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










Publication year range
1.
Immune Netw ; 23(5): e37, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37970231

ABSTRACT

Forkhead box P3-positive (Foxp3+)-inducible Tregs (iTregs) are readily generated by TGF-ß1 at low TCR signaling intensity. TGF-ß1-mediated Foxp3 expression is further enhanced by retinoic acid (RA) and lactoferrin (LF). However, the intensity of TCR signaling required for induction of Foxp3 expression by TGF-ß1 in combination with RA and LF is unknown. Here, we found that either RA or LF alone decreased TGF-ß1-mediated Foxp3 expression at low TCR signaling intensity. In contrast, at high TCR signaling intensity, the addition of either RA or LF strongly increased TGF-ß1-mediated Foxp3 expression. Moreover, decreased CD28 stimulation was more favorable for TGF-ß1/LF-mediated Foxp3 expression. Lastly, we found that at high signaling intensities of both TCR and CD28, combined treatment with TGF-ß1, RA, and LF induced robust expression of Foxp3, in parallel with powerful suppressive activity against responder T cell proliferation. Our findings that TGFß/RA/LF strongly generate high affinity Ag-specific iTreg population would be useful for the control of unwanted hypersensitive immune reactions such as various autoimmune diseases.

2.
Cytokine ; 172: 156413, 2023 12.
Article in English | MEDLINE | ID: mdl-37918054

ABSTRACT

Given the clinical success of cytokine blockade in managing diverse inflammatory human conditions, this approach could be exploited for numerous refractory or uncontrolled inflammatory conditions by identifying novel targets for functional blockade. Interleukin (IL)-18, a pro-inflammatory cytokine, is relatively underestimated as a therapeutic target, despite accumulated evidence indicating the unique roles of IL-18 in acute and chronic inflammatory conditions, such as macrophage activation syndrome. Herein, we designed a new form of IL-18 blockade, i.e., APB-R3, a long-acting recombinant human IL-18BP linked to human albumin-binding Fab fragment, SL335, for extending half-life. We then explored the pharmacokinetics and pharmacodynamics of APB-R3. In addition to an extended serum half-life, APB-R3 alleviates liver inflammation and splenomegaly in a model of the macrophage activation syndrome induced in IL-18BP knockout mice. Moreover, APB-R3 substantially controlled skin inflammation in a model of atopic dermatitis. Thus, we report APB-R3 as a new potent IL-18 blocking agent that could be applied to treat IL-18-mediated inflammatory diseases.


Subject(s)
Dermatitis, Atopic , Macrophage Activation Syndrome , Mice , Animals , Humans , Dermatitis, Atopic/drug therapy , Interleukin-18/therapeutic use , Serum Albumin, Human/therapeutic use , Macrophage Activation Syndrome/drug therapy , Cytokines/therapeutic use , Immunologic Factors/therapeutic use , Inflammation
3.
Immunology ; 168(1): 110-119, 2023 01.
Article in English | MEDLINE | ID: mdl-36054548

ABSTRACT

We recently reported that lactoferrin (LF) induces Foxp3+ Treg differentiation through binding to TGFß receptor III (TßRIII), and this activity was further enhanced by TGFß1. Generally, a low T-cell receptor (TCR) signal strength is favourable for Foxp3+ Treg differentiation. In the present study, we explored the effect of lactoferrin chimera (LFch, containing lactoferricin [aa 17-30] and lactoferrampin [aa 265-284]), along with TGFß1 on Foxp3+ Treg differentiation. LFch alone did not induce Foxp3 expression, yet LFch dramatically enhanced TGFß1-induced Foxp3 expression. LFch had little effect on the phosphorylation of Smad3, a canonical transcriptional factor of TGFß1. Instead, LFch attenuated the phosphorylation of S6 (a target of mTOR), IκB and PI3K. These activities of LFch were completely abrogated by pretreatment of LFch with soluble TGFß1 receptor III (sTßRIII). Consistent with this, the activity of LFch on TGFß1-induced Foxp3 expression was also abrogated by treatment with sTßRIII. Finally, the TGFß1/LFch-induced T cell population substantially suppressed the proliferation of responder CD4+ T cells. These results indicate that LFch robustly enhances TGFß1-induced Foxp3+ Treg differentiation by diminishing TCR/CD28 signal intensity.


Subject(s)
CD28 Antigens , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/metabolism , Lactoferrin/pharmacology , Lactoferrin/metabolism , Receptors, Antigen, T-Cell/metabolism , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
4.
J Immunol ; 207(10): 2456-2464, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34615735

ABSTRACT

Lactoferrin (LF) is known to possess anti-inflammatory activity, although its mechanisms of action are not well-understood. The present study asked whether LF affects the commitment of inducible regulatory T cells (Tregs). LF substantially promoted Foxp3 expression by mouse activated CD4+T cells, and this activity was further enhanced by TGF-ß1. Interestingly, blocking TGF-ß with anti-TGF-ß Ab completely abolished LF-induced Foxp3 expression. However, no significant amount of soluble TGF-ß was released by LF-stimulated T cells, suggesting that membrane TGF-ß (mTGF-ß) is associated. Subsequently, it was found that LF binds to TGF-ß receptor III, which induces reactive oxygen species production and diminishes the expression of mTGF-ß-bound latency-associated peptide, leading to the activation of mTGF-ß. It was followed by phosphorylation of Smad3 and enhanced Foxp3 expression. These results suggest that LF induces Foxp3+ Tregs through TGF-ß receptor III/reactive oxygen species-mediated mTGF-ß activation, triggering canonical Smad3-dependent signaling. Finally, we found that the suppressive activity of LF-induced Tregs is facilitated mainly by CD39/CD73-induced adenosine generation and that this suppressor activity alleviates inflammatory bowel disease.


Subject(s)
Lactoferrin/metabolism , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Colitis/immunology , Colitis/metabolism , Lactoferrin/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
5.
Immune Netw ; 20(5): e38, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33163246

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that initiate both T-cell responses and tolerance. Tolerogenic DCs (tDCs) are regulatory DCs that suppress immune responses through the induction of T-cell anergy and Tregs. Because lactoferrin (LF) was demonstrated to induce functional Tregs and has a protective effect against inflammatory bowel disease, we explored the tolerogenic effects of LF on mouse bone marrow-derived DCs (BMDCs). The expression of CD80/86 and MHC class II was diminished in LF-treated BMDCs (LF-BMDCs). LF facilitated BMDCs to suppress proliferation and elevate Foxp3+ induced Treg (iTreg) differentiation in ovalbumin-specific CD4+ T-cell culture. Foxp3 expression was further increased by blockade of the B7 molecule using CTLA4-Ig but was diminished by additional CD28 stimulation using anti-CD28 Ab. On the other hand, the levels of arginase-1 and indoleamine 2,3-dioxygenase-1 (known as key T-cell suppressive molecules) were increased in LF-BMDCs. Consistently, the suppressive activity of LF-BMDCs was partially restored by inhibitors of these molecules. Collectively, these results suggest that LF effectively causes DCs to be tolerogenic by both the suppression of T-cell proliferation and enhancement of iTreg differentiation. This tolerogenic effect of LF is due to the reduction of costimulatory molecules and enhancement of suppressive molecules.

6.
Immune Netw ; 18(4): e25, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30181913

ABSTRACT

γδ T cells are abundant in the gut mucosa and play an important role in adaptive immunity as well as innate immunity. Although γδ T cells are supposed to be associated with the enhancement of Ab production, the status of γδ T cells, particularly in the synthesis of IgA isotype, remains unclear. We compared Ig expression in T cell receptor delta chain deficient (TCRδ-/-) mice with wild-type mice. The amount of IgA in fecal pellets was substantially elevated in TCRδ-/- mice. This was paralleled by an increase in surface IgA expression and total IgA production by Peyer's patches (PPs) and mesenteric lymph node (MLN) cells. Likewise, the TCRδ-/- mice produced much higher levels of serum IgA isotype. Here, surface IgA expression and number of IgA secreting cells were also elevated in the culture of spleen and bone marrow (BM) B cells. Germ-line α transcript, an indicator of IgA class switch recombination, higher in PP and MLN B cells from TCRδ-/- mice, while it was not seen in inactivated B cells. Nevertheless, the frequency of IgA+ B cells was much higher in the spleen from TCRδ-/- mice. These results suggest that γδ T cells control the early phase of B cells, in order to prevent unnecessary IgA isotype switching. Furthermore, this regulatory role of γδ T cells had lasting effects on the long-lived IgA-producing plasma cells in the BM.

7.
Cell Immunol ; 322: 49-55, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29042055

ABSTRACT

The present study extends an earlier report that retinoic acid (RA) down-regulates IgE Ab synthesis in vitro. Here, we show the suppressive activity of RA on IgE production in vivo and its underlying mechanisms. We found that RA down-regulated IgE class switching recombination (CSR) mainly through RA receptor α (RARα). Additionally, RA inhibited histone acetylation of germ-line ε (GL ε) promoter, leading to suppression of IgE CSR. Consistently, serum IgE levels were substantially elevated in vitamin A-deficient (VAD) mice and this was more dramatic in VAD-lecithin:retinol acyltransferase deficient (LRAT-/-) mice. Further, serum mouse mast cell protease-1 (mMCP-1) level was elevated while frequency of intestinal regulatory T cells (Tregs) were diminished in VAD LRAT-/- mice, reflecting that deprivation of RA leads to allergic immune response. Taken together, our results reveal that RA has an IgE-repressive activity in vivo, which may ameliorate IgE-mediated allergic disease.


Subject(s)
Immunoglobulin Class Switching/drug effects , Immunoglobulin E/biosynthesis , Interleukin-4/metabolism , Tretinoin/pharmacology , Vitamin A Deficiency/blood , Acyltransferases/deficiency , Acyltransferases/genetics , Animals , Chymases/metabolism , Food Hypersensitivity/drug therapy , Food Hypersensitivity/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin E/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Retinoic Acid Receptor alpha/immunology , T-Lymphocytes, Regulatory/immunology , Vitamin A/genetics , Vitamin A Deficiency/genetics
8.
Article in English | MEDLINE | ID: mdl-28752079

ABSTRACT

There is a substantial need for biomarkers to distinguish latent stage from active Mycobacterium tuberculosis infections, for predicting disease progression. To induce the reactivation of tuberculosis, we present a new experimental animal model modified based on the previous model established by our group. In the new model, the reactivation of tuberculosis is induced without administration of immunosuppressive agents, which might disturb immune responses. To identify the immunological status of the persistent and chronic stages, we analyzed immunological genes in lung tissues from mice infected with M. tuberculosis. Gene expression was screened using cDNA microarray analysis and confirmed by quantitative RT-PCR. Based on the cDNA microarray results, 11 candidate cytokines genes, which were obviously up-regulated during the chronic stage compared with those during the persistent stage, were selected and clustered into three groups: (1) chemokine genes, except those of monocyte chemoattractant proteins (MCPs; CXCL9, CXCL10, CXCL11, CCL5, CCL19); (2) MCP genes (CCL2, CCL7, CCL8, CCL12); and (3) TNF and IFN-γ genes. Results from the cDNA microarray and quantitative RT-PCR analyses revealed that the mRNA expression of the selected cytokine genes was significantly higher in lung tissues of the chronic stage than of the persistent stage. Three chemokines (CCL5, CCL19, and CXCL9) and three MCPs (CCL7, CCL2, and CCL12) were noticeably increased in the chronic stage compared with the persistent stage by cDNA microarray (p < 0.01, except CCL12) or RT-PCR (p < 0.01). Therefore, these six significantly increased cytokines in lung tissue from the mouse tuberculosis model might be candidates for biomarkers to distinguish the two disease stages. This information can be combined with already reported potential biomarkers to construct a network of more efficient tuberculosis markers.


Subject(s)
Chemokines/genetics , Latent Tuberculosis/microbiology , Lung/immunology , Mycobacterium tuberculosis/physiology , Tuberculosis/drug therapy , Tuberculosis/genetics , Animals , Antitubercular Agents/administration & dosage , Chemokines/immunology , Chronic Disease , Disease Models, Animal , Gene Expression Profiling , Humans , Latent Tuberculosis/genetics , Latent Tuberculosis/immunology , Lung/microbiology , Mice , Tuberculosis/immunology , Tuberculosis/microbiology
9.
Cell Mol Immunol ; 13(6): 862-870, 2016 11.
Article in English | MEDLINE | ID: mdl-26277894

ABSTRACT

Lactoferrin (LF) and retinoic acid (RA) are enriched in colostrum, milk, and mucosal tissues. We recently showed that LF-induced IgA class switching through binding to betaglycan (transforming growth factor-beta receptor III, TßRIII) and activation of canonical TGF-ß signaling. We investigated the combined effect of LF and RA on the overall IgA response. An increase in IgA production by LF was further augmented by RA. This combination effect was also evident in Ig germ-line α (GLα) transcription and GLα promoter activity, indicating that LF in cooperation with RA increased IgA isotype switching. We subsequently found that RA enhanced TßRIII expression and that this increase contributed to LF-stimulated IgA production. In addition to the IgA response, LF and RA in combination also enhanced the expression of the gut-homing molecules C-C chemokine receptor 9 (CCR9) and α4ß7 on B cells. Finally, peroral administration of LF and RA enhanced the frequency of CCR9+IgA+ plasma cells in the lamina propria. Taken together, these results suggest that LF in cooperation with RA can contribute to the establishment of gut IgA responses.


Subject(s)
Immunoglobulin A/metabolism , Lactoferrin/pharmacology , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Tretinoin/pharmacology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cattle , Cell Proliferation/drug effects , Humans , Immunoglobulin Class Switching/genetics , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
BMB Rep ; 48(7): 419-25, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25936779

ABSTRACT

Ginseng has been widely used for therapeutic and preventive purposes for thousands of years. However, orally administered ginseng has very low bioavailability and absorption in the intestine. Therefore, fermented ginseng was developed to enhance the beneficial effects of ginseng in the intestine. In this study, we investigated the molecular mechanisms underlying the anti-inflammatory activity of fermented wild ginseng (FWG). We found that FWG significantly alleviated the severity of colitis in a dextran sodium sulfate (DSS)-induced colitis mouse model, and decreased expression level of pro-inflammatory cytokines in colonic tissue. Moreover, we observed that FWG suppressed the infiltration of macrophages in DSS-induced colitis. FWG also attenuated the transcriptional activity of nuclear factor-κB (NF-κB) by reducing the translocation of NF-κB into the nucleus. Our data indicate that FWG contains anti-inflammatory activity via NF-κB inactivation and could be useful for treating colitis.


Subject(s)
Colitis/chemically induced , Colitis/drug therapy , Intestinal Mucosa/pathology , NF-kappa B/metabolism , Panax/chemistry , Plant Extracts/therapeutic use , Signal Transduction , Acute Disease , Administration, Oral , Animals , Colitis/pathology , Colon/pathology , Cytokines/biosynthesis , Dextran Sulfate , Female , Fermentation , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Inflammation/pathology , Intestinal Mucosa/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Plant Extracts/pharmacology , RAW 264.7 Cells , Signal Transduction/drug effects , Zonula Occludens-1 Protein/metabolism
11.
Immune Netw ; 15(1): 37-43, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25713507

ABSTRACT

It is well established that TGF-ß1 and retinoic acid (RA) cause IgA isotype switching in mice. We recently found that lactoferrin (LF) also has an activity of IgA isotype switching in spleen B cells. The present study explored the effect of LF on the Ig production by mouse peritoneal B cells. LF, like TGF-ß1, substantially increased IgA production in peritoneal B1 cells but little in peritoneal B2 cells. In contrast, LF increased IgG2b production in peritoneal B2 cells much more strongly than in peritoneal B1 cells. LF in combination with RA further enhanced the IgA production and, interestingly, this enhancement was restricted to IgA isotype and B1 cells. Similarly, the combination of the two molecules also led to expression of gut homing molecules α4ß7 and CCR9 on peritoneal B1 cells, but not on peritoneal B2 cells. Thus, these results indicate that LF and RA can contribute to gut IgA response through stimulating IgA isotype switching and expression of gut-homing molecules in peritoneal B1 cells.

12.
Immune Netw ; 14(5): 237-40, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25360074

ABSTRACT

Endoglin (also known as CD105 or TGF-ß type III receptor) is a co-receptor involved in TGF-ß signaling. In atherosclerosis, TGF-ß signaling is crucial in regulating disease progression owing to its anti-inflammatory effects as well as its inhibitory effects on smooth muscle cell proliferation and migration. Endoglin is a regulator of TGF-ß signaling, but its role in atherosclerosis has yet to be defined. This review focuses on the roles of the various forms of endoglin in atherosclerosis. The expression of the two isoforms of endoglin (long-form and short-form) is increased in atherosclerotic lesions, and the expression of the soluble forms of endoglin is upregulated in sera of patients with hypercholesterolemia and atherosclerosis. Interestingly, long-form endoglin shows an atheroprotective effect via the induction of eNOS expression, while short-form and soluble endoglin enhance atherogenesis by inhibiting eNOS expression and TGF-ß signaling. This review summarizes evidence suggesting that the different forms of endoglin have distinct roles in atherosclerosis.

13.
Hum Immunol ; 75(8): 923-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24994461

ABSTRACT

Retinoic acid (RA) is known to have several functions that lead to a potent mucosal IgA response. Nevertheless, its exact role in human IgA synthesis has yet to be elucidated. Thus, we investigated the role of RA in promoting IgA isotype switching in human B cells. We found that RA increased IgA production and the expression of germ-line IgA1 and IgA2 transcripts (GLTα1 and GLTα2). This induction occurred alongside an increase in the frequency of IgA1-secreting B cell clones, as assessed by limiting dilution analysis. Under the same conditions, RA did not increase IgM and IgG production. Am80, an agonist of RA receptor α (RARα), increased IgA production. In addition, RA activity was abrogated by LE540, an antagonist of RAR, suggesting that the RAR pathway is involved in RA-induced IgA production. Taken together, these results indicate that RA induces IgA isotype switching mainly through RARα in human B cells.


Subject(s)
B-Lymphocytes/drug effects , Immunoglobulin A/genetics , Immunoglobulin Class Switching/drug effects , Tretinoin/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Benzoates/pharmacology , Dibenzazepines/pharmacology , Gene Expression Regulation , Humans , Immunoglobulin A/immunology , Immunoglobulin G , Immunoglobulin M , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/immunology , Retinoic Acid Receptor alpha , Signal Transduction , Tetrahydronaphthalenes/pharmacology , Transforming Growth Factor beta1/pharmacology
14.
J Leukoc Biol ; 94(2): 325-35, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23744644

ABSTRACT

The present study demonstrates that RA has activity of an IgA switch factor and is more specific than TGF-ß1. RA independently caused only IgA switching, whereas TGF-ß1 caused IgA and IgG2b switching. We found that RA increased IgA production and that this was a result of its ability to increase the frequency of IgA-secreting B cell clones. Increased IgA production was accompanied by an increase of GLTα. RA activity was abrogated by an antagonist of the RAR. Additionally, RA affected intestinal IgA production in mice. Surprisingly, RA, in combination with TGF-ß1, notably enhanced not only IgA production and GLTα expression but also CCR9 and α4ß7 expression on B cells. These results suggest that RA selectively induces IgA isotype switching through RAR and that RA and TGF-ß have important effects on the overall gut IgA antibody response.


Subject(s)
Immunoglobulin A/immunology , Immunoglobulin Class Switching/drug effects , Transforming Growth Factor beta1/immunology , Tretinoin/pharmacology , Animals , Cells, Cultured/immunology , Clonal Selection, Antigen-Mediated , Endotoxins/toxicity , Genes, Immunoglobulin , Immunity, Mucosal/drug effects , Immunoglobulin A/biosynthesis , Immunoglobulin G/immunology , Integrins/biosynthesis , Integrins/genetics , Lymph Nodes/immunology , Mesentery/immunology , Mice , Mice, Inbred BALB C , Peyer's Patches/immunology , Receptors, CCR/biosynthesis , Receptors, CCR/genetics , Receptors, Lymphocyte Homing/drug effects , Receptors, Lymphocyte Homing/immunology , Receptors, Retinoic Acid/physiology
15.
Immune Netw ; 13(1): 10-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23559895

ABSTRACT

Aluminum hydroxide (alum) is the most widely used adjuvant in human vaccines. Nevertheless, it is virtually unknown whether alum acts on B cells. In the present study, we explored the direct effect of alum on Ig expression by murine B cells in vitro. LPS-activated mouse spleen B cells were cultured with alum, and the level of isotype-specific Ig secretion, IgG1 secreting cell numbers, and Ig germ-line transcripts (GLT) were measured using ELISA, ELISPOT, and RT-PCR, respectively. Alum consistently enhanced total IgG1 production, numbers of IgG1 secreting cells, and GLTγ1 expression. These results demonstrate that alum can directly cause IgG1 isotype switching leading to IgG1 production.

16.
Immune Netw ; 12(3): 84-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22916043

ABSTRACT

B cell-activating factor belonging to the TNF family (BAFF) is primarily expressed by macrophages and stimulates B cell proliferation, differentiation, survival, and Ig production. In this study, we explored the effect of lactoferrin (LF) on BAFF expression by murine macrophages. We determined the level of BAFF expression at the transcriptional and protein levels using RT-PCR and ELISA, respectively. LF markedly enhanced BAFF expression in mouse macrophages at both the transcriptional and protein levels. Overexpression of Smad3/4 further increased LF-induced BAFF transcription while DN-Smad3 abolished the LF-induced BAFF expression. These results demonstrate that LF can enhance BAFF expression through Smad3/4 pathway.

17.
Mol Cells ; 32(3): 251-5, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21710205

ABSTRACT

A proliferation-inducing ligand (APRIL), a new TNF family member, supports B-cell survival and tumor cell proliferation. APRIL is secreted as a soluble protein by macrophages, dendritic cells and activated T cells. However, factors involved in regulation of APRIL expression are as yet unknown. In this study, we investigated the effect of TGF-ß1 on APRIL expression in P388D1, a mouse macrophage cell line. TGF-ß1 induced APRIL mRNA expression in a time- and dose-dependent manner. One nanogram per milliliter of TGF-ß1 was optimal and APRIL transcripts appeared as early as 3 h after stimulation. Based on our studies, which included overexpression of Smad3, DN-Smad3, and sh-Smad3, we found that Smad3 mediates APRIL transcription at least partially. Further, experiments using inhibitors revealed that p38MAPK and CREB are also involved in TGF-ß1-induced APRIL expression. These results suggest that TGF-ß1, through Smad3 and p38MAPK/CREB signaling pathways, stimulates APRIL expression in macrophages.


Subject(s)
Cell Proliferation/drug effects , Macrophages/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Macrophages/cytology , Macrophages/metabolism , Mice , Plasmids , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transfection , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
18.
Cytokine ; 47(1): 43-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19427796

ABSTRACT

APRIL (a proliferation-inducing ligand) is primarily expressed by macrophages and dendritic cells, and has profound effects on B cell physiology. In this study, we investigated the role of IL-4 in APRIL expression by mouse macrophages and the signaling mechanism involved. IL-4 markedly enhanced APRIL expression in mouse macrophages at the transcriptional and protein level. The p38MAPK inhibitor SB203580 completely abolished the IL-4 effect, whereas overexpression of CREB with IL-4 augmented APRIL expression. This increase was abolished by SB203580 treatment, indicating that p38MAPK may activate CREB. Overexpression of Stat6 also augmented IL-4-induced APRIL expression; this effect was partially abolished by SB203580 but not by the Jak inhibitor AG490, indicating that Stat6 mediates IL-4-induced APRIL expression in a Jak-independent manner and that p38MAPK acts as the intermediate. Our results demonstrate that IL-4 up-regulates APRIL expression through two divergent pathways in mouse macrophages, p38MAPK-CREB and p38MAPK-Stat6.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Interleukin-4/pharmacology , Macrophages/metabolism , STAT6 Transcription Factor/genetics , Signal Transduction/physiology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Gene Expression/drug effects , Imidazoles/pharmacology , Macrophages/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Transfection , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...