151
|
Affiliation(s)
- Michael L Dustin
- Skirball Institute for Molecular Medicine, New York University School of Medicine, New York, NY 10016, USA.
| | | |
Collapse
|
152
|
Alkhatib A, Werner M, Hug E, Herzog S, Eschbach C, Faraidun H, Köhler F, Wossning T, Jumaa H. FoxO1 induces Ikaros splicing to promote immunoglobulin gene recombination. ACTA ACUST UNITED AC 2012; 209:395-406. [PMID: 22291095 PMCID: PMC3280865 DOI: 10.1084/jem.20110216] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
During murine B cell development, PI3 kinase inhibits Ig gene rearrangement by suppressing FoxO1, which mediates Ikaros mRNA splicing; Ikaros is needed for Ig gene recombination. Somatic rearrangement of immunoglobulin (Ig) genes is a key step during B cell development. Using pro–B cells lacking the phosphatase Pten (phosphatase and tensin homolog), which negatively regulates phosphoinositide-3-kinase (PI3K) signaling, we show that PI3K signaling inhibits Ig gene rearrangement by suppressing the expression of the transcription factor Ikaros. Further analysis revealed that the transcription factor FoxO1 is crucial for Ikaros expression and that PI3K-mediated down-regulation of FoxO1 suppresses Ikaros expression. Interestingly, FoxO1 did not influence Ikaros transcription; instead, FoxO1 is essential for proper Ikaros mRNA splicing, as FoxO1-deficient cells contain aberrantly processed Ikaros transcripts. Moreover, FoxO1-induced Ikaros expression was sufficient only for proximal VH to DJH gene rearrangement. Simultaneous expression of the transcription factor Pax5 was needed for the activation of distal VH genes; however, Pax5 did not induce any Ig gene rearrangement in the absence of Ikaros. Together, our results suggest that ordered Ig gene rearrangement is regulated by distinct activities of Ikaros, which mediates proximal VH to DJH gene rearrangement downstream of FoxO1 and cooperates with Pax5 to activate the rearrangement of distal VH genes.
Collapse
Affiliation(s)
- Alabbas Alkhatib
- Department of Molecular Immunology, Faculty of Biology, Albert-Ludwigs University of Freiburg and Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Abstract
In the last decade, the availability of genetically modified animals has revealed interesting roles for phosphoinositide 3-kinases (PI3Ks) as signaling platforms orchestrating multiple cellular responses, both in health and pathology. By acting downstream distinct receptor types, PI3Ks nucleate complex signaling assemblies controlling several biological process, ranging from cell proliferation and survival to immunity, cancer, metabolism and cardiovascular control. While the involvement of these kinases in modulating immune reactions and neoplastic transformation has long been accepted, recent progress from our group and others has highlighted new and unforeseen roles of PI3Ks in controlling cardiovascular function. Hence, the view is emerging that pharmacological targeting of distinct PI3K isoforms could be successful in treating disorders such as myocardial infarction and heart failure, besides inflammatory diseases and cancer. Currently, PI3Ks represent attractive drug targets for companies interested in the development of novel and safe treatments for such diseases. Numerous hit and lead compounds are now becoming available and, for some of them, clinical trials can be envisaged in the near future. In the following sections, we will outline the impact of specific PI3K isoforms in regulating different cellular contexts, including immunity, metabolism, cancer and cardiovascular system, both in physiological and disease conditions.
Collapse
|
154
|
Abstract
Phosphoinositide 3-kinases (PI3Ks) control cell growth, proliferation, cell survival, metabolic activity, vesicular trafficking, degranulation, and migration. Through these processes, PI3Ks modulate vital physiology. When over-activated in disease, PI3K promotes tumor growth, angiogenesis, metastasis or excessive immune cell activation in inflammation, allergy and autoimmunity. This chapter will introduce molecular activation and signaling of PI3Ks, and connections to target of rapamycin (TOR) and PI3K-related protein kinases (PIKKs). The focus will be on class I PI3Ks, and extend into current developments to exploit mechanistic knowledge for therapy.
Collapse
Affiliation(s)
- Matthias Wymann
- Institute Biochemistry & Genetics, Department Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland,
| |
Collapse
|
155
|
Abstract
UNLABELLED Several phosphoinositide 3-kinase (PI3K) inhibitors are in the clinic and many more are in preclinical development. CAL-101, a selective inhibitor of the PI3Kδ isoform, has shown remarkable success in certain hematologic malignancies. Although PI3Kδ signaling plays a central role in lymphocyte biology, the degree of single-agent therapeutic activity of CAL-101 during early-phase development has been somewhat unexpected. CAL-101 works in part by blocking signals from the microenvironment that normally sustain leukemia and lymphoma cells in a protective niche. As PI3Ks enter the arena of molecular-targeted therapies, CAL-101 provides proof of principle that isoform-selective compounds can be effective in selected cancer types and patient populations. SIGNIFICANCE A key question is whether compounds targeting a single PI3K catalytic isoform can provide meaningful single agent efficacy in cancer cells that express multiple isoforms. Clinical studies of the drug CAL-101 have provided a significant advance by showing that selective targeting of PI3Kδ achieves efficacy in chronic lymphocytic leukemia, in part through targeting the tumor microenvironment.
Collapse
Affiliation(s)
- David A Fruman
- Department of Molecular Biology and Biochemistry and Institute for Immunology, University of California, Irvine, Irvine, California 92697-3900, USA.
| | | |
Collapse
|
156
|
Green JA, Suzuki K, Cho B, Willison LD, Palmer D, Allen CDC, Schmidt TH, Xu Y, Proia RL, Coughlin SR, Cyster JG. The sphingosine 1-phosphate receptor S1P₂ maintains the homeostasis of germinal center B cells and promotes niche confinement. Nat Immunol 2011; 12:672-80. [PMID: 21642988 PMCID: PMC3158008 DOI: 10.1038/ni.2047] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 05/02/2011] [Indexed: 01/14/2023]
Abstract
Mice deficient in sphingosine 1-phosphate receptor type 2 (S1P(2)) develop diffuse large B cell lymphoma. However, the role of S1P(2) in normal germinal center (GC) physiology is unknown. Here we show that S1P(2)-deficient GC B cells outgrew their wild-type counterparts in chronically established GCs. We found that antagonism of the kinase Akt mediated by S1P(2) and its downstream mediators Gα(12), Gα(13) and p115RhoGEF regulated cell viability and was required for growth control in chronically proliferating GCs. Moreover, S1P(2) inhibited GC B cell responses to follicular chemoattractants and helped confine cells to the GC. In addition, S1P(2) overexpression promoted the centering of activated B cells in the follicle. We suggest that by inhibiting Akt activation and migration, S1P(2) helps restrict GC B cell survival and localization to an S1P-low niche at the follicle center.
Collapse
Affiliation(s)
- Jesse A Green
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Baracho G, Miletic A, Omori S, Cato M, Rickert R. Emergence of the PI3-kinase pathway as a central modulator of normal and aberrant B cell differentiation. Curr Opin Immunol 2011; 23:178-83. [PMID: 21277760 PMCID: PMC3070849 DOI: 10.1016/j.coi.2011.01.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 12/22/2010] [Accepted: 01/02/2011] [Indexed: 01/16/2023]
Abstract
Phosphoinositide 3-kinase (PI3K) defines a family of lipid kinases that direct a wide range of cellular processes and cell fate decisions. Since its discovery, and that of its enzymatic antagonist PTEN, much of the focus on PI3K has been on its oncogenic potential. In recent years, studies on PI3K signaling in B lymphocytes have established the importance of this pathway in effecting B cell differentiation and associated molecular events such as V(D)J recombination and class switch recombination. Intriguing new findings also indicate that there is specificity in the PI3K pathway in B cells, including preferential expression or usage of particular PI3K isoforms and counter-regulation by the PTEN and SHIP phosphatases. The role of PI3K adaptor proteins (CD19, BCAP, and TC21) has also undergone revision to reflect both shared and unique properties. The emergence of Foxo1 as a critical PI3K regulatory target for B cell differentiation has united membrane proximal regulatory events orchestrated by PI3K/PTEN/SHIP with key transcriptional targets. Insights into the regulation and impact of PI3K signaling have been brought to bear in new treatments for B cell malignancies, and will also be an important topic of consideration for B cell-dependent autoimmune diseases.
Collapse
Affiliation(s)
- G.V. Baracho
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - A.V. Miletic
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - S.A. Omori
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - M.H. Cato
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | - R.C. Rickert
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
158
|
14-3-3sigma regulates B-cell homeostasis through stabilization of FOXO1. Proc Natl Acad Sci U S A 2011; 108:1555-60. [PMID: 21205887 DOI: 10.1073/pnas.1017729108] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
14-3-3σ regulates cytokinesis and cell cycle arrest induced by DNA damage but its role in the immune system is unknown. Using gene-targeted 14-3-3σ-deficient (i.e., KO) mice, we studied the role of 14-3-3σ in B-cell functions. Total numbers of B cells were reduced by spontaneous apoptosis of peripheral B cells. Upon B-cell antigen receptor engagement in vitro, KO B cells did not proliferate properly or up-regulate CD86. In response to T cell-independent antigens, KO B cells showed poor secretion of antigen-specific IgM. This deficit led to increased lethality of KO mice after vesicular stomatitis virus infection. KO B cells showed elevated total FOXO transcriptional activity but also increased FOXO1 degradation. Coimmunoprecipitation revealed that endogenous 14-3-3σ protein formed a complex with FOXO1 protein. Our results suggest that 14-3-3σ maintains FOXO1 at a consistent level critical for normal B-cell antigen receptor signaling and B-cell survival.
Collapse
|
159
|
Cato MH, Chintalapati SK, Yau IW, Omori SA, Rickert RC. Cyclin D3 is selectively required for proliferative expansion of germinal center B cells. Mol Cell Biol 2011; 31:127-37. [PMID: 20956554 PMCID: PMC3019862 DOI: 10.1128/mcb.00650-10] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 07/27/2010] [Accepted: 10/11/2010] [Indexed: 01/09/2023] Open
Abstract
The generation of robust T-cell-dependent humoral immune responses requires the formation and expansion of germinal center structures within the follicular regions of the secondary lymphoid tissues. B-cell proliferation in the germinal center drives ongoing antigen-dependent selection and the generation of high-affinity class-switched plasma and memory B cells. However, the mechanisms regulating B-cell proliferation within this microenvironment are largely unknown. Here, we report that cyclin D3 is uniquely required for germinal center progression. Ccnd3(-/-) mice exhibit a B-cell-intrinsic defect in germinal center maturation and fail to generate an affinity-matured IgG response. We determined that the defect resulted from failed proliferative expansion of GL7(+) IgD(-) PNA(+) B cells. Mechanistically, sustained expression of cyclin D3 was found to be regulated at the level of protein stability and controlled by glycogen synthase kinase 3 in a cyclic AMP-protein kinase A-dependent manner. The specific defect in proliferative expansion of GL7(+) IgD(-) PNA(+) B cells in Ccnd3(-/-) mice defines an underappreciated step in germinal center progression and solidifies a role for cyclin D3 in the immune response, and as a potential therapeutic target for germinal center-derived B-cell malignancies.
Collapse
Affiliation(s)
- Matthew H. Cato
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, and Program of Signal Transduction, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Suresh K. Chintalapati
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, and Program of Signal Transduction, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Irene W. Yau
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, and Program of Signal Transduction, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Sidne A. Omori
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, and Program of Signal Transduction, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Robert C. Rickert
- Program on Inflammatory Disease Research, Infectious and Inflammatory Disease Center, and Program of Signal Transduction, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
160
|
Abstract
A byproduct of the largely stochastic generation of a diverse B-cell specificity repertoire is production of cells that recognize autoantigens. Indeed, recent studies indicate that more than half of the primary repertoire consists of autoreactive B cells that must be silenced to prevent autoimmunity. While this silencing can occur by multiple mechanisms, it appears that most autoreactive B cells are silenced by anergy, wherein they populate peripheral lymphoid organs and continue to express unoccupied antigen receptors yet are unresponsive to antigen stimulation. Here we review molecular mechanisms that appear operative in maintaining the antigen unresponsiveness of anergic B cells. In addition, we present new data indicating that the failure of anergic B cells to mobilize calcium in response to antigen stimulation is not mediated by inactivation of stromal interacting molecule 1, a critical intermediary in intracellular store depletion-induced calcium influx.
Collapse
Affiliation(s)
- Yuval Yarkoni
- Integrated Department of Immunology, University of Colorado School of Medicine and National Jewish Health, Denver, CO, USA
| | | | | |
Collapse
|
161
|
Abstract
Engagement of the B-cell antigen receptor (BCR) or its precursor, the pre-BCR, induces a cascade of biochemical reactions that regulate the differentiation, selection, survival, and activation of B cells. This cascade is initiated by receptor-associated tyrosine kinases that activate multiple downstream signaling pathways. Since it is required for metabolism, cell growth, development, and survival, the activation of phosphoinositide 3-kinase (PI3K)-dependent pathways represents a crucial event of BCR/pre-BCR signaling. The phosphorylated substrates of the PI3K promote specific recruitment of selected signaling proteins to the plasma membrane, where important signaling complexes are formed to mediate the above-mentioned biological processes. Here, we review the principles of PI3K signaling and highlight the role of an important PI3K-driven module in VDJ recombination of immunoglobulin (Ig) genes during early B-cell development as compared with class switch recombination of Ig genes in mature B cells after activation by specific antigens. Furthermore, we discuss the role of PI3K in the survival of mature B cells, which is strictly dependent on BCR expression and basal BCR signaling.
Collapse
Affiliation(s)
- Markus Werner
- Faculty of Biology, Department of Molecular Immunology, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | | | | |
Collapse
|
162
|
Waterman PM, Cambier JC. The conundrum of inhibitory signaling by ITAM-containing immunoreceptors: potential molecular mechanisms. FEBS Lett 2010; 584:4878-82. [PMID: 20875413 PMCID: PMC2998577 DOI: 10.1016/j.febslet.2010.09.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 09/14/2010] [Accepted: 09/17/2010] [Indexed: 11/28/2022]
Abstract
Immunoreceptor signals must be appropriately transduced and regulated to achieve effective immunity while controlling inflammation and autoimmunity. It is generally held that these processes are mediated by the interplay of distinct activating and inhibitory receptors via conserved activating (ITAM) and inhibitory (ITIM) signaling motifs. However, recent evidence indicates that under certain conditions incomplete phosphorylation of ITAM tyrosines leads to inhibitory signaling. This new regulatory function of ITAMs has been termed ITAMi (inhibitory ITAM). Here we discuss the potential molecular mechanisms of inhibitory signaling by ITAM-containing receptors.
Collapse
Affiliation(s)
- Paul M Waterman
- Integrated Department of Immunology, University of Colorado School of Medicine, Denver CO 80206, USA
| | | |
Collapse
|
163
|
Bach2 represses plasma cell gene regulatory network in B cells to promote antibody class switch. EMBO J 2010; 29:4048-61. [PMID: 20953163 DOI: 10.1038/emboj.2010.257] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 09/21/2010] [Indexed: 01/03/2023] Open
Abstract
Two transcription factors, Pax5 and Blimp-1, form a gene regulatory network (GRN) with a double-negative loop, which defines either B-cell (Pax5 high) or plasma cell (Blimp-1 high) status as a binary switch. However, it is unclear how this B-cell GRN registers class switch DNA recombination (CSR), an event that takes place before the terminal differentiation to plasma cells. In the absence of Bach2 encoding a transcription factor required for CSR, mouse splenic B cells more frequently and rapidly expressed Blimp-1 and differentiated to IgM plasma cells as compared with wild-type cells. Genetic loss of Blimp-1 in Bach2(-/-) B cells was sufficient to restore CSR. These data with mathematical modelling of the GRN indicate that Bach2 achieves a time delay in Blimp-1 induction, which inhibits plasma cell differentiation and promotes CSR (Delay-Driven Diversity model for CSR). Reduction in mature B-cell numbers in Bach2(-/-) mice was not rescued by Blimp-1 ablation, indicating that Bach2 regulates B-cell differentiation and function through Blimp-1-dependent and -independent GRNs.
Collapse
|
164
|
Rolf J, Bell SE, Kovesdi D, Janas ML, Soond DR, Webb LMC, Santinelli S, Saunders T, Hebeis B, Killeen N, Okkenhaug K, Turner M. Phosphoinositide 3-kinase activity in T cells regulates the magnitude of the germinal center reaction. THE JOURNAL OF IMMUNOLOGY 2010; 185:4042-52. [PMID: 20826752 DOI: 10.4049/jimmunol.1001730] [Citation(s) in RCA: 182] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The generation of high-affinity Abs is essential for immunity and requires collaboration between B and T cells within germinal centers (GCs). By using novel mouse models with a conditional deletion of the p110δ catalytic subunit of the PI3K pathway, we established that p110δ is required in T cells, but not in B cells, for the GC reaction. We found the formation of T follicular helper (T(FH)) cells to be critically dependent on p110δ in T cells. Furthermore, by deleting phosphatase and tensin homolog deleted on chromosome 10, which opposes p110δ in activated T cells, we found a positive correlation between increased numbers of T(FH) cells and GC B cells. These results are consistent with the hypothesis that T cell help is the limiting factor in the GC reaction. P110δ was not required for the expression of B cell lymphoma 6, the downregulation of CCR7, or T cell entry into primary follicles. Instead, p110δ was the critical catalytic subunit for ICOS downstream signaling and the production of key T(FH) cytokines and effector molecules. Our findings support a model in which the magnitude of the GC reaction is controlled by the activity of the PI3K pathway in T(FH) cells.
Collapse
Affiliation(s)
- Julia Rolf
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Taher TE, Parikh K, Flores-Borja F, Mletzko S, Isenberg DA, Peppelenbosch MP, Mageed RA. Protein phosphorylation and kinome profiling reveal altered regulation of multiple signaling pathways in B lymphocytes from patients with systemic lupus erythematosus. ACTA ACUST UNITED AC 2010; 62:2412-23. [PMID: 20506108 DOI: 10.1002/art.27505] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The cause of B lymphocyte hyperactivity and autoantibody production in systemic lupus erythematosus (SLE) remains unclear. Previously, we identified abnormalities in the level and translocation of signaling molecules in B cells in SLE patients. The present study was undertaken to examine the extent of signaling abnormalities that relate to altered B cell responses in SLE. METHODS B lymphocytes from 88 SLE patients and 72 healthy controls were isolated from blood by negative selection. Protein tyrosine phosphorylation and cellular kinase levels were analyzed by Western blotting, flow cytometry, and a kinome array protocol. Changes in protein phosphorylation were determined in ex vivo B cells and following B cell receptor engagement. RESULTS Differences in tyrosine phosphorylation in B cells from patients with SLE, compared with matched controls, were demonstrated. Further, the kinome array analysis identified changes in the activation of key kinases, i.e., the activity of phosphatidylinositol 3-kinase, which regulates survival and differentiation, was up-regulated and the activity of Rac and Rho kinases, which regulate the cytoskeleton and migration, was increased. In contrast, the activity of ATR, which regulates the cell cycle, was down-regulated in SLE patients compared with controls. Differences in signaling pathways were seen in all SLE B lymphocyte subsets that manifested phenotypic features of immature, mature, and memory cells. CONCLUSION This study revealed dysregulation in multiple signaling pathways that control key responses in B cells of SLE patients. Data generated in this study provide a molecular basis for further analysis of the altered B lymphocyte responses in SLE.
Collapse
Affiliation(s)
- Taher E Taher
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | | | | | | | |
Collapse
|
166
|
Abstract
In response to an assault by foreign organisms, peripheral B cells can change their antibody affinity and isotype by somatically mutating their genomic DNA. The ability of a cell to modify its DNA is exceptional in light of the potential consequences of genetic alterations to cause human disease and cancer. Thus, as expected, this mechanism of antibody diversity is tightly regulated and coordinated through one protein, activation-induced deaminase (AID). AID produces diversity by converting cytosine to uracil within the immunoglobulin loci. The deoxyuracil residue is mutagenic when paired with deoxyguanosine, since it mimics thymidine during DNA replication. Additionally, B cells can manipulate the DNA repair pathways so that deoxyuracils are not faithfully repaired. Therefore, an intricate balance exists which is regulated at multiple stages to promote mutation of immunoglobulin genes, while retaining integrity of the rest of the genome. Here we discuss and summarize the current understanding of how AID functions to cause somatic hypermutation.
Collapse
Affiliation(s)
- Robert W Maul
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | | |
Collapse
|
167
|
Oracki SA, Walker JA, Hibbs ML, Corcoran LM, Tarlinton DM. Plasma cell development and survival. Immunol Rev 2010; 237:140-59. [DOI: 10.1111/j.1600-065x.2010.00940.x] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
168
|
North CM, Crawford RB, Lu H, Kaminski NE. 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated suppression of toll-like receptor stimulated B-lymphocyte activation and initiation of plasmacytic differentiation. Toxicol Sci 2010; 116:99-112. [PMID: 20348231 PMCID: PMC2886857 DOI: 10.1093/toxsci/kfq095] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 03/23/2010] [Indexed: 01/20/2023] Open
Abstract
2,3,7,8-Tetrachlordibenzo-p-dioxin (TCDD) is a potent suppressor of humoral immunity, disrupting antibody production in response to both T cell-dependent and T cell-independent antigens. Among the cell types required for humoral responses, the B cell is highly, and directly, sensitive to TCDD. B cells become antibody-secreting cells via plasmacytic differentiation, a process regulated by several transcription factors, including activator protein-1, B-cell CLL/lymphoma 6 (BCL-6), and B lymphocyte-induced maturation protein 1 (Blimp-1). The overarching conceptual framework guiding experimentation is that TCDD disrupts plasmacytic differentiation by altering the expression or activity for upstream regulators of Blimp-1. Multiparametric flow cytometry was used to investigate TCDD-induced alterations in both activation marker and transcription factor expression following lipopolysaccharide (LPS) activation of purified B cells. TCDD significantly impaired LPS-activated expression of major histocompatibility complex class II, cluster of differentiation (CD)69, CD80, and CD86. Immunosuppressive concentrations of TCDD also suppressed LPS-activated Blimp-1 and phosphorylated c-Jun expression, whereas elevating BCL-6 expression. Because BCL-6 and c-Jun are directly and indirectly regulated by the kinases AKT, extracellular signal-regulated kinase (ERK), and Jun N-terminal kinase (JNK), it was hypothesized that TCDD alters toll-like receptor-activated kinase phosphorylation. TCDD at 0.03 and 0.3 nM significantly impaired phosphorylation of AKT, ERK, and JNK in CH12.LX B cells activated with LPS, CpG oligonucleotides, or resiquimod (R848). In primary B cells, R848-activated phosphorylation of AKT, ERK, and JNK was also impaired by TCDD at 30 nM. These results suggest that impairment of plasmacytic differentiation by TCDD involves altered transcription factor expression, in part, by suppressed kinase phosphorylation.
Collapse
Affiliation(s)
| | | | | | - Norbert E. Kaminski
- Center for Integrative Toxicology, Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
169
|
Ghigo A, Damilano F, Braccini L, Hirsch E. PI3K inhibition in inflammation: Toward tailored therapies for specific diseases. Bioessays 2010; 32:185-196. [PMID: 20162662 DOI: 10.1002/bies.200900150] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the past decade, the availability of genetically modified animals has enabled the discovery of interesting roles for phosphatidylinositol 3-kinase-gamma (PI3Kgamma) and -delta (PI3Kdelta) in different cell types orchestrating innate and adaptive immune responses. Therefore, these PI3K isoforms appear to be attractive drug targets for the treatment of diseases caused by unrestrained immune reactions. Currently, pharmacological targeting of PI3Kgamma and/or PI3Kdelta represents one of the most promising challenges for companies interested in the development of novel safe treatments for inflammatory diseases. In this review we provide a general outline of PI3Kgamma- and PI3Kdelta-specific functions in distinct subsets of inflammatory cells. We also discuss the therapeutic impact of novel compounds targeting PI3Kgamma, PI3Kdelta or both, in mouse models of autoimmune disorders (systemic lupus erythematosus (SLE) and rheumatoid arthritis), respiratory diseases (allergic asthma and chronic obstructive pulmonary disease) and cardiovascular dysfunctions (atherosclerosis and myocardial infarction).
Collapse
Affiliation(s)
- Alessandra Ghigo
- Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Federico Damilano
- Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Laura Braccini
- Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Emilio Hirsch
- Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy
| |
Collapse
|
170
|
Kracker S, Gardes P, Mazerolles F, Durandy A. Immunoglobulin class switch recombination deficiencies. Clin Immunol 2010; 135:193-203. [DOI: 10.1016/j.clim.2010.01.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 01/25/2010] [Accepted: 01/25/2010] [Indexed: 01/01/2023]
|
171
|
Kövesdi D, Bell SE, Turner M. The development of mature B lymphocytes requires the combined function of CD19 and the p110δ subunit of PI3K. SELF NONSELF 2010; 1:144-153. [PMID: 21487516 DOI: 10.4161/self.1.2.11796] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 02/25/2010] [Accepted: 03/11/2010] [Indexed: 11/19/2022]
Abstract
Mice lacking either CD19 or p110δ have reduced numbers of marginal zone and B1 B cells but normal numbers of naïve B2 cells which occupy the follicles of the lymphoid organs. We show here that mice lacking both CD19 and p110δ have normal B cell development in the bone marrow but have a significant reduction in the number of naïve B2 cells in the bone marrow, spleen and lymph nodes. These p110δ/CD19 double mutant B cells show a survival defect and reduced responsiveness to the pro-survival cytokine BAFF despite normal NFκB2/p100 processing and elevated expression of Bcl-2. Although the combined loss of p110δ and CD19 did not increase switching to Ig-lambda in immature B cells, mature B lymphocytes from the lymph nodes of p110δ/CD19 double mutant mice express highly elevated levels of mRNA encoding RAG-1 and RAG-2, which confirms the existing synergy between CD19 and p110δ-mediated signaling.
Collapse
Affiliation(s)
- Dorottya Kövesdi
- Laboratory of Lymphocyte Signalling and Development; The Babraham Institute; Babraham, Cambridge UK
| | | | | |
Collapse
|
172
|
Okkenhaug K, Fruman DA. PI3Ks in lymphocyte signaling and development. Curr Top Microbiol Immunol 2010; 346:57-85. [PMID: 20563708 DOI: 10.1007/82_2010_45] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lymphocyte development and function are regulated by tyrosine kinase and G-protein coupled receptors. Each of these classes of receptors activates phosphoinositide 3-kinase (PI3K). In this chapter, we summarize current understanding of how PI3K contributes to key aspects of the adaptive immune system.
Collapse
Affiliation(s)
- Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Cambridge, UK.
| | | |
Collapse
|
173
|
Srinivasan L, Sasaki Y, Calado DP, Zhang B, Paik JH, DePinho RA, Kutok JL, Kearney JF, Otipoby KL, Rajewsky K. PI3 kinase signals BCR-dependent mature B cell survival. Cell 2009; 139:573-86. [PMID: 19879843 DOI: 10.1016/j.cell.2009.08.041] [Citation(s) in RCA: 527] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 07/17/2009] [Accepted: 08/26/2009] [Indexed: 01/01/2023]
Abstract
Previous work has shown that mature B cells depend upon survival signals delivered to the cells by their antigen receptor (BCR). To identify the molecular nature of this survival signal, we have developed a genetic approach in which ablation of the BCR is combined with the activation of specific, BCR dependent signaling cascades in mature B cells in vivo. Using this system, we provide evidence that the survival of BCR deficient mature B cells can be rescued by a single signaling pathway downstream of the BCR, namely PI3K signaling, with the FOXO1 transcription factor playing a central role.
Collapse
Affiliation(s)
- Lakshmi Srinivasan
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Zhang TT, Al-Alwan M, Marshall AJ. The pleckstrin homology domain adaptor protein Bam32/DAPP1 is required for germinal center progression. THE JOURNAL OF IMMUNOLOGY 2009; 184:164-72. [PMID: 19949096 DOI: 10.4049/jimmunol.0902505] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ab affinity maturation within germinal centers (GCs) requires weeks to complete. Several signaling pathways in B cells have been shown to be required for initiation of the GC response; however, the signaling checkpoints controlling progression and eventual dissolution of the GC reaction are poorly understood. The adaptor protein Bam32/DAPP1 was originally isolated from human GCs and functions downstream of phosphoinositide 3-kinase enzymes, which are known to have critical roles in B cell activation and GC responses. In this study we identify a unique role of Bam32/DAPP1 in promoting GC progression. Bam32-deficient mice show normal GC initiation, but premature GC dissolution after immunization with protein Ag in alum or low doses of sheep red blood cells. Adoptive transfer studies confirmed that Bam32-deficient B cells have an intrinsic impairment in the ability to mount sustained GC responses. Bam32 deficiency was also associated with impaired Ab affinity maturation. Proliferation of Bam32-deficient GC B cells was not compromised; however, these cells show impaired switch to IgG1 and increased apoptosis in situ. GCs formed by Bam32-deficient B cells contain fewer T cells, indicating that Bam32 is required for B cell-dependent T cell accumulation within established GCs. Exogenous CD40 ligand restored GC B cell numbers and switch to IgG1, indicating that Bam32-deficient B cells are competent to respond to CD40 stimulation when ligand is available. These data demonstrate that Bam32 is not required for GC initiation, but rather functions in a late checkpoint of GC progression associated with T cell recruitment and GC B cell survival.
Collapse
Affiliation(s)
- Ting-ting Zhang
- Department of Immunology, University of Manitoba, Manitoba, Canada
| | | | | |
Collapse
|
175
|
Browne CD, Del Nagro CJ, Cato MH, Dengler HS, Rickert RC. Suppression of phosphatidylinositol 3,4,5-trisphosphate production is a key determinant of B cell anergy. Immunity 2009; 31:749-60. [PMID: 19896393 DOI: 10.1016/j.immuni.2009.08.026] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 06/15/2009] [Accepted: 08/21/2009] [Indexed: 01/01/2023]
Abstract
Anergy is a critical physiologic mechanism to sensor self-reactive B cells. However, a biochemical understanding of how anergy is achieved and maintained is lacking. Herein, we investigated the role of the phosphoinositide 3-kinase (PI3K) lipid product PI(3,4,5)P(3) in B cell anergy. We found reduced generation of PI(3,4,5)P(3) in anergic B cells, which was attributable to reduced phosphorylation of the PI3K membrane adaptor CD19, as well as increased expression of the inositol phosphatase PTEN. Sustained production of PI(3,4,5)P(3) in B cells, achieved through conditional deletion of Pten, resulted in failed tolerance induction and abundant autoantibody production. In contrast to wild-type immature B cells, B cell receptor engagement of PTEN-deficient immature B cells resulted in activation and proliferation, indicating a central defect in early B cell responsiveness. These findings establish repression of the PI3K signaling pathway as a necessary condition to avert the generation, activation, and persistence of self-reactive B cells.
Collapse
Affiliation(s)
- Cecille D Browne
- Program of Inflammatory Disease Research, Infectious and Inflammatory Disease Center & Program of Signal Transduction, Cancer Center, Burnham Institute for Medical Research, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
176
|
Hinman RM, Nichols WA, Diaz TM, Gallardo TD, Castrillon DH, Satterthwaite AB. Foxo3-/- mice demonstrate reduced numbers of pre-B and recirculating B cells but normal splenic B cell sub-population distribution. Int Immunol 2009; 21:831-42. [PMID: 19502585 DOI: 10.1093/intimm/dxp049] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
B cell antigen receptor (BCR) cross-linking promotes proliferation and survival of mature B cells. Phosphoinositide-3-kinase-mediated down-regulation of pro-apoptotic and anti-mitogenic genes such as the Foxo family of transcription factors is an important component of this process. Previously, we demonstrated that BCR signaling decreases expression of transcripts for Foxo1, Foxo3 and Foxo4. We now show that BCR-induced down-regulation of Foxo3 and Foxo4 mRNA expression occurs via distinct mechanisms from those established for Foxo1. While Foxo1, Foxo3 and Foxo4 bind the same DNA sequence, the differential control of their expression upon B cell activation suggests that they may have unique functions in the B lineage. To begin to address this issue, we evaluated B cell development and function in Foxo3-/- mice. No effect of Foxo3 deficiency was observed with respect to the following parameters in the splenic B cell compartment: sub-population distribution, proliferation, in vitro differentiation and expression of the Foxo target genes cyclin G2 and B cell translocation gene 1. However, Foxo3-/- mice demonstrated increased basal levels of IgG2a, IgG3 and IgA. A significant reduction in pre-B cell numbers was also observed in Foxo3-/- bone marrow. Finally, recirculating B cells in the bone marrow and peripheral blood were decreased in Foxo3-/- mice, perhaps due to lower than normal expression of receptor for sphingosine-1 phosphate, which mediates egress from lymphoid organs. Thus, Foxo3 makes a unique contribution to B cell development, B cell localization and control of Ig levels.
Collapse
Affiliation(s)
- Rochelle M Hinman
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
177
|
Abstract
The phosphoinositide 3-kinase (PI3K) family of lipid kinases regulates diverse aspects of lymphocyte behavior. This review discusses how genetic and pharmacological tools have yielded an increasingly detailed understanding of how PI3K enzymes function at different stages of lymphocyte development and activation. Following antigen receptor engagement, activated PI3K generates 3-phosphorylated inositol lipid products that serve as membrane targeting signals for numerous proteins involved in the assembly of multiprotein complexes, termed signalosomes, and immune synapse formation. In B cells, class IA PI3K is the dominant subgroup whose loss causes profound defects in development and antigen responsiveness. In T cells, both class IA and IB PI3K contribute to development and immune function. PI3K also regulates both chemokine responsiveness and antigen-driven changes in lymphocyte trafficking. PI3K modulates the function not only of effector T cells, but also regulatory T cells; these disparate functions culminate in unexpected autoimmune phenotypes in mice with PI3K-deficient T cells. Thus, PI3K signaling is not a simple switch to promote cellular activation, but rather an intricate web of interactions that must be properly balanced to ensure appropriate cellular responses and maintain immune homeostasis. Defining these complexities remains a challenge for pharmaceutical development of PI3K inhibitors to combat inflammation and autoimmunity.
Collapse
Affiliation(s)
- David A Fruman
- Department of Molecular Biology and Biochemistry, and Center for Immunology, University of California at Irvine, Irvine, CA, USA.
| | | |
Collapse
|
178
|
Dil N, Marshall AJ. Role of phosphoinositide 3-kinase p110 delta in TLR4- and TLR9-mediated B cell cytokine production and differentiation. Mol Immunol 2009; 46:1970-8. [PMID: 19362372 DOI: 10.1016/j.molimm.2009.03.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 03/12/2009] [Accepted: 03/14/2009] [Indexed: 11/15/2022]
Abstract
Phosphoinositide 3-kinase (PI3K) enzymes play key roles in signaling via antigen receptors and cytokine receptors and isoform-selective PI3K inhibitors are being evaluated as targets for treatment of allergic and inflammatory diseases. The specific roles of PI3K isoforms in TLR-mediated activation of lymphocytes have not been defined. In this study we assess the role of p110 delta PI3K in TLR4, TLR9, or TLR4+TLR9-mediated B cell responses. Utilizing both p110 delta-mutant mice and p110 delta-specific inhibitor IC87114, we find that signaling via p110 delta is required for optimal B cell proliferation, but is not required for TLR-mediated B cell differentiation into plasma cells or Ig isotype switch. However PI3K blockade led to increased frequencies of IgG1 and IgE expressing cells, and partially reversed ability of CpG to inhibit IgG1 and IgE. Examination of B cell cytokine production revealed that p110 delta blockade markedly reduced IL-6 and IL-10 production. In contrast, p110 delta signaling was clearly not required for IL-12 production, with p110 delta-mutant B cells in fact showing enhanced IL-12 p70 production. TLR4- and TLR9-ligands act in synergy to drive IL-6 and IL-10 production, but not IL-12, and this additive effect is independent of p110 delta signaling. Together, these results indicate that PI3K delta functions in influencing the type of B cell cytokine production and differentiation response induced by TLR-ligands.
Collapse
Affiliation(s)
- Nyla Dil
- Department of Immunology, University of Manitoba, Winnipeg, MB, R3E 0W2, Canada.
| | | |
Collapse
|
179
|
Niu H, Sobel ES, Morel L. Defective B-cell response to T-dependent immunization in lupus-prone mice. Eur J Immunol 2009; 38:3028-40. [PMID: 18924209 DOI: 10.1002/eji.200838417] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Lupus anti-nuclear Ab show the characteristics of Ag-driven T-cell-dependent (TD) humoral responses. If autoAg elicit the same response as exogenous Ag, lupus should enhance humoral responses to immunization. Blunted responses to various immunizations have, however, been reported in a significant portion of lupus patients. In this study, we show that lupus-prone C57BL/6.Sle1.Sle2.Sle3 (B6.TC) mice produce significantly less Ab in response to TD immunization than congenic controls, while producing significantly more total Ig. This blunted Ab response to TD Ag could be reconstituted with B6.TC B and CD4+ T cells. Multiple defects were found in the B6.TC response to 4-hydroxy-3-nitrophenylacetyl-keyhole limpet hemocyanin (NP-KLH) compared with total Ig, including a smaller percentage of B cells participating in the NP-response, a reduced entry into germinal centers, and highly defective production of NP-specific long-lived plasma cells (PC) in the bone marrow. B6.TC PC expressed reduced levels of FcgammaRIIb, which suggests that reduced apoptosis in resident PC prevents the establishment of newly formed NP-specific PC in bone marrow niches. Overall, these results show that lupus-prone mice responded differently to auto- and exogenous Ag and suggest that low FcgammaRIIb, hypergammaglobulinemia, and high autoAb production would be predictive of a poor response to immunization in lupus patients.
Collapse
Affiliation(s)
- Haitao Niu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610-0275, USA
| | | | | |
Collapse
|
180
|
The role of PI3K signalling in the B cell response to antigen. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 633:43-53. [PMID: 19209680 DOI: 10.1007/978-0-387-79311-5_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The PI3K signalling pathway is crucial to normal B cell development and response to antigen. The p1108 catalytic subunit plays an important and non-redundant role within this pathway although other catalytic isoforms may also contribute. Although CD40, TLR and cytokines all activate PI3K the BCR seems especially dependent upon PI3K signalling. The downstream effects of PI3K may be mediated to a large extent by activation of PKB. In B cell development PI3K promotes development of MZ and Bl cells. In the response to antigen PI3K is crucial to BCR-mediated proliferation. PI3K activity has been shown to be inhibitory to CSR. The effects on immunoglobulin production and ASC differentiation are harder to disentangle from the developmental effects on cell populations and at present remain uncertain.
Collapse
|
181
|
Zhang TT, Okkenhaug K, Nashed BF, Puri KD, Knight ZA, Shokat KM, Vanhaesebroeck B, Marshall AJ. Genetic or pharmaceutical blockade of p110delta phosphoinositide 3-kinase enhances IgE production. J Allergy Clin Immunol 2008; 122:811-819.e2. [PMID: 19014771 DOI: 10.1016/j.jaci.2008.08.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 07/02/2008] [Accepted: 08/11/2008] [Indexed: 12/20/2022]
Abstract
BACKGROUND Recent studies indicate that pharmaceutical blockade of phosphoinositide 3-kinase (PI3K) signaling enzymes might be effective in reducing allergic airway inflammation. Signals generated by the p110delta PI3K isoform play critical roles in signaling through antigen and cytokine receptors and were shown to be required for induction of type 2, but not type 1, cytokine responses. OBJECTIVE We sought to determine the effect of genetic or pharmaceutical inactivation of p110delta PI3K on induction of IgE responses. METHODS We determined the effect of p110delta inactivation on induction of systemic IgE responses and on the ability of purified B lymphocytes to undergo IgE isotype switch in vitro. IgG and IgE germline transcription, postswitch transcription, protein expression, and secretion were measured, as well as cell division and expression of activation-induced cytidine deaminase, an enzyme required for isotype switch. RESULTS Paradoxically, inactivation of p110delta PI3K led to markedly increased IgE responses, despite reduced production of other antibody isotypes. This result was seen by using genetic inactivation of p110delta inhibition with IC87114 compound or blockade with the broad-spectrum PI3K inhibitors PIK-90 and PI-103. Significant increases in IgG1/IgE double-positive cells were observed, indicating that inactivation of PI3K leads to uncontrolled sequential switching from IgG1 to IgE. Disruption of p110delta signaling results in increased germline transcription at the epsilon locus and increased activation-induced cytidine deaminase expression, suggesting deregulation at the level of the isotype switch process. CONCLUSION Blockade of PI3K signaling leads to markedly enhanced B-cell switch to IgE and increased IgE levels in vivo, despite reduced type 2 cytokine production.
Collapse
Affiliation(s)
- Ting-Ting Zhang
- CIHR National Training Program in Allergy and Asthma Research, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, the Babraham Institute, Cambridge, United Kingdom
| | - Baher F Nashed
- CIHR National Training Program in Allergy and Asthma Research, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Zachary A Knight
- Howard Hughes Medical Institute, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, Calif
| | - Kevan M Shokat
- Howard Hughes Medical Institute, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, Calif
| | - Bart Vanhaesebroeck
- Institute of Cancer, Queen Mary's School of Medicine and Dentistry, University of London, London, United Kingdom
| | - Aaron J Marshall
- CIHR National Training Program in Allergy and Asthma Research, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
182
|
Yu Q, Quinn WJ, Salay T, Crowley JE, Cancro MP, Sen JM. Role of beta-catenin in B cell development and function. THE JOURNAL OF IMMUNOLOGY 2008; 181:3777-83. [PMID: 18768830 DOI: 10.4049/jimmunol.181.6.3777] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
beta-Catenin is a central mediator of Wnt signaling pathway, components of which have been implicated in B cell development and function. B cell progenitors and bone marrow stromal cells express Wnt ligands, Frizzled receptors and Wnt antagonists, suggesting fine tuned regulation of this pathway in B cell development. In particular, deletion of Frizzled 9 gene results in developmental defects at the pre-B stage of development and an accumulation of plasma cells. Furthermore, Wnt signals regulate B cell proliferation through lymphocyte enhancer-binding factor-1. However, it is not known whether Wnt signaling in B cell development is mediated by beta-catenin and whether beta-catenin plays a role in mature B cell function. In this report, we show that mice bearing B cell-specific deletion of beta-catenin have normal B cell development in bone marrow and periphery. A modest defect in plasma cell generation in vitro was documented, which correlated with a defective expression of IRF-4 and Blimp-1. However, B cell response to T-dependent and T-independent Ags in vivo was found to be normal. Thus, beta-catenin expression was found to be dispensable for normal B cell development and function.
Collapse
Affiliation(s)
- Qing Yu
- Lymphocyte Development Unit, Laboratory of Immunology, National Institute on Aging, National Institutes of Health, Baltimore MD 21224, USA
| | | | | | | | | | | |
Collapse
|
183
|
Dengler HS, Baracho GV, Omori SA, Bruckner S, Arden KC, Castrillon DH, DePinho RA, Rickert RC. Distinct functions for the transcription factor Foxo1 at various stages of B cell differentiation. Nat Immunol 2008; 9:1388-98. [PMID: 18978794 PMCID: PMC2679692 DOI: 10.1038/ni.1667] [Citation(s) in RCA: 261] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Accepted: 09/25/2008] [Indexed: 12/11/2022]
Abstract
The Foxo transcription factors (Foxo1, Foxo3, Foxo4) modulate cell fate decisions in diverse systems. Here we show that Foxo1-dependent gene expression was critical at multiple stages of B cell differentiation. Early deletion of Foxo1 caused a severe block at the pro-B cell stage, due to a failure to express interleukin 7 receptor α (IL-7Rα). Foxo1 inactivation in late pro-B cells resulted in an arrest at the pre-B cell stage due to a reduction in Rag1 and Rag2 expression. Deletion of Foxo1 in peripheral B cells led to fewer lymph node B cells due to reduced L-selectin expression, and failed class switch recombination due to impaired Aicda upregulation. Thus, Foxo1 regulates a transcriptional program that is essential for early B cell development and peripheral B cell function.
Collapse
Affiliation(s)
- Hart S Dengler
- Program of Inflammatory Disease Research, Burnham Institute for Medical Research, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
184
|
Abstract
B lymphocyte-induced maturation protein-1 (Blimp-1), discovered 16 years ago as a transcriptional repressor of the IFNbeta promoter, plays fundamentally important roles in many cell lineages and in early development. This review focuses on Blimp-1 in lymphocytes. In the B cell lineage, Blimp-1 is required for development of immunoglobulin-secreting cells and for maintenance of long-lived plasma cells (LLPCs). Direct targets of Blimp-1 and the transcriptional cascades Blimp-1 initiates to trigger plasmacytic differentiation are described. Blimp-1 also affects the homeostasis and function of CD4(+), CD8(+), and regulatory CD4(+) T cells, and Blimp-1 levels are highest in antigen-experienced T cells. Blimp-1 attenuates T cell proliferation and survival and modulates differentiation. Roles for Blimp-1 in Th1/Th2 specification, regulatory T cell function, and CD8 differentiation and function are under investigation. Signals that induce Blimp-1 in B cells include Toll-like receptor ligands and cytokines; in T cells, T cell receptors and cytokines induce Blimp-1. In spite of some commonalities, different targets and regulators of Blimp-1 in B and T cells suggest intriguing evolutionary divergence of this regulatory machinery.
Collapse
Affiliation(s)
- Gislâine Martins
- Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA.
| | | |
Collapse
|
185
|
Calame K. Activation-dependent induction of Blimp-1. Curr Opin Immunol 2008; 20:259-64. [PMID: 18554885 DOI: 10.1016/j.coi.2008.04.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 04/15/2008] [Accepted: 04/15/2008] [Indexed: 11/25/2022]
Abstract
B lymphocyte induced maturation protein-1 (Blimp-1) mRNA is induced upon antigen-dependent activation of both T and B lymphocytes, in spite of the fact that it plays very different roles in the two lineages. B cells have at least four different mechanisms to repress Blimp-1 and repression is relieved before induction. Only one repressor, Bcl-6, is known in T cells. Activators must also be present to induce Blimp-1 in both T and B cells. Cytokines IL-21, IL-10, and IL-6, activating STAT3, are crucial in B cells along with toll-like receptor (TLR) signals, whereas IL-2 is crucial in T cells. AP-1, NF-kappaB, and IRF4 also activate Blimp-1.
Collapse
Affiliation(s)
- Kathryn Calame
- Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
186
|
Janas ML, Hodson D, Stamataki Z, Hill S, Welch K, Gambardella L, Trotman LC, Pandolfi PP, Vigorito E, Turner M. The effect of deleting p110delta on the phenotype and function of PTEN-deficient B cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:739-46. [PMID: 18178811 DOI: 10.4049/jimmunol.180.2.739] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Control of the intracellular levels of phosphatidylinositol-(3, 4, 5)-trisphosphate by PI3K and phosphatase and tensin homolog (PTEN) is essential for B cell development and differentiation. Deletion of the PI3K catalytic subunit p110delta leads to a severe reduction in B1 and marginal zone (MZ) B cells, whereas deletion of PTEN results in their expansion. We have examined the relationship between these two molecules by generating mice with a B cell-specific deletion of PTEN (PTENB) and a concurrent germline deletion of p110delta. The expanded B1 cell population of PTENB mice was reduced to normal levels in PTENB/p110delta mutant mice, indicating a critical role for the p110delta isoform in the expansion of B1 cells. However, numbers of MZ B cells in the PTENB/p110delta mutants was intermediate between wild-type and PTENB-deficient mice, suggesting an additional role for other PI3K catalytic isoforms in MZ differentiation. Furthermore, the defective class switch recombination in PTENB B cells was only partially reversed in PTENB/p110delta double mutant B cells. These results demonstrate an epistatic relationship between p110delta and PTEN. In addition, they also suggest that additional PI3K catalytic subunits contribute to B cell development and function.
Collapse
Affiliation(s)
- Michelle L Janas
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Doi T, Obayashi K, Kadowaki T, Fujii H, Koyasu S. PI3K is a negative regulator of IgE production. Int Immunol 2008; 20:499-508. [PMID: 18303010 DOI: 10.1093/intimm/dxn009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The production of IgE, a main player in allergic disorders such as asthma and atopic dermatitis, is strictly regulated and the serum concentrations of IgE are normally kept at a much lower level than other isotypes. We found that mice deficient for the p85alpha regulatory subunit of class IA phosphoinositide 3-kinase (PI3K) produced increasing amounts of serum IgE. Purified p85alpha-/- B cells produced more IgE than wild-type B cells in vitro in response to anti-CD40 mAb and IL-4. PI3K inhibitors wortmannin and IC87114 enhanced IgE production by wild-type B cells stimulated with anti-CD40 mAb and IL-4. Under the same condition, antigen receptor cross-linking induced the expression of inhibitor of differentiation-2 and suppressed the expression of activation-induced cytidine deaminase and class switch recombination (CSR) in a PI3K-dependent manner. IgE production was also suppressed in a concentrated cell culture condition, which was completely reversed by PI3K inhibition. The selective suppression of IgE production by PI3K was also observed at a protein level after CSR. Our results indicate that PI3K negatively regulates IgE production at both CSR and protein levels.
Collapse
Affiliation(s)
- Tomomitsu Doi
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | |
Collapse
|
188
|
Kuo CC, Lin SC. Altered FOXO1 transcript levels in peripheral blood mononuclear cells of systemic lupus erythematosus and rheumatoid arthritis patients. Mol Med 2008; 13:561-6. [PMID: 17873969 PMCID: PMC1976859 DOI: 10.2119/2007-00021.kuo] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2007] [Accepted: 08/20/2007] [Indexed: 11/06/2022] Open
Abstract
FOXO forkhead transcription factors play an important role in controlling lymphocyte activation and proliferation. To evaluate the possibility that FOXO transcriptional expression is dysregulated in systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) patients, we determined the quantities of FOXO1, FOXO3a, and FOXO4 transcripts in peripheral blood mononuclear cells (PBMCs) from normal controls as well as from SLE and RA patients. Results showed that FOXO1 and FOXO3a are dominant FOXO factors at the transcript level in PBMCs. Statistical analysis showed that the FOXO1 transcript levels in RA patients and in SLE patients with active disease activity were significantly lower than those in normal controls, and the FOXO1 transcript levels were inversely correlated with lupus disease activity. In contrast, the differences in FOXO3a and FOXO4 transcript levels between normal controls and patients were not significant. These data suggest that the transcriptional dysregulation in FOXO1 is possibly linked to the pathogenesis of SLE and RA.
Collapse
Affiliation(s)
- Chia-Chen Kuo
- Division of Allergy and Immunology, Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | | |
Collapse
|
189
|
Donahue AC, Fruman DA. Distinct signaling mechanisms activate the target of rapamycin in response to different B-cell stimuli. Eur J Immunol 2007; 37:2923-36. [PMID: 17724683 DOI: 10.1002/eji.200737281] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Phosphoinositide 3-kinase (PI3K) and the mammalian target of rapamycin (mTOR), a downstream kinase, are both required for proliferation of splenic B cells. However, the functions of PI3K and mTOR in response to different stimuli and among B cell subsets have not been fully elucidated. We used flow cytometry and magnetic cell sorting to examine the requirement for PI3K and mTOR in responses of splenic B cell subsets to BCR and LPS stimulation. BCR-mediated phosphorylation of Akt and Erk is sensitive to the PI3K catalytic inhibitor wortmannin in both marginal zone (MZ) and follicular (FO) cells. BCR-mediated mTOR activation in both subsets is inhibited by wortmannin, though less strongly in MZ cells. In contrast, LPS-induced mTOR signaling is strikingly resistant to wortmannin in both subsets. Similarly, functional responses to LPS are partially wortmannin resistant yet sensitive to mTOR inhibition by rapamycin. We also observed mitogen-independent mTOR activity that is regulated by nutrient availability, and is significantly elevated in MZ cells relative to FO cells. These data define both similarities and differences in PI3K/mTOR signaling mechanisms in MZ and FO cells, and suggest that mTOR signaling can occur in the absence of PI3K activation to promote B cell responses to LPS.
Collapse
Affiliation(s)
- Amber C Donahue
- Department of Molecular Biology & Biochemistry, and Center for Immunology, University of California Irvine, Irvine, CA 92697-3900, USA
| | | |
Collapse
|
190
|
Heltemes-Harris LM, Gearhart PJ, Ghosh P, Longo DL. Activation-induced deaminase-mediated class switch recombination is blocked by anti-IgM signaling in a phosphatidylinositol 3-kinase-dependent fashion. Mol Immunol 2007; 45:1799-806. [PMID: 17983655 DOI: 10.1016/j.molimm.2007.09.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 09/21/2007] [Accepted: 09/22/2007] [Indexed: 11/28/2022]
Abstract
Activation-induced deaminase (AID) is expressed in activated B lymphocytes and initiates somatic hypermutation and class switch recombination. To determine if different stimuli affect the expression and function of AID, we monitored AID activity in murine B cells stimulated ex vivo with various ligands. AID was rapidly expressed at both the RNA and protein levels following stimulation with LPS, LPS plus IL-4, and anti-CD40 plus IL-4, but was delayed after stimulation with anti-IgM plus IL-4. By day 4, AID was expressed in all groups; however, cells stimulated with anti-IgM plus IL-4 did not undergo switch recombination. These cells expressed normal levels of gamma 1 germline transcripts, implying that the gamma 1 switch region was accessible. Furthermore, switching was suppressed by the addition of anti-IgM to cells stimulated with LPS plus IL-4 or anti-CD40 plus IL-4, even though AID was expressed. The lack of class switch recombination could be reversed by inhibition of phosphatidylinositol 3-kinase (PI3K). This suggests that activation through the B cell receptor induces PI3K, which interferes with the function of AID.
Collapse
Affiliation(s)
- Lynn M Heltemes-Harris
- Laboratory of Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | | | | |
Collapse
|
191
|
Ji H, Rintelen F, Waltzinger C, Bertschy Meier D, Bilancio A, Pearce W, Hirsch E, Wymann MP, Rückle T, Camps M, Vanhaesebroeck B, Okkenhaug K, Rommel C. Inactivation of PI3Kgamma and PI3Kdelta distorts T-cell development and causes multiple organ inflammation. Blood 2007; 110:2940-7. [PMID: 17626838 DOI: 10.1182/blood-2007-04-086751] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mice lacking both the p110gamma and p110delta isoforms display severe impairment of thymocyte development. Here, we show that this phenotype is recapitulated in p110gamma-/-/p110delta(D910A/D910A) (p110gamma(KO)delta(D910A)) mice where the p110delta isoform has been inactivated by a point mutation. Moreover, we have examined the pathological consequences of the p110gammadelta deficiency, which include profound T-cell lymphopenia, T-cell and eosinophil infiltration of mucosal organs, elevated IgE levels, and a skewing toward Th2 immune responses. Using small-molecule selective inhibitors, we demonstrated that in mature T cells, p110delta, but not p110gamma, controls Th1 and Th2 cytokine secretion. Thus, the pathology in the p110gammadelta-deficient mice is likely to be secondary to a developmental block in the thymus that leads to lymphopenia-associated inflammatory responses.
Collapse
Affiliation(s)
- Hong Ji
- Merck Serono S. A., Geneva Research Center, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Péron S, Pan-Hammarström Q, Imai K, Du L, Taubenheim N, Sanal O, Marodi L, Bergelin-Besançon A, Benkerrou M, de Villartay JP, Fischer A, Revy P, Durandy A. A primary immunodeficiency characterized by defective immunoglobulin class switch recombination and impaired DNA repair. ACTA ACUST UNITED AC 2007; 204:1207-16. [PMID: 17485519 PMCID: PMC2118580 DOI: 10.1084/jem.20070087] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Immunoglobulin class switch recombination (CSR) deficiencies are rare primary immunodeficiencies, characterized by a lack of switched isotype (IgG, IgA, or IgE) production, variably associated with abnormal somatic hypermutation (SHM). Deficiencies in CD40 ligand, CD40, activation-induced cytidine deaminase, and uracil-N-glycosylase may account for this syndrome. We previously described another Ig CSR deficiency condition, characterized by a defect in CSR downstream of the generation of double-stranded DNA breaks in switch (S) μ regions. Further analysis performed with the cells of five affected patients showed that the Ig CSR deficiency was associated with an abnormal formation of the S junctions characterized by microhomology and with increased cell radiosensitivity. In addition, SHM was skewed toward transitions at G/C residues. Overall, these findings suggest that a unique Ig CSR deficiency phenotype could be related to an as-yet-uncharacterized defect in a DNA repair pathway involved in both CSR and SHM events.
Collapse
Affiliation(s)
- Sophie Péron
- Institut National de la Santé et de la Recherche Médicale, U768, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Kallies A, Nutt SL. Terminal differentiation of lymphocytes depends on Blimp-1. Curr Opin Immunol 2007; 19:156-62. [PMID: 17291741 DOI: 10.1016/j.coi.2007.01.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Accepted: 01/31/2007] [Indexed: 01/05/2023]
Abstract
B lymphocyte induced maturation protein 1 (Blimp-1) has long been considered a master regulator of the terminal differentiation of B cells into antibody-secreting plasma cells. Gene-targeting experiments have now demonstrated that quantitative changes in Blimp-1 expression define plasma cell ontogeny--a process that requires the continual function of Blimp-1. Recently, new roles for Blimp-1 have been revealed, as a suppressor of diffuse large B cell lymphoma and as a key regulator of T-cell differentiation. Blimp-1 is expressed in differentiated effector T cells and controls their homeostasis. These new findings suggest that Blimp-1 has a conserved function in the final differentiation of both the cellular and the humoral arm of the adaptive immune response.
Collapse
Affiliation(s)
- Axel Kallies
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, Australia
| | | |
Collapse
|
194
|
Okkenhaug K, Ali K, Vanhaesebroeck B. Antigen receptor signalling: a distinctive role for the p110delta isoform of PI3K. Trends Immunol 2007; 28:80-7. [PMID: 17208518 PMCID: PMC2358943 DOI: 10.1016/j.it.2006.12.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Revised: 11/24/2006] [Accepted: 12/18/2006] [Indexed: 11/24/2022]
Abstract
The activation of antigen receptors triggers two important signalling pathways originating from phosphatidylinositol(4,5)-bisphosphate [PtdIns(4,5)P2]. The first is phospholipase Cγ (PLCγ)-mediated hydrolysis of PtdIns(4,5)P2, resulting in the activation of Ras, protein kinase C and Ca2+ flux. This culminates in profound alterations in gene expression and effector-cell responses, including secretory granule exocytosis and cytokine production. By contrast, phosphoinositide 3-kinases (PI3Ks) phosphorylate PtdIns(4,5)P2 to yield phosphatidylinositol(3,4,5)-trisphosphate, activating signalling pathways that overlap with PLCγ or are PI3K-specific. Pathways that are PI3K-specific include Akt-mediated inactivation of Foxo transcription factors and transcription-independent regulation of glucose uptake and metabolism. The p110δ isoform of PI3K is the main source of PI3K activity following antigen recognition by B cells, T cells and mast cells. Here, we review the roles of p110δ in regulating antigen-dependent responses in these cell types.
Collapse
Affiliation(s)
- Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK, CB2 4AT
| | - Khaled Ali
- Ludwig Institute for Cancer Research, London, UK, W1W 7BS
- Department of Biochemistry and Molecular Biology, University College London, London, UK, WC1E 6BT
| | - Bart Vanhaesebroeck
- Ludwig Institute for Cancer Research, London, UK, W1W 7BS
- Department of Biochemistry and Molecular Biology, University College London, London, UK, WC1E 6BT
| |
Collapse
|
195
|
Pan-Hammarström Q, Zhao Y, Hammarström L. Class switch recombination: a comparison between mouse and human. Adv Immunol 2007; 93:1-61. [PMID: 17383538 DOI: 10.1016/s0065-2776(06)93001-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Humans and mice separated more than 60 million years ago. Since then, evolution has led to a multitude of changes in their genomic sequences. The divergence of genes has resulted in differences both in the innate and adaptive immune systems. In this chapter, we focus on species difference with regard to immunoglobulin class switch recombination (CSR). We have compared the immunoglobulin constant region gene loci from human and mouse, with an emphasis on the switch regions, germ line transcription promoters, and 3' enhancers. We have also compared pathways/factors that are involved in CSR. Although there are remarkable similarities in the cellular machinery involved in CSR, there are also a number of unique features in each species.
Collapse
Affiliation(s)
- Qiang Pan-Hammarström
- Department of Laboratory Medicine, Division of Clinical Immunology, Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden
| | | | | |
Collapse
|