Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Nat Immunol ; 24(12): 2108-2120, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37932457

ABSTRACT

Regulatory T cells (Treg cells) are instrumental in establishing immunological tolerance. However, the precise effector mechanisms by which Treg cells control a specific type of immune response in a given tissue remains unresolved. By simultaneously studying Treg cells from different tissue origins under systemic autoimmunity, in the present study we show that interleukin (IL)-27 is specifically produced by intestinal Treg cells to regulate helper T17 cell (TH17 cell) immunity. Selectively increased intestinal TH17 cell responses in mice with Treg cell-specific IL-27 ablation led to exacerbated intestinal inflammation and colitis-associated cancer, but also helped protect against enteric bacterial infection. Furthermore, single-cell transcriptomic analysis has identified a CD83+CD62Llo Treg cell subset that is distinct from previously characterized intestinal Treg cell populations as the main IL-27 producers. Collectively, our study uncovers a new Treg cell suppression mechanism crucial for controlling a specific type of immune response in a particular tissue and provides further mechanistic insights into tissue-specific Treg cell-mediated immune regulation.


Subject(s)
Interleukin-27 , T-Lymphocytes, Regulatory , Mice , Animals , T-Lymphocytes, Helper-Inducer , Immune Tolerance , Immunity, Cellular , Th17 Cells
2.
J Exp Med ; 220(10)2023 10 02.
Article in English | MEDLINE | ID: mdl-37516921

ABSTRACT

Effector regulatory T cells (eTregs) exhibit distinct homeostatic properties and superior suppressor capacities pivotal for controlling immune responses mediated by their conventional T cell counterpart. While the role of microRNAs (miRNAs) in Tregs has been well-established, how miRNAs regulate eTregs remains poorly understood. Here, we demonstrate that miR-15/16 clusters act as key regulators in limiting eTreg responses. Loss of miR-15/16 clusters leads to increased eTreg frequencies with enhanced suppressor function. Consequently, mice with Treg-specific ablation of miR-15/16 clusters display attenuated immune responses during neuroinflammation and upon both infectious and non-infectious challenges. Mechanistically, miR-15/16 clusters exert their regulatory effect in part through repressing IRF4, a transcription factor essential for eTreg differentiation and function. Moreover, miR-15/16 clusters also directly target neuritin, an IRF4-dependent molecule, known for its role in Treg-mediated regulation of plasma cell responses. Together, we identify an miRNA family that controls an important Treg subset and further demonstrate that eTreg responses are tightly regulated at both transcriptional and posttranscriptional levels.


Subject(s)
MicroRNAs , T-Lymphocytes, Regulatory , Animals , Mice , Lymphocyte Activation , Cell Differentiation/genetics , Homeostasis , MicroRNAs/genetics
3.
bioRxiv ; 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36865314

ABSTRACT

Regulatory T (Treg) cells are instrumental in establishing immunological tolerance. However, the precise effector mechanisms by which Treg cells control a specific type of immune response in a given tissue remains unresolved. By simultaneously studying Treg cells from different tissue origins under systemic autoimmunity, here we show that IL-27 is specifically produced by intestinal Treg cells to regulate Th17 immunity. Selectively increased intestinal Th17 responses in mice with Treg cell-specific IL-27 ablation led to exacerbated intestinal inflammation and colitis-associated cancer, but also helped protect against enteric bacterial infection. Furthermore, single-cell transcriptomic analysis has identified a CD83+TCF1+ Treg cell subset that is distinct from previously characterized intestinal Treg cell populations as the main IL-27 producers. Collectively, our study uncovers a novel Treg cell suppression mechanism crucial for controlling a specific type of immune response in a particular tissue, and provides further mechanistic insights into tissue-specific Treg cell-mediated immune regulation.

4.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34554189

ABSTRACT

IL-27 controls a diverse range of immune responses in many disease settings. Here, we identify intestinal epithelial cells (IECs) as one of the major IL-27 cellular sources in the gut-associated tissue. Unlike IL-27 secreted by innate immune cells, gut epithelial IL-27 is dispensable for T-bet+ regulatory T cell (T reg cell) differentiation or IL-10 induction. Rather, IEC-derived IL-27 specifically promotes a distinct CD8αα+CD4+ intraepithelial lymphocyte (IEL) population that acquires their functional differentiation at the intestinal epithelium. Loss of IL-27 in IECs leads to a selective defect in CD8αα+CD4+ IELs over time. Consequently, mice with IEC-specific IL-27 ablation exhibited elevated pathogen burden during parasitic infection, and this could be rescued by transfer of exogenous CD8αα+CD4+ IELs. Collectively, our data reveal that in addition to its known regulatory properties in preventing immune hyperactivity, gut epithelial IL-27 confers barrier immunity by inducing a specific IEL subset and further suggest that IL-27 produced by different cell types plays distinct roles in maintaining intestinal homeostasis.


Subject(s)
Epithelial Cells/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Intraepithelial Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Female , Homeostasis/immunology , Male , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/immunology , Signal Transduction/immunology
5.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33125052

ABSTRACT

During thymocyte development, medullary thymic epithelial cells (mTECs) provide appropriate instructive cues in the thymic microenvironment for not only negative selection but also the generation of regulatory T (T reg) cells. Here, we identify that miR-155, a microRNA whose expression in T reg cells has previously been shown to be crucial for their development and homeostasis, also contributes to thymic T reg (tT reg) cell differentiation by promoting mTEC maturation. Mechanistically, we show that RANKL stimulation induces expression of miR-155 to safeguard the thymic medulla through targeting multiple known and previously uncharacterized molecules within the TGFß signaling pathway, which is recognized for its role in restricting the maturation and expansion of mTECs. Our work uncovers a miR-155-TGFß axis in the thymic medulla to determine mTEC maturity and, consequently, the quantity of tT reg cells and suggests that miR-155 ensures proper tT reg cell development in both cell-intrinsic and -extrinsic manners.


Subject(s)
Epithelial Cells/immunology , MicroRNAs/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Signal Transduction/immunology , Transforming Growth Factor beta/immunology
6.
Sci Adv ; 5(12): eaaw1715, 2019 12.
Article in English | MEDLINE | ID: mdl-31844658

ABSTRACT

Follicular helper T (TFH) cells are essential for generating protective humoral immunity. To date, microRNAs (miRNAs) have emerged as important players in regulating TFH cell biology. Here, we show that loss of miR-23~27~24 clusters in T cells resulted in elevated TFH cell frequencies upon different immune challenges, whereas overexpression of this miRNA family led to reduced TFH cell responses. Mechanistically, miR-23~27~24 clusters coordinately control TFH cells through targeting a network of genes that are crucial for TFH cell biology. Among them, thymocyte selection-associated HMG-box protein (TOX) was identified as a central transcription regulator in TFH cell development. TOX is highly up-regulated in both mouse and human TFH cells in a BCL6-dependent manner. In turn, TOX promotes the expression of multiple molecules that play critical roles in TFH cell differentiation and function. Collectively, our results establish a key miRNA regulon that maintains optimal TFH cell responses for resultant humoral immunity.


Subject(s)
Cell Differentiation/genetics , Immunity, Humoral/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes/immunology , Animals , Gene Expression Regulation, Developmental/immunology , High Mobility Group Proteins/genetics , Humans , Immunity, Humoral/immunology , Lymphocyte Activation/immunology , Mice , MicroRNAs/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Signal Transduction , T-Lymphocytes, Helper-Inducer/metabolism
7.
Nat Commun ; 9(1): 2757, 2018 07 16.
Article in English | MEDLINE | ID: mdl-30013024

ABSTRACT

Reciprocal interactions between B and follicular T helper (Tfh) cells orchestrate the germinal center (GC) reaction, a hallmark of humoral immunity. Abnormal GC responses could lead to the production of pathogenic autoantibodies and the development of autoimmunity. Here we show that miR-146a controls GC responses by targeting multiple CD40 signaling pathway components in B cells; by contrast, loss of miR-146a in T cells does not alter humoral responses. However, specific deletion of both miR-146a and its paralog, miR-146b, in T cells increases Tfh cell numbers and enhanced GC reactions. Thus, our data reveal differential cell-intrinsic regulations of GC B and Tfh cells by miR-146a and miR-146b. Together, members of the miR-146 family serve as crucial molecular brakes to coordinately control GC reactions to generate protective humoral responses without eliciting unwanted autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , MicroRNAs/genetics , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoantibodies/biosynthesis , Autoimmunity/genetics , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , CD40 Antigens/genetics , CD40 Antigens/immunology , Cell Differentiation , Gene Expression Regulation , Germinal Center/cytology , Germinal Center/drug effects , Immunity, Humoral/genetics , Interleukin-4/pharmacology , Mice , Mice, Transgenic , MicroRNAs/immunology , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects
8.
Article in English | WPRIM (Western Pacific) | ID: wpr-691352

ABSTRACT

<p><b>OBJECTIVE</b>To explore brain activations associated with electroacupuncture simulation at Tongli (HT 5) and its comparison with brain activations during picture-naming task.</p><p><b>METHODS</b>Twenty healthy subjects were enrolled in this study. Half of them received electroacupuncture stimulation at HT 5 (ACUP group) and the other half of them received stimulation at a nonmeridian sham acupoint (SHAM group). All subjects performed picture-naming task. Each subject finished two runs of functional magnetic resonance imaging examinations in one session and picture-naming task was performed before electroacupuncture stimulation. Subjective brain activations were obtained using generalized linear model and inter-group analyses were performed after that.</p><p><b>RESULTS</b>The electroacupuncture stimulation at HT 5 induced significant brain activations in both the anterior and posterior language regions, including the left inferior frontal gyrus, which was in consistent with activations induced during picture-naming task. Group analysis showed a tendency of increased activation of ACUP group in left inferior frontal gyrus compared with SHAM group (P<0.05 FDR corrected).</p><p><b>CONCLUSIONS</b>Electroacupuncture treatment at the acupoint HT 5 has modulation effect on typical language-implicated brain regions in healthy subjects, which provides supporting evidence for beneficial effects of needling at HT 5 for recovery of language function in aphasia.</p>

9.
J Immunol ; 198(10): 3919-3926, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28404635

ABSTRACT

miR-23∼27∼24 was recently implicated in restricting Th2 immunity, as well as the differentiation and function of other effector T cell lineages. Interestingly, miR-24, unlike other family members, actually promotes Th1 and Th17 responses. In this article, we show that miR-24 drives the production of IFN-γ and IL-17 in T cells at least in part through targeting TCF1, a transcription factor known for its role in limiting Th1 and Th17 immunity. Surprisingly, whereas TCF1 was previously shown to promote Th2 responses through inducing GATA3, enforced TCF1 expression in miR-24-overexpressing T cells led to further downregulation of IL-4 and GATA3 expression, suggesting miR-24-mediated inhibition of Th2 immunity cannot be attributed to TCF1 repression by miR-24. Together, our data demonstrate a novel miR-24-TCF1 pathway in controlling effector cytokine production by T cells and further suggest miR-24 could function as a key upstream molecule regulating TCF1-mediated immune responses.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , T-Lymphocyte Subsets/immunology , Animals , Cell Differentiation , Cytokines/biosynthesis , Cytokines/immunology , Down-Regulation , GATA3 Transcription Factor/biosynthesis , Hepatocyte Nuclear Factor 1-alpha/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocyte Activation , Mice , Signal Transduction , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
10.
J Clin Invest ; 127(2): 530-542, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28067667

ABSTRACT

MicroRNAs (miRs) are tightly regulated in the immune system, and aberrant expression of miRs often results in hematopoietic malignancies and autoimmune diseases. Previously, it was suggested that elevated levels of miR-27 in T cells isolated from patients with multiple sclerosis facilitate disease progression by inhibiting Th2 immunity and promoting pathogenic Th1 responses. Here we have demonstrated that, although mice with T cell-specific overexpression of miR-27 harbor dysregulated Th1 responses and develop autoimmune pathology, these disease phenotypes are not driven by miR-27 in effector T cells in a cell-autonomous manner. Rather, dysregulation of Th1 responses and autoimmunity resulted from a perturbed Treg compartment. Excessive miR-27 expression in murine T cells severely impaired Treg differentiation. Moreover, Tregs with exaggerated miR-27-mediated gene regulation exhibited diminished homeostasis and suppressor function in vivo. Mechanistically, we determined that miR-27 represses several known as well as previously uncharacterized targets that play critical roles in controlling multiple aspects of Treg biology. Collectively, our data show that miR-27 functions as a key regulator in Treg development and function and suggest that proper regulation of miR-27 is pivotal to safeguarding Treg-mediated immunological tolerance.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , Immune Tolerance , MicroRNAs/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/genetics , Mice , Mice, Transgenic , MicroRNAs/genetics , Th1 Cells/immunology , Th2 Cells/immunology
11.
J Exp Med ; 213(2): 235-49, 2016 Feb 08.
Article in English | MEDLINE | ID: mdl-26834155

ABSTRACT

Coordinated repression of gene expression by evolutionarily conserved microRNA (miRNA) clusters and paralogs ensures that miRNAs efficiently exert their biological impact. Combining both loss- and gain-of-function genetic approaches, we show that the miR-23∼27∼24 clusters regulate multiple aspects of T cell biology, particularly helper T (Th) 2 immunity. Low expression of this miRNA family confers proper effector T cell function at both physiological and pathological settings. Further studies in T cells with exaggerated regulation by individual members of the miR-23∼27∼24 clusters revealed that miR-24 and miR-27 collaboratively limit Th2 responses through targeting IL-4 and GATA3 in both direct and indirect manners. Intriguingly, although overexpression of the entire miR-23 cluster also negatively impacts other Th lineages, enforced expression of miR-24, in contrast to miR-23 and miR-27, actually promotes the differentiation of Th1, Th17, and induced regulatory T cells, implying that under certain conditions, miRNA families can fine tune the biological effects of their regulation by having individual members antagonize rather than cooperate with each other. Together, our results identify a miRNA family with important immunological roles and suggest that tight regulation of miR-23∼27∼24 clusters in T cells is required to maintain optimal effector function and to prevent aberrant immune responses.


Subject(s)
MicroRNAs/genetics , MicroRNAs/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , Asthma/genetics , Asthma/immunology , Asthma/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Disease Models, Animal , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/genetics , Gene Expression Regulation , Gene Regulatory Networks , Interleukin-4/biosynthesis , Interleukin-4/genetics , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Multigene Family , Phenotype , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Th2 Cells/cytology , Th2 Cells/immunology
12.
Immunity ; 43(1): 52-64, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26163372

ABSTRACT

MicroRNA (miRNA)-dependent regulation of gene expression confers robustness to cellular phenotypes and controls responses to extracellular stimuli. Although a single miRNA can regulate expression of hundreds of target genes, it is unclear whether any of its distinct biological functions can be due to the regulation of a single target. To explore in vivo the function of a single miRNA-mRNA interaction, we mutated the 3' UTR of a major miR-155 target (SOCS1) to specifically disrupt its regulation by miR-155. We found that under physiologic conditions and during autoimmune inflammation or viral infection, some immunological functions of miR-155 were fully or largely attributable to the regulation of SOCS1, whereas others could be accounted only partially or not at all by this interaction. Our data suggest that the role of a single miRNA-mRNA interaction is dependent on cell type and biological context.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , MicroRNAs/genetics , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes, Regulatory/immunology , 3' Untranslated Regions/genetics , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Profiling , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Killer Cells, Natural/transplantation , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Mutation , RNA, Messenger/genetics , Suppressor of Cytokine Signaling 1 Protein
13.
PLoS Pathog ; 11(2): e1004635, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25658840

ABSTRACT

IFNγ signaling drives dendritic cells (DCs) to promote type I T cell (Th1) immunity. Here, we show that activation of DCs by IFNγ is equally crucial for the differentiation of a population of T-bet+ regulatory T (Treg) cells specialized to inhibit Th1 immune responses. Conditional deletion of IFNγ receptor in DCs but not in Treg cells resulted in a severe defect in this specific Treg cell subset, leading to exacerbated immune pathology during parasitic infections. Mechanistically, IFNγ-unresponsive DCs failed to produce sufficient amount of IL-27, a cytokine required for optimal T-bet induction in Treg cells. Thus, IFNγ signalling endows DCs with the ability to efficiently control a specific type of T cell immunity through promoting a corresponding Treg cell population.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Interferon-gamma/immunology , T-Lymphocytes, Regulatory/immunology , Toxoplasmosis/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Inflammation/immunology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , T-Box Domain Proteins/immunology , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Th1 Cells/immunology
14.
Cell ; 142(6): 914-29, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20850013

ABSTRACT

Foxp3(+) regulatory T (Treg) cells maintain immune homeostasis by limiting different types of inflammatory responses. Here, we report that miR-146a, one of the miRNAs prevalently expressed in Treg cells, is critical for their suppressor function. The deficiency of miR-146a in Treg cells resulted in a breakdown of immunological tolerance manifested in fatal IFNγ-dependent immune-mediated lesions in a variety of organs. This was likely due to augmented expression and activation of signal transducer and activator transcription 1 (Stat1), a direct target of miR-146a. Likewise, heightened Stat1 activation in Treg cells subjected to a selective ablation of SOCS1, a key negative regulator of Stat1 phosphorylation downstream of the IFNγ receptor, was associated with analogous Th1-mediated pathology. Our results suggest that specific aspects of Treg suppressor function are controlled by a single miRNA and that an optimal range of Stat1 activation is important for Treg-mediated control of Th1 responses and associated autoimmunity.


Subject(s)
MicroRNAs/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Interferon-gamma/immunology , Mice , Mice, Knockout , MicroRNAs/genetics , STAT1 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology
15.
Zhongguo Gu Shang ; 21(8): 626-8, 2008 Aug.
Article in Chinese | MEDLINE | ID: mdl-19108384

ABSTRACT

OBJECTIVE: To study the therapeutic effect of combined multifunctional external fixator for treatment of tibiofibular fracture. METHODS: From Oct 1999 to Apr 2006, 37 patients of tibiofibular fractures were treated with combined multifunctional external fixator. There were 28 males and 9 females with an average of 47.8 years (range,from 22 to 76 years). There were 7 cases with transverse fracture, 18 comminuted fracture, 8 spiral fracture and 4 oblique fracture;closed fracture was in 29 cases and open fracture was in 8. RESULTS: All the patients were followed up 6 to 13 months (mean,9 months)and achieved union of the fractures. CONCLUSION: Combined multifunctional external fixator for tibiofibular fracture has many advantages such as minimal invasion, convenient operation, reliable fixation and high union ratio of fracture. Moreover,the treatment has no influence on motion of knee and ankle joint, even patient can walk with external fixator without long-term bed rest.


Subject(s)
External Fixators , Fibula/injuries , Tibial Fractures/surgery , Adult , Aged , Female , Humans , Male , Middle Aged
16.
Blood ; 110(1): 193-200, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17360936

ABSTRACT

The recruitment of tumor necrosis factor receptor-associated factors (TRAFs) 1, 2, 3, 5, and 6 to the CD40 cytoplasmic tail upon CD40 trimerization results in downstream signaling events that ultimately lead to CD40-dependent, thymus-dependent (TD) humoral immune responses. Previously, we have shown signaling through the C-terminal tail of CD40 in the absence of canonical TRAF-binding sites is capable of signaling through an alternative TRAF2-binding site. Here, we demonstrate that B cells from mice harboring CD40 with only the C-terminal tail can activate both canonical and noncanonical NFkappaB signaling pathways. Moreover, while lacking germinal center formation, several hallmarks of humoral immune responses including clonal B-cell activation/expansion, antibody isotype switching, and affinity maturation remain normal. This study demonstrates a new functional domain in CD40 that controls critical aspects of B-cell immunity in an in vivo setting.


Subject(s)
B-Lymphocytes/cytology , CD40 Antigens/physiology , TNF Receptor-Associated Factor 2/metabolism , Animals , Antibody Formation , Binding Sites , CD40 Antigens/chemistry , CD40 Antigens/immunology , Cell Differentiation , Cell Proliferation , Mice , NF-kappa B/metabolism , Signal Transduction
17.
J Exp Med ; 199(1): 91-8, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14707116

ABSTRACT

Long-lived humoral immunity is manifested by the ability of bone marrow plasma cells (PCs) to survive for extended periods of time. Recent studies have underscored the importance of BLyS and APRIL as factors that can support the survival of B lineage lymphocytes. We show that BLyS can sustain PC survival in vitro, and this survival can be further enhanced by interleukin 6. Selective up-regulation of Mcl-1 in PCs by BLyS suggests that this alpha-apoptotic gene product may play an important role in PC survival. Blockade of BLyS, via transmembrane activator and cyclophilin ligand interactor-immunoglobulin treatment, inhibited PC survival in vitro and in vivo. Heightened expression of B cell maturation antigen (BCMA), and lowered expression of transmembrane activator and cyclophilin ligand interactor and BAFF receptor in PCs relative to resting B cells suggests a vital role of BCMA in PC survival. Affirmation of the importance of BCMA in PC survival was provided by studies in BCMA-/- mice in which the survival of long-lived bone marrow PCs was impaired compared with wild-type controls. These findings offer new insights into the molecular basis for the long-term survival of PCs.


Subject(s)
Bone Marrow Cells/cytology , Cell Survival/physiology , Plasma Cells/cytology , Receptors, Tumor Necrosis Factor/physiology , Animals , B-Cell Activating Factor , B-Cell Maturation Antigen , Base Sequence , Bone Marrow Cells/physiology , DNA Primers , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/physiology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/physiology
18.
Proc Natl Acad Sci U S A ; 100(22): 12905-10, 2003 Oct 28.
Article in English | MEDLINE | ID: mdl-14555759

ABSTRACT

The coordinated regulation of chemokine responsiveness plays a critical role in the development of humoral immunity. After antigen challenge and B cell activation, the emerging plasma cells (PCs) undergo CXCL12-induced chemotaxis to the bone marrow, where they produce Ab and persist. Here we show that PCs, but not B cells or T cells from lupus-prone NZM mice, are deficient in CXCL12-induced migration. PC unresponsiveness to CXCL12 results in a marked accumulation of PCs in the spleen of mice, and a concordant decrease in bone marrow PCs. Unlike normal mice, in NZM mice, a majority of the splenic PCs are long-lived. This deficiency is a consequence of the genetic interactions of multiple systemic lupus erythematosus susceptibility loci.


Subject(s)
Bone Marrow Cells/immunology , Chemokines, CXC/physiology , Plasma Cells/immunology , Receptors, Lymphocyte Homing/immunology , Adoptive Transfer , Animals , Chemokine CXCL12 , Chemokines, CXC/deficiency , Chemokines, CXC/genetics , Chemotaxis, Leukocyte , Disease Susceptibility/immunology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Plasma Cells/physiology , Spleen/immunology
19.
J Biol Chem ; 278(46): 45414-8, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-12960157

ABSTRACT

Tumor necrosis factor receptor-associated factors (TRAFs) belong to a family of adapter proteins that are involved in tumor necrosis factor receptor superfamily signaling. It has been shown that the recruitment of TRAFs to the CD40 cytoplasmic tail is essential for CD40-mediated B cell responses. However, it has also been shown that some early B cell responses, such as up-regulation of cell surface molecules and B cell proliferation are only marginally impaired by the disruption of previously defined TRAF binding sites (Ahonen, C., Manning, E., Erickson, L. D., O'Connor, B. P., Lind, E. F., Pullen, S. S., Kehry, M. R., and Noelle, R. J. (2002) Nat. Immunol. 3, 451-456; and Manning, E., Pullen, S. S., Souza, D. J., Kehry, M., and Noelle, R. J. (2002) Eur. J. Immunol. 32, 39-49). In this report, we identify a second TRAF2 binding site in the CD40 C terminus. The binding motif "SVQE" fits into the major TRAF2 binding consensus sequence, and its disruption resulted in the loss of remaining CD40 functions. Hence, like CD30, the CD40 cytoplasmic tail contains two distinct and functionally important TRAF2 binding sites.


Subject(s)
CD40 Antigens/metabolism , Proteins/metabolism , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , CD40 Ligand/biosynthesis , Cell Division , Cell Line , Cytoplasm/metabolism , DNA/metabolism , Ki-1 Antigen/biosynthesis , Mice , Models, Genetic , Molecular Sequence Data , Mutation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , TNF Receptor-Associated Factor 2 , Transfection , Two-Hybrid System Techniques , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL