in PV. Herein, potential attenuation of skin inflammation in the IL-23-induced mouse model of psoriasiform dermatitis by functional inhibition of IL-36 receptor (IL-36R) was interrogated. Anti-mouse IL-36R monoclonal antibodies (mAbs) were generated and validated in vitro by inhibiting IL-36α-induced secretion of CXCL1 from NIH 3T3 cells. Antibody target engagement was demonstrated by inhibition of CXCL1 production in a novel acute model of IL-36α systemic injection in mice. In addition, anti-IL-36R mAbs inhibited tissue inflammation and inflammatory gene expression in an IL-36α ear injection model of psoriasiform dermatitis demonstrating engagement of the target in the ear skin. To elucidate the possible role of IL-36 signalling in IL-23/Th17 pathway, the ability of anti-IL-36R mAbs to inhibit skin inflammation in an IL-23 ear injection model was assessed. Inhibiting the IL-36 pathway resulted in significant attenuation of skin thickening and psoriasis-relevant gene expression. Taken together, these data suggest a role for IL-36 signalling in the IL-23/Th17 signalling axis in PV.


Subject(s)
Antibodies, Monoclonal/immunology , Dermatitis/immunology , Inflammation/immunology , Interleukins/immunology , Psoriasis/immunology , Receptors, Interleukin/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Chemokine CXCL1/metabolism , Cytokines/metabolism , Dermatitis/therapy , Disease Models, Animal , Female , Gene Expression Profiling , Inflammation/metabolism , Interleukin-1/immunology , Interleukin-23/pharmacology , Ligands , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Psoriasis/therapy , Rats , Rats, Sprague-Dawley , Receptors, Interleukin/immunology , Receptors, Interleukin-1/immunology , Signal Transduction , Skin/metabolism , Skin/pathology , Th17 Cells/cytology
5.
Immunity ; 29(4): 615-27, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18835195

ABSTRACT

Fas is highly expressed in activated and germinal center (GC) B cells but can potentially be inactivated by misguided somatic hypermutation. We employed conditional Fas-deficient mice to investigate the physiological functions of Fas in various B cell subsets. B cell-specific Fas-deficient mice developed fatal lymphoproliferation due to activation of B cells and T cells. Ablation of Fas specifically in GC B cells reproduced the phenotype, indicating that the lymphoproliferation initiates in the GC environment. B cell-specific Fas-deficient mice also showed an accumulation of IgG1(+) memory B cells expressing high amounts of CD80 and the expansion of CD28-expressing CD4(+) Th cells. Blocking T cell-B cell interaction and GC formation completely prevented the fatal lymphoproliferation. Thus, Fas-mediated selection of GC B cells and the resulting memory B cell compartment is essential for maintaining the homeostasis of both T and B lymphocytes.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , T-Lymphocytes/immunology , fas Receptor/metabolism , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , B-Lymphocytes/metabolism , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CTLA-4 Antigen , Cell Communication , Cell Differentiation , Cell Proliferation , Cytokines/blood , Germinal Center/metabolism , Homeostasis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , T-Lymphocytes/metabolism , fas Receptor/deficiency , fas Receptor/immunology
6.
J Exp Med ; 204(4): 747-58, 2007 Apr 16.
Article in English | MEDLINE | ID: mdl-17420268

ABSTRACT

We describe a mouse strain in which B cell development relies either on the expression of membrane-bound immunoglobulin (Ig) gamma1 or mu heavy chains. Progenitor cells expressing gamma1 chains from the beginning generate a peripheral B cell compartment of normal size with all subsets, but a partial block is seen at the pro- to pre-B cell transition. Accordingly, gamma1-driven B cell development is disfavored in competition with developing B cells expressing a wild-type (WT) IgH locus. However, the mutant B cells display a long half-life and accumulate in the mature B cell compartment, and even though partial truncation of the Ig alpha cytoplasmic tail compromises their development, it does not affect their maintenance, as it does in WT cells. IgG1-expressing B cells showed an enhanced Ca(2+) response upon B cell receptor cross-linking, which was not due to a lack of inhibition by CD22. The enhanced Ca(2+) response was also observed in mature B cells that had been switched from IgM to IgG1 expression in vivo. Collectively, these results suggest that the gamma1 chain can exert a unique signaling function that can partially replace that of the Ig alpha/beta heterodimer in B cell maintenance and may contribute to memory B cell physiology.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD79 Antigens/metabolism , Immunoglobulins/metabolism , Receptors, Antigen, B-Cell/metabolism , Sialic Acid Binding Ig-like Lectin 2/metabolism , Signal Transduction , Animals , Calcium/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Dimerization , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulins/genetics , Immunoglobulins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Protein Binding , Sialic Acid Binding Ig-like Lectin 2/genetics , Spleen/cytology , Spleen/metabolism , Toll-Like Receptors/metabolism
7.
J Immunol ; 187(6): 2853-8, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21841126

ABSTRACT

Igα serine 191 and 197 and threonine 203, which are located in proximity of the Igα ITAM, dampen Igα ITAM tyrosine phosphorylation. In this study, we show that mice with targeted mutations of Igα S191, 197, and T203 displayed elevated serum IgG2c and IgG2b concentrations and had elevated numbers of IgG2c- and IgG2b-secreting cells in the bone marrow. BCR-induced Igα tyrosine phosphorylation was slightly increased in splenic B cells. Our results suggest that Igα serine/threonines limit formation of IgG2c- and IgG2b-secreting bone marrow plasma cells, possibly by fine-tuning Igα tyrosine-mediated BCR signaling.


Subject(s)
Bone Marrow Cells/cytology , Mutation/immunology , Plasma Cells/cytology , Receptors, Antigen, B-Cell/chemistry , Receptors, Antigen, B-Cell/immunology , Amino Acid Sequence , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Separation , Cytoplasm/chemistry , Cytoplasm/immunology , Cytoplasm/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, Antigen, B-Cell/genetics , Serine/chemistry , Serine/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Threonine/chemistry , Threonine/immunology , Tyrosine/metabolism
8.
Viruses ; 14(5)2022 05 04.
Article in English | MEDLINE | ID: mdl-35632703

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a recently emerged human coronavirus. COVID-19 vaccines have proven to be successful in protecting the vaccinated from infection, reducing the severity of disease, and deterring the transmission of infection. However, COVID-19 vaccination faces many challenges, such as the decline in vaccine-induced immunity over time, and the decrease in potency against some SARS-CoV-2 variants including the recently emerged Omicron variant, resulting in breakthrough infections. The challenges that COVID-19 vaccination is facing highlight the importance of the discovery of antivirals to serve as another means to tackle the pandemic. To date, neutralizing antibodies that block viral entry by targeting the viral spike protein make up the largest class of antivirals that has received US FDA emergency use authorization (EUA) for COVID-19 treatment. In addition to the spike protein, other key targets for the discovery of direct-acting antivirals include viral enzymes that are essential for SARS-CoV-2 replication, such as RNA-dependent RNA polymerase and proteases, as judged by US FDA approval for remdesivir, and EUA for Paxlovid (nirmatrelvir + ritonavir) for treating COVID-19 infections. This review presents an overview of the current status and future direction of antiviral drug discovery for treating SARS-CoV-2 infections, covering important antiviral targets such as the viral spike protein, non-structural protein (nsp) 3 papain-like protease, nsp5 main protease, and the nsp12/nsp7/nsp8 RNA-dependent RNA polymerase complex.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Drug Discovery , Antiviral Agents/pharmacology , COVID-19 Vaccines , Coronavirus 3C Proteases/antagonists & inhibitors , Humans , RNA-Dependent RNA Polymerase/antagonists & inhibitors , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Viral Proteins/metabolism
9.
Science ; 375(6578): eabl6251, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34855508

ABSTRACT

Many studies have examined the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants on neutralizing antibody activity after they have become dominant strains. Here, we evaluate the consequences of further viral evolution. We demonstrate mechanisms through which the SARS-CoV-2 receptor binding domain (RBD) can tolerate large numbers of simultaneous antibody escape mutations and show that pseudotypes containing up to seven mutations, as opposed to the one to three found in previously studied variants of concern, are more resistant to neutralization by therapeutic antibodies and serum from vaccine recipients. We identify an antibody that binds the RBD core to neutralize pseudotypes for all tested variants but show that the RBD can acquire an N-linked glycan to escape neutralization. Our findings portend continued emergence of escape variants as SARS-CoV-2 adapts to humans.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immune Evasion , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , BNT162 Vaccine/immunology , Betacoronavirus/immunology , COVID-19/immunology , COVID-19/virology , Cross Reactions , Cryoelectron Microscopy , Crystallography, X-Ray , Epitopes , Evolution, Molecular , Humans , Models, Molecular , Mutation , Polysaccharides/analysis , Protein Binding , Protein Domains , Receptors, Coronavirus/chemistry , Receptors, Coronavirus/metabolism , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Viral Pseudotyping
10.
J Immunol ; 183(1): 732-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19535619

ABSTRACT

CCL2 is a key CC chemokine that has been implicated in a variety of inflammatory autoimmune diseases and in tumor progression and it is therefore an important target for therapeutic intervention in these diseases. Soluble receptor-based therapy is a known approach for neutralizing the in vivo functions of soluble mediators. Owing to the complexity of seven-transmembrane G protein-coupled receptors, efforts to generate neutralizing soluble chemokine receptors have so far failed. We developed a strategy that is based on the generation of short recombinant proteins encoding different segments of a G protein-coupled receptor, and tested the ability of each of them to bind and neutralize its target chemokine. We show that a fusion protein comprised of as few as 20 aa of the third extracellular (E3) domain of the CCL2 receptor, stabilized by the IgG H chain Fc domain (E3-IgG or BL-2030), selectively binds CCL2 and CCL16 and effectively neutralizes their biological activities. More importantly, E3-IgG (BL-2030) could effectively suppress the in vivo biological activity of CCL2, attenuating ongoing experimental autoimmune encephalomyelitis, as well as the development of human prostate tumor in SCID mice.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/physiology , Receptors, CCR2/physiology , Recombinant Fusion Proteins/physiology , Animals , Cell Line , Cell Line, Tumor , Cell Migration Inhibition/immunology , Cell Proliferation , Chemokine CCL2/metabolism , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, CCR2/metabolism , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/metabolism
11.
J Exp Med ; 198(10): 1609-19, 2003 Nov 17.
Article in English | MEDLINE | ID: mdl-14623914

ABSTRACT

In B lymphocytes, immunoglobulin (Ig)M receptors drive development and construction of naive repertoire, whereas IgG receptors promote formation of the memory B cell compartment. This isotype switching process requires appropriate B cell activation and T cell help. In the absence of T cell help, activated B cells undergo Fas-mediated apoptosis, a peripheral mechanism contributing to the establishment of self-tolerance. Using Igmicro-deficient microMT mouse model, where B cell development is blocked at pro-B stage, here we show an alternative developmental pathway used by isotype-switched B cell precursors. We find that isotype switching occurs normally in B cell precursors and is T independent. Ongoing isotype switching was found in both normal and microMT B cell development as reflected by detection of IgG1 germline and postswitch transcripts as well as activation-induced cytidine deaminase expression, resulting in the generation of IgG-expressing cells. These isotype-switched B cells are negatively selected by Fas pathway, as blocking the Fas/FasL interaction rescues the development of isotype-switched B cells in vivo and in vitro. Similar to memory B cells, isotype-switched B cells have a marginal zone phenotype. We suggest a novel developmental pathway used by isotype-switched B cell precursors that effectively circumvents peripheral tolerance requirements. This developmental pathway, however, is strictly controlled by Fas/FasL interaction to prevent B cell autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Clonal Deletion/immunology , Membrane Glycoproteins/immunology , fas Receptor/immunology , Animals , Cell Differentiation/immunology , Fas Ligand Protein , Immunoglobulin Class Switching/immunology , Mice , fas Receptor/genetics
12.
Science ; 363(6428): 748-753, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30765568

ABSTRACT

B2 cells engage in classical antibody responses, whereas B1 cells are considered carriers of innate immunity, biased toward recognizing epitopes present on the surfaces of common pathogens and self antigens. To explore the role of B cell antigen receptor (BCR) specificity in driving B1 cell differentiation, we developed a transgenic system allowing us to change BCR specificity in B cells in an inducible and programmed manner. Mature B2 cells differentiated into bona fide B1 cells upon acquisition of a B1 cell-typical self-reactive BCR through a phase of proliferative expansion. Thus, B2 cells have B1 cell differentiation potential in addition to their classical capacity to differentiate into memory and plasma cells, and B1 differentiation can be instructed by BCR-mediated self-reactivity, in the absence of B1-lineage precommitment.


Subject(s)
B-Lymphocyte Subsets/cytology , Cell Differentiation/immunology , Cell Plasticity/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Cell Differentiation/genetics , Cell Lineage , Cell Plasticity/genetics , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Mice , Mice, Transgenic , Receptors, Antigen, B-Cell/genetics , Transcriptome
13.
J Clin Invest ; 129(1): 349-363, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30530991

ABSTRACT

While immune checkpoint blockade leads to potent antitumor efficacy, it also leads to immune-related adverse events in cancer patients. These toxicities stem from systemic immune activation resulting in inflammation of multiple organs, including the gastrointestinal tract, lung, and endocrine organs. We developed a dual variable domain immunoglobulin of anti-CTLA4 antibody (anti-CTLA4 DVD, where CTLA4 is defined as cytotoxic T lymphocyte-associated antigen-4) possessing an outer tumor-specific antigen-binding site engineered to shield the inner anti-CTLA4-binding domain. Upon reaching the tumor, the outer domain was cleaved by membrane type-serine protease 1 (MT-SP1) present in the tumor microenvironment, leading to enhanced localization of CTLA4 blockade. Anti-CTLA4 DVD markedly reduced multiorgan immune toxicity by preserving tissue-resident Tregs in Rag 1-/- mice that received naive donor CD4+ T cells from WT C57BL/6j mice. Moreover, anti-CTLA4 DVD induced potent antitumor effects by decreasing tumor-infiltrating Tregs and increasing the infiltration of antigen-specific CD8+ T lymphocytes in TRAMP-C2-bearing C57BL/6j mice. Treg depletion was mediated through the antibody-dependent cellular cytotoxicity (ADCC) mechanism, as anti-CTLA4 without the FcγR-binding portion (anti-CTLA4 DANA) spared Tregs, preventing treatment-induced toxicities. In summary, our results demonstrate an approach to anti-CTLA4 blockade that depletes tumor-infiltrating, but not tissue-resident, Tregs, preserving antitumor effects while minimizing toxicity. Thus, our tumor-conditional anti-CTLA4 DVD provides an avenue for uncoupling antitumor efficacy from immunotherapy-induced toxicities.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy , Neoplasms/therapy , Single-Chain Antibodies/pharmacology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Immunological/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Line, Tumor , HEK293 Cells , Humans , Immunity, Cellular , Male , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Single-Chain Antibodies/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Cancer Cell ; 18(6): 580-9, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21156282

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) comprises disease entities with distinct genetic profiles, including germinal center B cell (GCB)-like and activated B cell (ABC)-like DLBCLs. Major differences between these two subtypes include genetic aberrations leading to constitutive NF-κB activation and interference with terminal B cell differentiation through BLIMP1 inactivation, observed in ABC- but not GCB-DLBCL. Using conditional gain-of-function and/or loss-of-function mutagenesis in the mouse, we show that constitutive activation of the canonical NF-κB pathway cooperates with disruption of BLIMP1 in the development of a lymphoma that resembles human ABC-DLBCL. Our work suggests that both NF-κB signaling, as an oncogenic event, and BLIMP1, as a tumor suppressor, play causal roles in the pathogenesis of ABC-DLBCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/etiology , NF-kappa B/metabolism , Transcription Factors/physiology , Animals , Cell Proliferation , Germinal Center/physiology , I-kappa B Kinase/genetics , Mice , Mutation , Plasma Cells/physiology , Positive Regulatory Domain I-Binding Factor 1 , Transcription Factors/genetics
15.
Blood ; 111(3): 1448-55, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18006702

ABSTRACT

The Epstein-Barr virus (EBV) protein LMP1 is considered to be a functional homologue of the CD40 receptor. However, in contrast to the latter, LMP1 is a constitutively active signaling molecule. To compare B cell-specific LMP1 and CD40 signaling in an unambiguous manner, we generated transgenic mice conditionally expressing a CD40/LMP1 fusion protein, which retained the LMP1 cytoplasmic tail but has lost the constitutive activity of LMP1 and needs to be activated by the CD40 ligand. We show that LMP1 signaling can completely substitute CD40 signaling in B cells, leading to normal B-cell development, activation, and immune responses including class-switch recombination, germinal center formation, and somatic hypermutation. In addition, the LMP1-signaling domain has a unique property in that it can induce class-switch recombination to IgG1 independent of cytokines. Thus, our data indicate that LMP1 has evolved to imitate T-helper cell function allowing activation, proliferation, and differentiation of EBV-infected B cells independent of T cells.


Subject(s)
B-Lymphocytes/immunology , CD40 Antigens/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Signal Transduction/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies/immunology , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Antigens/metabolism , Cells, Cultured , Enzyme Activation , Germinal Center/immunology , Germinal Center/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Mutation/genetics , NF-kappa B/metabolism , Transgenes/genetics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
16.
Proc Natl Acad Sci U S A ; 103(19): 7396-401, 2006 May 09.
Article in English | MEDLINE | ID: mdl-16651521

ABSTRACT

Germinal centers (GCs) represent the main sites for the generation of high-affinity, class-switched antibodies during T cell-dependent antibody responses. To study gene function specifically in GC B cells, we generated Cgamma1-cre mice in which the expression of Cre recombinase is induced by transcription of the Ig gamma1 constant region gene segment (Cgamma1). In these mice, Cre-mediated recombination at the fas, Igbeta, IgH, and Rosa26 loci occurred in GC B cells as early as 4 days after immunization with T cell-dependent antigens and involved >85% of GC B cells at the peak of the GC reaction. Less than 2% of IgM(+) B cells showed Cre-mediated recombination. These cells carried few Ig somatic mutations, expressed germ-line Cgamma1- and activation-induced cytidine deaminase-specific transcripts and likely include GC B cell founders and/or plasma cell precursors. Cre-mediated recombination involved most IgG1, but also a fraction of IgG3-, IgG2a-, IgG2b-, and IgA-expressing GC and post-GC B cells. This result indicates that a GC B cell can transcribe more than one downstream C(H) gene before undergoing class switch recombination. The efficient induction of Cre expression in GC B cells makes the Cgamma1-cre allele a powerful tool for the genetic analysis of these cells, as well as, in combination with a suitable marker for Cre-mediated recombination, the tracking of class-switched memory B and plasma cells in vivo. To expedite the genetic analysis of GC B cells, we have established Cgamma1-cre F(1) embryonic stem cells, allowing further rounds of gene targeting and the cloning of compound mutants by tetraploid embryo complementation.


Subject(s)
B-Lymphocytes/metabolism , Germ Cells/metabolism , Germinal Center/metabolism , Immunoglobulin gamma-Chains/genetics , Immunoglobulin gamma-Chains/metabolism , Alleles , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Blastocyst/metabolism , Cells, Cultured , DNA, Recombinant/genetics , Gene Rearrangement, B-Lymphocyte , Germ Cells/immunology , Germinal Center/immunology , Immunoglobulin gamma-Chains/immunology , Interleukin-4/pharmacology , Mice , Mice, Transgenic , Mutation/genetics , Ploidies , Time Factors , Transcription, Genetic/genetics
17.
Immunology ; 112(2): 265-73, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15147570

ABSTRACT

Class switch recombination (CSR) is a T-cell-dependent mechanism regulating isotype switching in activated mature B cells. Recently we showed that T-cell-independent CSRs occur spontaneously during B lymphopoiesis, but such cells are negatively selected by Fas signalling. In immunoglobulin mu-deficient mice, lack of Fas rescues isotype-switched B cells, resulting in generation of an autoimmune primary immunoglobulin G (IgG) repertoire in muMT/lpr mice. In the present study, we studied the role of alphabeta and gammadelta T cells in regulating this primary gammaH-driven repertoire. We found that a lack of alphabeta T cells significantly inhibited IgG production and autoimmunity in muMT/lpr mice, whereas a lack of gammadelta T cells resulted in augmented IgG production and autoimmunity. Also, a lack of T cells in muMT mice rescued isotype-switched B cells and serum IgG, probably owing to the lack of available FasL. We suggest that although CSRs in B-cell lymphopoiesis are T-cell independent, alphabeta T cells are important in the expansion of isotype-switched B-cell precursors and in promoting gammaH-driven autoimmunity, whereas gammadelta T cells regulate these cells.


Subject(s)
Autoimmunity , Immunoglobulin G/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/immunology , Animals , B-Lymphocytes/immunology , Cell Differentiation/immunology , Chromatin/immunology , Fas Ligand Protein , Immunoglobulin Class Switching/immunology , Immunoglobulin G/blood , Immunoglobulin mu-Chains/immunology , Lymphocyte Cooperation/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , fas Receptor/immunology
18.
Int Immunol ; 16(7): 905-13, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15148286

ABSTRACT

Class switch recombination (CSR) is a well-regulated process that occurs in peripheral lymphoid tissue, and is thought of as an important factor constructing the memory repertoire. We have recently shown that CSR normally occurs during bone marrow (BM) development, and these isotype-switched B cells are negatively selected by Fas signaling. This novel pathway of B cell development may generate a primary repertoire driven by gamma-heavy receptors, the nature of which is yet unknown. To study this gammaH-driven repertoire we used mice lacking IgM-transmembrane tail exon ( micro MT), where B cell development is limited by their ability to undergo CSR. We already showed that lack of Fas signaling rescues development of a significant population of isotype-switched B cells and production of high titers of non-IgM serum antibodies in micro MT mice deficient in Fas ( micro MT/lpr), thereby providing a mouse model allowing the assessment of gammaH-driven repertoire. Using a tissue array and phage display epitope library we report here that IgG repertoire in micro MT/lpr mice is oligo-monoclonal, bearing self-tissue reactivity. This is supported by analysis of the Vkappa utilization in peripheral B cells from micro MT/lpr mice, which revealed a strikingly restricted repertoire. In contrast, micro MT/lpr B cells that are grown in non-selective BM cultures utilize a wide repertoire. These results suggest that the Fas pathway is an important regulator in the generation and selection of an autoimmune gammaH-driven repertoire in vivo.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin gamma-Chains/immunology , Immunoglobulin mu-Chains/immunology , Lymphopoiesis/immunology , Recombination, Genetic/immunology , fas Receptor/immunology , Animals , Autoantigens/immunology , Immunoglobulin gamma-Chains/genetics , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Immunoglobulin mu-Chains/genetics , Lymphopoiesis/genetics , Mice , Mice, Knockout , Signal Transduction/immunology , fas Receptor/genetics
19.
Int Immunol ; 15(9): 1045-52, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12917256

ABSTRACT

The expression of muH chain is an important checkpoint in B cell development. In mice deficient for IgM transmembrane tail exons (muMT mice) B cell development is blocked at the pro-B stage. However, we showed that Fas-deficient muMT mice (muMT/lpr) develop a very small population of isotype-switched B cells and produce high titers of self-reactive serum antibodies. In addition, muMT/lpr mice develop severe lymphoproliferation and both pathologic processes occur at young ages. This may suggest that lack of Fas-Fas ligand signaling exacerbates murine lupus in B cell lymphopenic mice. To test this we analyzed antibody and plasma cell formation, and accumulation of abnormal T cells in muMT/lpr mice. Our results show that the muMT/lpr mouse is particularly permissive for the development and accumulation of antibody-producing cells, thereby explaining the high titers of serum antibodies in these mice. In addition, we found that accumulating cells in spleen and lymph nodes of muMT/lpr mice are alphabeta T cells expressing the abnormal B220+/CD3+ surface markers, a phenotype also described for other Fas-deficient mouse models. Strikingly, we found that accumulating cells in muMT/lpr mice express the membrane proteoglycan syndecan-1, a known plasma cell marker. Development of these cells is blocked in mice deficient for TCRbeta and TCRdelta. We also found that both antibody production and lymphoproliferation in muMT/lpr mice are Th1 regulated. Our results, therefore, suggest that in the muMT/lpr mouse model a small population of isotype-switched B cells is sufficient for the initiation and propagation of Th1-regulated murine lupus.


Subject(s)
Immunoglobulin mu-Chains/genetics , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/analysis , Plasma Cells/immunology , Proteoglycans/analysis , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , CD3 Complex/physiology , Cell Differentiation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoglobulin mu-Chains/biosynthesis , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Mutant Strains , Mice, Transgenic , Receptors, Antigen, B-Cell/genetics , Spleen/immunology , Syndecan-1 , Syndecans , Th1 Cells/immunology , fas Receptor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL