Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1251127, 2023.
Article in English | MEDLINE | ID: mdl-37822931

ABSTRACT

Immunotherapeutic targeting of surface regulatory proteins and pharmacologic inhibition of critical signaling pathways has dramatically shifted our approach to the care of individuals with B cell malignancies. This evolution in therapy reflects the central role of the B cell receptor (BCR) signaling complex and its co-receptors in the pathogenesis of B lineage leukemias and lymphomas. Members of the Fc receptor-like gene family (FCRL1-6) encode cell surface receptors with complex tyrosine-based regulation that are preferentially expressed by B cells. Among them, FCRL1 expression peaks on naïve and memory B cells and is unique in terms of its intracellular co-activation potential. Recent studies in human and mouse models indicate that FCRL1 contributes to the formation of the BCR signalosome, modulates B cell signaling, and promotes humoral responses. Progress in understanding its regulatory properties, along with evidence for its over-expression by mature B cell leukemias and lymphomas, collectively imply important yet unmet opportunities for FCRL1 in B cell development and transformation. Here we review recent advances in FCRL1 biology and highlight its emerging significance as a promising biomarker and therapeutic target in B cell lymphoproliferative disorders.


Subject(s)
Lymphoma , Neoplasms , Animals , Mice , Humans , Neoplasms/metabolism , B-Lymphocytes/metabolism , Receptors, Fc/genetics , Receptors, Fc/metabolism , Receptors, Cell Surface/metabolism , Lymphoma/metabolism , Membrane Proteins/metabolism
2.
Oncol Res ; 32(1): 163-174, 2023.
Article in English | MEDLINE | ID: mdl-38188684

ABSTRACT

Hepatocellular carcinoma (HCC) is a leading cause of death worldwide. Current therapies are effective for HCC patients with early disease, but many patients suffer recurrence after surgery and have a poor response to chemotherapy. Therefore, new therapeutic targets are needed. We analyzed gene expression profiles between HCC tissues and normal adjacent tissues from public databases and found that the expression of genes involved in lipid metabolism was significantly different. The analysis showed that AKR1C3 was upregulated in tumors, and high AKR1C3 expression was associated with a poorer prognosis in HCC patients. In vitro, assays demonstrated that the knockdown of AKR1C3 or the addition of the AKR1C3 inhibitor indomethacin suppressed the growth and colony formation of HCC cell lines. Knockdown of AKR1C3 in Huh7 cells reduced tumor growth in vivo. To explore the mechanism, we performed pathway enrichment analysis, and the results linked the expression of AKR1C3 with prostaglandin F2 alpha (PGF2α) downstream target genes. Suppression of AKR1C3 activity reduced the production of PGF2α, and supplementation with PGF2α restored the growth of indomethacin-treated Huh7 cells. Knockdown of the PGF receptor (PTGFR) and treatment with a PTGFR inhibitor significantly reduced HCC growth. We showed that indomethacin potentiated the sensitivity of Huh7 cells to sorafenib. In summary, our results indicate that AKR1C3 upregulation may promote HCC growth by promoting the production of PGF2α, and suppression of PTGFR limited HCC growth. Therefore, targeting the AKR1C3-PGF2α-PTGFR axis may be a new strategy for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Aldo-Keto Reductases/genetics , Dinoprost , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Cell Line , Indomethacin/pharmacology , Aldo-Keto Reductase Family 1 Member C3
3.
Int J Mol Sci ; 22(6)2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33809908

ABSTRACT

Endoglin (CD105) is a type-1 integral transmembrane glycoprotein and coreceptor for transforming growth factor-ß (TGF-ß) ligands. The endoglin/TGF-ß signaling pathway regulates hemostasis, cell proliferation/migration, extracellular matrix (ECM) synthesis and angiogenesis. Angiogenesis contributes to early progression, invasion, postoperative recurrence, and metastasis in hepatocellular carcinoma (HCC), one of the most widespread malignancies globally. Endoglin is overexpressed in newly formed HCC microvessels. It increases microvessel density in cirrhotic and regenerative HCC nodules. In addition, circulating endoglin is present in HCC patients, suggesting potential for use as a diagnostic or prognostic factor. HCC angiogenesis is dynamic and endoglin expression varies by stage. TRC105 (carotuximab) is an antibody against endoglin, and three of its clinical trials were related to liver diseases. A partial response was achieved when combining TRC105 with sorafenib. Although antiangiogenic therapy still carries some risks, combination therapy with endoglin inhibitors or other targeted therapies holds promise.


Subject(s)
Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Disease Susceptibility , Endoglin/genetics , Endoglin/metabolism , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Animals , Biomarkers, Tumor , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/therapy , Endoglin/blood , Endoglin/chemistry , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Humans , Liver Neoplasms/pathology , Molecular Diagnostic Techniques , Molecular Targeted Therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Signal Transduction , Structure-Activity Relationship , Viral Core Proteins/metabolism
4.
Int J Mol Sci ; 21(18)2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32962123

ABSTRACT

Smoothened (SMO) belongs to the Hedgehog (HH) signaling pathway, which regulates cell growth, migration, invasion and stem cells in cancer. The HH signaling pathway includes both canonical and noncanonical pathways. The canonical HH pathway functions through major HH molecules such as HH ligands, PTCH, SMO and GLI, whereas the noncanonical HH pathway involves the activation of SMO or GLI through other pathways. The role of SMO has been discussed in different types of cancer, including breast, liver, pancreatic and colon cancers. SMO expression correlates with tumor size, invasiveness, metastasis and recurrence. In addition, SMO inhibitors can suppress cancer formation, reduce the proliferation of cancer cells, trigger apoptosis and suppress cancer stem cell activity. A better understanding of the role of SMO in cancer could contribute to the development of novel therapeutic approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/metabolism , Smoothened Receptor/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Female , Hedgehog Proteins/metabolism , Humans , Liver Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Signal Transduction/drug effects , Signal Transduction/genetics , Smoothened Receptor/metabolism
5.
Immunol Lett ; 226: 22-30, 2020 10.
Article in English | MEDLINE | ID: mdl-32622933

ABSTRACT

OBJECTIVE: MicroRNA-122 (miR-122) is the most abundant miRNA in the liver and it plays an important role in regulating liver metabolism and tumor formation. Previous studies also reveal an anti-inflammatory function of miR-122; however, relatively little is known about the mechanisms by which miR-122 suppresses inflammation. This study aims to search the effect of miR-122 on proinflammatory chemokines/cytokines production in mice. METHODS: Quantitative real-time PCR, Western blot analysis, and ELISA were performed to examine gene expression. TargetScan, miRanda, and microT v3.0 were used to search for possible miR-122 target sites in the 3'-untranslated regions (3'-UTR) of candidate genes. Luciferase reporter assay and site-directed mutagenesis were applied to verify miR-122 target sequences. LPS was applied to peritoneal macrophages and mice to evaluate inflammatory response. RESULTS: The expression of proinflammatory chemokines, including Ccl2, Ccl4, Ccl20, Cxcl2, and Cxcl10, and Relb in the livers of miR-122 knockout (KO) mice was increased. We identified Relb as a direct miR-122 target. Overexpressing RelB in the mouse liver increased the expression of Ccl2, Ccl4, Ccl20, Cxcl2, and Cxcl10. Peritoneal macrophages from miR-122 KO mice had a higher level of RelB, and they showed a stronger NF-κB activation and more TNF-α and IL-6 secretion after LPS stimulation. Overexpression of RelB in a macrophage cell line augmented LPS-induced TNF-α and IL-6 production. miR-122 KO mice showed a greatly increased mortality rate and generated a stronger and lasting inflammatory response to LPS. CONCLUSIONS: Deletion of miR-122 caused an upregulation of proinflammatory chemokines and RelB in the liver. Increased RelB may contribute to increases in these chemokine in the liver. Intriguingly, deletion of miR-122 also enhanced the sensitivity of macrophages and mice to LPS. Our results reveal that reducing RelB expression is a new mechanism by which miR-122 regulates inflammation.


Subject(s)
Liver/physiology , Macrophages/physiology , MicroRNAs/genetics , Transcription Factor RelB/metabolism , Animals , Chemokines/metabolism , Cytokines/metabolism , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Transcription Factor RelB/genetics , Up-Regulation
6.
Arthritis Res Ther ; 22(1): 64, 2020 03 30.
Article in English | MEDLINE | ID: mdl-32228715

ABSTRACT

BACKGROUND: Cytotoxic T lymphocyte-associated antigen-4-Ig (CTLA-4-Ig) competes with CD28 for binding CD80/CD86 on antigen-presenting cells (APCs) to limit T cell activation. B cells are believed to be important APCs in the pathogenesis of autoimmune diseases and express CD80/CD86 after activation; however, relatively little is known about the effect of CTLA-4-Ig on B cells. This study tested the impact of CTLA-4-Ig on human B cell responses. METHODS: Human blood B cells were purified from healthy donors and activated in the presence of CTLA-4-Ig or the L6-Ig control protein in vitro. RT-q-PCR and immunofluorescence staining were performed to detect activation marker expression. ELISA was conducted to measure cytokine secretion. The CD80/CD86 levels on the surface of the memory B cells in the blood of 18 patients with rheumatoid arthritis (RA) were detected using immunofluorescence staining. RESULTS: CTLA-4-Ig suppressed the expression of Staphylococcus aureus (SAC)-induced CD80, CD86, TNFA, and IL6 in human B cells at the transcriptional level. Furthermore, CTLA-4-Ig concomitantly decreased SAC-induced CD80/CD86 surface expression on and TNF-α and IL-6 secretion from B cells. On the other hand, T cell-dependent (TD) stimulation-induced B cell activation, proliferation, plasma cell differentiation, and antibody secretion were not affected by CTLA-4-Ig. As expected, TD stimulation-induced surface CD80 was hindered by CTLA-4-Ig. Notably, a blockade of CD80/CD86 on the surface of the memory B cells was observed in the patients with RA after abatacept (CTLA-4-Ig) treatment. In a portion of the RA patients, restoration of CD80/CD86 staining on the surface of the memory B was detected starting in the 3rd month of abatacept treatment. Interestingly, the surface levels of CD80/CD86 on the patients' memory B cells positively correlated with disease activity. CONCLUSIONS: We found that CTLA-4-Ig directly suppressed SAC-induced B cell activation in vitro. Obstruction of CD80 and CD86 on the surface of the memory B cells was detected in the RA patients after abatacept treatment. Blocking CD80/CD86 on B cells by CTLA-4-Ig may hinder T cell activation and associated with the disease activity of RA in vivo. Our findings indicate that CTLA-4-Ig may regulate humoral responses by modulating B cell activation and interfering T cell-B cell interaction.


Subject(s)
Abatacept/pharmacology , B-Lymphocytes/drug effects , B7-2 Antigen/metabolism , CD28 Antigens/metabolism , Cytokines/metabolism , T-Lymphocytes, Cytotoxic/drug effects , Abatacept/immunology , Abatacept/metabolism , Adult , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , B-Lymphocytes/metabolism , B-Lymphocytes/microbiology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , CD28 Antigens/genetics , CD28 Antigens/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression/drug effects , Gene Expression/immunology , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , Male , Middle Aged , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
7.
Oncol Lett ; 18(5): 4377-4384, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31611946

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-associated mortality worldwide. Hepatocarcinogenesis involves numerous interlinked factors and processes, including the Sonic hedgehog (Shh) signaling pathway, which participates in the carcinogenesis, progression, invasiveness, recurrence and cancer stem cell maintenance of HCC. The Shh signaling pathway is activated by ligands that bind to their receptor protein, Protein patched homolog (Ptch). The process of Shh ligand binding to Ptch weakens the inhibition of smoothened homolog (SMO) and activates signal transduction via glioma-associated oncogene homolog (Gli) transcription factors. The overexpression of Shh pathway molecules, including Shh, Ptch-1, Gli and SMO has been indicated in patients with HCC. It has also been suggested that the Shh signaling pathway exhibits cross-talk between numerous other signaling pathways. The inactivation of the Shh signaling pathway reduces HCC growth, increases radio-sensitivity and increases the beneficial effect of chemotherapy in HCC treatment. Therefore, inhibition of the Shh pathway may be an effective target therapy that can be used in the treatment of HCC.

8.
Biochem Pharmacol ; 163: 308-320, 2019 05.
Article in English | MEDLINE | ID: mdl-30822403

ABSTRACT

Chronic myelogenous leukemia (CML) is clinically treated with imatinib, which inhibits the kinase activity of the Bcr-Abl oncoprotein. However, imatinib resistance remains a common clinical issue. Andrographolide, the major compound of the medicinal plant Andrographis paniculata, was reported to exhibit anticancer activity. In this study, we explored the therapeutic potential of andrographolide and its derivative, NCTU-322, against both imatinib-sensitive and imatinib-resistant human CML cell lines. Both andrographolide and NCTU-322 downregulated the Bcr-Abl oncoprotein in imatinib-resistant CML cells through an Hsp90-dependent mechanism similar to that observed in imatinib-sensitive CML cells. In addition, NCTU-322 had stronger effects than andrographolide on downregulation of Bcr-Abl oncoprotein, induction of Hsp90 cleavage and cytotoxicity of CML cells. Notably, andrographolide and NCTU-322 could induce differentiation, mitotic arrest and apoptosis of both imatinib-sensitive and imatinib-resistant CML cells. Finally, the anticancer activity of NCTU-322 against imatinib-resistant CML cells was demonstrated in vivo. In summary, our data demonstrated that andrographolide and NCTU-322 inhibit Bcr-abl function via a mechanism different from that of imatinib, and they induced multiple anticancer effects in both imatinib-sensitive and resistant CML cells. Our findings demonstrate that andrographolide and NCTU-322 are potential therapeutic agents again CML.


Subject(s)
Antineoplastic Agents/pharmacology , Diterpenes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, abl/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/chemistry , Drug Resistance, Neoplasm , Genes, abl/genetics , Humans , Imatinib Mesylate/pharmacology , Leukocytes, Mononuclear/drug effects , Molecular Structure
10.
Rheumatology (Oxford) ; 58(4): 719-728, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30629240

ABSTRACT

OBJECTIVE: Deletion of Deltex1 (DTX1) in mice caused hyperactivation of T cells and lupus-like autoimmune syndromes, however, the association of DTX1 with human autoimmune diseases is totally unknown. This study investigated the role of DTX1 in human T cell functions and its correlation with disease activity in patients with SLE. METHODS: The influence of DTX1 on T cell function was evaluated using human primary cells. DTX1 expression in peripheral blood mononuclear cells (PBMCs) from healthy controls and SLE patients was measured by quantitative real-time PCR and the SLEDAI was used to assess disease activity. RESULTS: After stimulation with anti-CD3 and anti-CD28, silencing of DTX1 expression enhanced IFN-γ secretion by human T cells. The expression of DTX1 in PBMCs was significantly lower in 100 SLE patients than in 50 age- and sex-matched healthy controls (DTX1/glyceraldehyde 3-phosphate dehydrogenase, 0.452 vs 1.269, P < 0.001). The area under the receiver operator characteristics curve of the model was 0.737 (95% CI 0.658, 0.815). Intriguingly, a low DTX1 level in T cells led to high IFN-γ production in SLE patients and had a correlation with severe disease activity. In addition, low DTX1 expression in SLE patients was associated with active LN, lung involvement or hypocomplementaemia. CONCLUSION: Knockdown DTX1 expression in human T cells reduced IFN-γ secretion. DTX1 expression in the PBMCs was significantly lower in SLE patients and had an inverse correlation with disease activity, indicating that the DTX1 level may be a good disease marker of SLE.


Subject(s)
Lupus Erythematosus, Systemic/blood , T-Lymphocytes/metabolism , Ubiquitin-Protein Ligases/blood , Biomarkers/blood , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/metabolism
11.
Proc Natl Acad Sci U S A ; 115(34): E8027-E8036, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30087184

ABSTRACT

Activated T cells undergo metabolic reprogramming and effector-cell differentiation but the factors involved are unclear. Utilizing mice lacking DUSP6 (DUSP6-/-), we show that this phosphatase regulates T cell receptor (TCR) signaling to influence follicular helper T (TFH) cell differentiation and T cell metabolism. In vitro, DUSP6-/- CD4+ TFH cells produced elevated IL-21. In vivo, TFH cells were increased in DUSP6-/- mice and in transgenic OTII-DUSP6-/- mice at steady state. After immunization, DUSP6-/- and OTII-DUSP6-/- mice generated more TFH cells and produced more antigen-specific IgG2 than controls. Activated DUSP6-/- T cells showed enhanced JNK and p38 phosphorylation but impaired glycolysis. JNK or p38 inhibitors significantly reduced IL-21 production but did not restore glycolysis. TCR-stimulated DUSP6-/- T cells could not induce phosphofructokinase activity and relied on glucose-independent fueling of mitochondrial respiration. Upon CD28 costimulation, activated DUSP6-/- T cells did not undergo the metabolic commitment to glycolysis pathway to maintain viability. Unexpectedly, inhibition of fatty acid oxidation drastically lowered IL-21 production in DUSP6-/- TFH cells. Our findings suggest that DUSP6 connects TCR signaling to activation-induced metabolic commitment toward glycolysis and restrains TFH cell differentiation via inhibiting IL-21 production.


Subject(s)
Cell Differentiation/physiology , Dual Specificity Phosphatase 6 , Glycolysis/physiology , Receptors, Antigen, T-Cell , Signal Transduction/physiology , T-Lymphocytes, Helper-Inducer , Animals , Antibody Formation/physiology , CD28 Antigens/genetics , CD28 Antigens/immunology , CD28 Antigens/metabolism , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/immunology , Dual Specificity Phosphatase 6/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/immunology , Mitochondria/metabolism , Oxygen Consumption/physiology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Cell Rep ; 23(8): 2330-2341, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791845

ABSTRACT

T cells are a versatile immune cell population responding to challenges by differentiation and proliferation followed by contraction and memory formation. Dynamic metabolic reprogramming is essential for T cells to meet the biosynthetic needs and the reutilization of biomolecules, processes that require active participation of metabolite transporters. Here, we show that equilibrative nucleoside transporter 3 (ENT3) is highly expressed in peripheral T cells and has a key role in maintaining T cell homeostasis by supporting the proliferation and survival of T cells. ENT3 deficiency leads to an enlarged and disturbed lysosomal compartment, resulting in accumulation of surplus mitochondria, elevation of intracellular reactive oxygen species, and DNA damage in T cells. Our results identify ENT3 as a vital metabolite transporter that supports T cell homeostasis and activation by regulating lysosomal integrity and the availability of nucleosides. Moreover, we uncovered that T cell lysosomes are an important source of salvaged metabolites for survival and proliferation.


Subject(s)
Homeostasis , Lysosomes/metabolism , Nucleoside Transport Proteins/metabolism , Nucleosides/metabolism , T-Lymphocytes/metabolism , Animals , Cell Proliferation , Cell Size , Cell Survival , DNA/biosynthesis , DNA Repair , Lymphopenia/immunology , Lymphopenia/pathology , Lysosomes/ultrastructure , Mice , Mitochondria/metabolism , Phenotype , Reactive Oxygen Species/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/ultrastructure
13.
J Biomed Sci ; 21: 99, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25407417

ABSTRACT

BACKGROUND: Highly pathogenic influenza viruses cause high levels of morbidity, including excessive infiltration of leukocytes into the lungs, high viral loads and a cytokine storm. However, the details of how these pathological features unfold in severe influenza infections remain unclear. Accumulation of Gr1 + CD11b + myeloid cells has been observed in highly pathogenic influenza infections but it is not clear how and why they accumulate in the severely inflamed lung. In this study, we selected this cell population as a target to investigate the extreme inflammatory response during severe influenza infection. RESULTS: We established H1N1 IAV-infected mouse models using three viruses of varying pathogenicity and noted the accumulation of a defined Gr1 + CD11b + myeloid population correlating with the pathogenicity. Herein, we reported that CCR2+ inflammatory monocytes are the major cell compartments in this population. Of note, impaired clearance of the high pathogenicity virus prolonged IFN expression, leading to CCR2+ inflammatory monocytes amplifying their own recruitment via an interferon-α/ß receptor 1 (IFNAR1)-triggered chemokine loop. Blockage of IFNAR1-triggered signaling or inhibition of viral replication by Oseltamivir significantly suppresses the expression of CCR2 ligands and reduced the influx of CCR2+ inflammatory monocytes. Furthermore, trafficking of CCR2+ inflammatory monocytes from the bone marrow to the lung was evidenced by a CCR2-dependent chemotaxis. Importantly, leukocyte infiltration, cytokine storm and expression of iNOS were significantly reduced in CCR2-/- mice lacking infiltrating CCR2+ inflammatory monocytes, enhancing the survival of the infected mice. CONCLUSIONS: Our results indicated that uncontrolled viral replication leads to excessive production of inflammatory innate immune responses by accumulating CCR2+ inflammatory monocytes, which contribute to the fatal outcomes of high pathogenicity virus infections.


Subject(s)
Chemokines/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/physiopathology , Orthomyxoviridae Infections/physiopathology , Receptor, Interferon alpha-beta/genetics , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/virology , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Orthomyxoviridae Infections/virology , Oseltamivir/pharmacology , Receptor, Interferon alpha-beta/metabolism , Receptors, CCR2/metabolism , Severity of Illness Index , Specific Pathogen-Free Organisms , Virus Replication/drug effects
14.
Clin Exp Rheumatol ; 32(5): 715-23, 2014.
Article in English | MEDLINE | ID: mdl-25084482

ABSTRACT

OBJECTIVES: The decoy receptor 3 (DcR3) is a member of the tumour necrosis factor (TNF) receptor superfamily and may regulate inflammation. The aim of this study was to investigate the role of DcR3 in B cell functions and its correlation to disease activity in patients with rheumatoid arthritis (RA). METHODS: The concentrations of DcR3 and TNF-α were measured by ELISA. B cell proliferation was assessed by quantification of 3H-thymidine uptake. Staphylococcus aureus Cowan (SAC) strain were used to stimulate B cell proliferation and TNF-α production. RESULTS: Compared to the osteoarthritis (OA) patients, the RA group had higher synovial DcR3 levels (3273.6±1623.2 vs. 1594.8±1190.0 pg/ml, p=0.003), which were negatively correlated with the serum erythrocyte sedimentation rate and Disease Activity Score using 28 joint counts (DAS28) scores (r=-0.560, p=0.002; r=-0.579, p<0.001, respectively). Although the RA B cells have more active characteristics, B cell proliferation induced by SAC was successfully suppressed by recombinant DcR3.Fc fusion protein with an average inhibition of 44.8%. Moreover, DcR3.Fc fusion protein was found to suppress SAC-induced TNF-α production by B cells in 8 RA patients (average inhibition 47.0%). CONCLUSIONS: The results of our study indicated that the inhibition of B cell functions by DcR3 may partially explain the negative correlation between DcR3 level and disease activity in RA patients. Our findings imply that DcR3 may be used as a biomarker for disease activity and a potential therapeutic agent in the treatment of RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , B-Lymphocytes/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Aged , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , Biomarkers/metabolism , Case-Control Studies , Cell Proliferation , Cells, Cultured , Female , Humans , Lymphocyte Activation , Male , Middle Aged , Osteoarthritis, Knee/diagnosis , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/metabolism , Severity of Illness Index , Signal Transduction , Synovial Fluid/immunology , Synovial Fluid/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
15.
Immunobiology ; 219(11): 880-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25123430

ABSTRACT

The Hepatitis B virus (HBV) e antigen (HBeAg) is a secretory, non-structural protein, and associated with persistent infection of HBV. Previous studies indicate that HBeAg is able to regulate T cell-mediated responses, however, the interaction between HBeAg and the innate immune system is poorly understood. In this study, we demonstrated that recombinant HBeAg (rHBe) bound to human peripheral blood monocytes, neutrophils, and B lymphocytes but not to T lymphocytes. We focused on investigating the effects of HBeAg on monocytes and neutrophils and found that rHBe decreased the respiratory burst in both types of cells. Furthermore, we observed that cell migration in monocytes and neutrophils was suppressed by rHBe in a transwell assay. The attenuation of rHBe was not caused by a general cytotoxic effect because rHBe treatment stimulated low levels of cytokine and chemokine production by monocytes and it promoted neutrophil survival. Since the recruitment of monocytes and neutrophils to the infected site is crucial for the initiation of inflammation, HBeAg may modulate innate immune responses by diminishing the respiratory burst and migration of monocytes and neutrophils, which might interfere with the subsequent innate and adaptive immune responses against HBV, leading to the establishment of chronic infection.


Subject(s)
Chemotaxis, Leukocyte/immunology , Hepatitis B e Antigens/immunology , Monocytes/immunology , Neutrophils/immunology , Respiratory Burst/immunology , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chemokines/biosynthesis , Cytokines/biosynthesis , Hepatitis B e Antigens/metabolism , Humans , Monocytes/metabolism , Neutrophils/metabolism , Protein Binding , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
J Immunol ; 188(12): 5867-76, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22581854

ABSTRACT

Decoy receptor 3 (DcR3) is a soluble protein in the TNFR superfamily. Its known ligands include Fas ligand, homologous to lymphotoxin, showing inducible expression, and competing with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, TNF-like molecule 1A, and heparan sulfate proteoglycans. DcR3 has been reported to modulate the functions of T cells, dendritic cells, and macrophages; however, its role in regulating B cell activation is largely unknown. In this study, we found that the DcR3.Fc fusion protein bound to human and mouse B cells and suppressed the activation of B cells. DcR3.Fc attenuated Staphylococcus aureus, IgM-, Pam(3)CSK(4)-, and LPS-mediated B cell proliferation but did not affect cytokine-induced B cell growth. In the presence of these mitogens, DcR3.Fc did not induce B cell apoptosis, suggesting that DcR3 may inhibit the signal(s) important for B cell activation. Because the combination of Fas.Fc, LT-ßR.Fc (homologous to lymphotoxin, showing inducible expression, and competing with HSV glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes receptor), and DR3.Fc (TNF-like molecule 1A receptor) did not suppress B cell proliferation and because the biological effect of DcR3.Fc on B cells was not blocked by heparin, we hypothesize that a novel ligand(s) of DcR3 mediates its inhibitory activity on B cells. Moreover, we found that TLR2-stimulated NF-κB p65 activation and NF-κB-driven luciferase activity were attenuated by DcR3.Fc. The TLR2-induced cytokine production by B cells was consistently reduced by DcR3. These results imply that DcR3 may regulate B cell activation by suppressing the activation of NF-κB.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Activation/immunology , NF-kappa B/immunology , Receptors, Tumor Necrosis Factor, Member 6b/immunology , Toll-Like Receptor 2/immunology , Animals , Apoptosis/immunology , B-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Humans , Mice , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Toll-Like Receptor 2/metabolism
18.
Int Immunol ; 22(7): 583-92, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20497960

ABSTRACT

MicroRNAs 125a and 125b are predicted to be able to bind to the B lymphocyte-induced maturation protein-1 (BLIMP-1) and IFN regulatory protein-4 (IRF-4) transcription factors, which are essential for plasma cell differentiation. A computational survey of the human and mouse genomes revealed that miR-125a and miR-125b are members of a multigene family located in paralogous clusters. The miR-125a cluster on chromosome 19 in humans includes miR-99b and let-7e, whereas the miR-125b cluster on chromosome 21 includes miR-99a and miR-let-7c. Our analysis of the expression profiles for these six miRs during B lineage differentiation indicated that mature miR-125a, miR-125b, miR-99b and let-7e transcripts are preferentially expressed by the actively dividing centroblasts in germinal centers (GC). However, miR-99b and let-7e are not predicted to bind BLIMP-1 or IRF-4 transcripts, and binding to the untranslated region of BLIMP-1 and IRF-4 messenger RNAs could be confirmed only for miR-125b. When the effect of miR-125b over-expression on terminal B cell differentiation was evaluated in an LPS-responsive B cell line, the induction of BLIMP-1 expression and IgM secretion was inhibited in this model system. Furthermore, miR-125b over-expression inhibited the differentiation of primary B cells and compromised the survival of cultured myeloma cells. These findings suggest that miR-125b promotes B lymphocyte diversification in GC by inhibiting premature utilization of essential transcription factors for plasma cell differentiation.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Germinal Center/cytology , MicroRNAs/genetics , Animals , B-Lymphocytes/immunology , Base Sequence , Cell Line , Cluster Analysis , Conserved Sequence , Germinal Center/immunology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Mice , Models, Animal , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/genetics , Repressor Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/genetics
20.
J Exp Med ; 202(6): 783-91, 2005 Sep 19.
Article in English | MEDLINE | ID: mdl-16157685

ABSTRACT

The FcRH4 transmembrane molecule, a member of the Fc receptor homologue family, can potently inhibit B cell receptor (BCR) signaling. We show that cell surface expression of this immunoregulatory molecule is restricted to a subpopulation of memory B cells, most of which lack the classical CD27 marker for memory B cells in humans. The FcRH4+ and FcRH4- memory B cells have undergone comparable levels of immunoglobulin isotype switching and somatic hypermutation, while neither subpopulation expresses the transcription factors involved in plasma cell differentiation. The FcRH4+ memory cells are morphologically distinctive large lymphocytes that express the CD69, CD80, and CD86 cell activation markers. They are also shown to be poised to secrete high levels of immunoglobulins in response to stimulation with T cell cytokines, but they fail to proliferate in response either to BCR ligation or Staphylococcus aureus stimulation. A heightened expression of the CCR1 and CCR5 chemokine receptors may facilitate their preferential localization in lymphoid tissues near epithelial surfaces. Cell surface FcRH4 expression thus marks a unique population of memory B cells with distinctive morphology, functional capabilities, and tissue localization.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Memory , Receptors, Fc/biosynthesis , Receptors, Fc/genetics , Antibodies, Monoclonal/biosynthesis , B-Lymphocyte Subsets/metabolism , Cells, Cultured , Humans , Immunoglobulin Variable Region/genetics , Immunologic Memory/genetics , Lymphocyte Activation/immunology , Receptors, Cell Surface , Receptors, Chemokine/metabolism , Receptors, Fc/immunology , Somatic Hypermutation, Immunoglobulin
SELECTION OF CITATIONS
SEARCH DETAIL
...