Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Biol Sci ; 20(7): 2592-2606, 2024.
Article in English | MEDLINE | ID: mdl-38725855

ABSTRACT

Transcriptional coactivator with a PDZ-binding motif (TAZ) plays a key role in normal tissue homeostasis and tumorigenesis through interaction with several transcription factors. In particular, TAZ deficiency causes abnormal alveolarization and emphysema, and persistent TAZ overexpression contributes to lung cancer and pulmonary fibrosis, suggesting the possibility of a complex mechanism of TAZ function. Recent studies suggest that nuclear factor erythroid 2-related factor 2 (NRF2), an antioxidant defense system, induces TAZ expression during tumorigenesis and that TAZ also activates the NRF2-mediated antioxidant pathway. We thus thought to elucidate the cross-regulation of TAZ and NRF2 and the underlying molecular mechanisms and functions. TAZ directly interacted with NRF2 through the N-terminal domain and suppressed the transcriptional activity of NRF2 by preventing NRF2 from binding to DNA. In addition, the return of NRF2 to basal levels after signaling was inhibited in TAZ deficiency, resulting in sustained nuclear NRF2 levels and aberrantly increased expression of NRF2 targets. TAZ deficiency failed to modulate optimal NRF2 signaling and concomitantly impaired lysosomal acidification and lysosomal enzyme function, accumulating the abnormal autophagy vesicles and reactive oxygen species and causing protein oxidation and cellular damage in the lungs. TAZ restoration to TAZ deficiency normalized dysregulated NRF2 signaling and aberrant lysosomal function and triggered the normal autophagy-lysosomal pathway. Therefore, TAZ is indispensable for the optimal regulation of NRF2-mediated autophagy-lysosomal pathways and for preventing pulmonary damage caused by oxidative stress and oxidized proteins.


Subject(s)
Autophagy , Lysosomes , NF-E2-Related Factor 2 , NF-E2-Related Factor 2/metabolism , Autophagy/physiology , Lysosomes/metabolism , Animals , Mice , Humans , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing
2.
Front Pharmacol ; 14: 1203033, 2023.
Article in English | MEDLINE | ID: mdl-37469871

ABSTRACT

Background: Pulmonary fibrosis (PF) is a progressive lung disease characterized by fibroblast accumulation and collagen deposition, resulting in lung scarring and impaired gas exchange. Current treatments for idiopathic pulmonary fibrosis (IPF) have limited efficacy and significant side effects. Heat shock protein 27 (HSP27) has emerged as a potential therapeutic target for PF due to its involvement in fibrotic processes. However, effective HSP27 inhibitors for PF treatment are still lacking. Methods: To assess the anti-fibrotic effects of NA49, we utilized murine PF models induced by radiation (IR) or bleomycin (BLM). We administered NA49 to the PF mice and evaluated its impact on lung fibrosis progression. We also investigated the molecular mechanisms underlying NA49's effects, focusing on its inhibition of EMT-related signaling pathways. Results: In our study, we evaluated the potential of a novel HSP27 inhibitor, NA49, in preclinical models of PF. NA49 effectively suppressed PF development in radiation and bleomycin-induced PF models. It reduced fibrosis, inhibited NFkB signaling, and downregulated EMT-related molecules. Importantly, we evaluated the safety profile of NA49 by assessing its impact on DNA strand breakage. Compared to previous HSP27 inhibitors, NA49 showed lower levels of DNA damage in human lung epithelial cells, and suggests that NA49 may have reduced toxicity compared to other HSP27 inhibitors. Overall, our results demonstrate that NA49 effectively inhibits PF development in preclinical models. It reduces lung fibrosis, inhibits EMT-related signaling pathways, and exhibits improved safety profiles. These findings highlight the potential of NA49 as a promising candidate for the treatment of PF. Conclusion: NA49 exhibited significant anti-fibrotic effects, inhibiting fibrosis development and EMT-related signaling pathways. Moreover, NA49 showed improved safety profiles compared to previous HSP27 inhibitors.

3.
Cell Mol Life Sci ; 79(3): 186, 2022 Mar 13.
Article in English | MEDLINE | ID: mdl-35279781

ABSTRACT

Transcriptional co-activator with PDZ-binding motif (TAZ) is a key mediator of the Hippo signaling pathway and regulates structural and functional homeostasis in various tissues. TAZ activation is associated with the development of pancreatic cancer in humans, but it is unclear whether TAZ directly affects the structure and function of the pancreas. So we sought to identify the TAZ function in the normal pancreas. TAZ defect caused structural changes in the pancreas, particularly islet cell shrinkage and decreased insulin production and ß-cell markers expression, leading to hyperglycemia. Interestingly, TAZ physically interacted with the pancreatic and duodenal homeobox 1 (PDX1), a key insulin transcription factor, through the N-terminal domain of TAZ and the homeodomain of PDX1. TAZ deficiency decreased the DNA-binding and transcriptional activity of PDX1, whereas TAZ overexpression promoted PDX1 activity and increased insulin production even in a low glucose environment. Indeed, high glucose increased insulin production by turning off the Hippo pathway and inducing TAZ activation in pancreatic ß-cells. Ectopic TAZ overexpression along with PDX1 activation was sufficient to produce insulin in non-ß-cells. TAZ deficiency impaired the mesenchymal stem cell differentiation into insulin-producing cells (IPCs), whereas TAZ recovery restored normal IPCs differentiation. Compared to WT control, body weight increased in TAZ-deficient mice with age and even more with a high-fat diet (HFD). TAZ deficiency significantly exacerbated HFD-induced glucose intolerance and insulin resistance. Therefore, TAZ deficiency impaired pancreatic insulin production, causing hyperglycemia and exacerbating HFD-induced insulin resistance, indicating that TAZ may have a beneficial effect in treating insulin deficiency in diabetes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Homeodomain Proteins/metabolism , Insulin/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation , Cell Line , Diet, High-Fat , Glucose/pharmacology , Hippo Signaling Pathway/drug effects , Homeodomain Proteins/genetics , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperglycemia/veterinary , Insulin/genetics , Insulin Resistance , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Trans-Activators/genetics , Transcriptional Activation
4.
FASEB J ; 34(4): 5332-5347, 2020 04.
Article in English | MEDLINE | ID: mdl-32067268

ABSTRACT

Transcriptional coactivator with PDZ-binding motif (TAZ) plays crucial role in maintaining testicular structure and function via regulation of senescence of spermatogenic cells. However, it remains unclear whether TAZ is involved in testosterone biosynthesis in testicular Leydig cells. We found that TAZ deficiency caused aberrant Leydig cell expansion and increased lipid droplet formation, which was significantly associated with increased lipogenic enzyme expression. Additionally, the expression of key steroidogenic enzymes, including steroidogenic acute regulatory protein, cytochrome P450 (CYP) 11A1, CYP17A1, and 3ß-hydroxysteroid dehydrogenase, was greatly increased in TAZ-deficient testes and primary Leydig cells. Interestingly, the transcriptional activity of nuclear receptor 4 A1 (NR4A1) was dramatically suppressed by TAZ; however, the protein expression and the subcellular localization of NR4A1 were not affected by TAZ. TAZ directly associated with the N-terminal region of NR4A1 and substantially suppressed its DNA-binding and transcriptional activities. Stable expression of TAZ in the mouse Leydig TM3 cell line decreased the expression of key steroidogenic enzymes, whereas knockdown of endogenous TAZ in TM3 cells increased transcripts of steroidogenic genes induced by NR4A1. Consistently, testosterone production was enhanced within TAZ-deficient Leydig cells. However, TAZ deficiency resulted in decreased testosterone secretion caused by dysfunctional mitochondria and lysosomes. Therefore, TAZ plays essential role in NR4A1-induced steroidogenic enzyme expression and testosterone production in Leydig cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Leydig Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphoproteins/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Testosterone/metabolism , Trans-Activators/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
5.
Clin Cancer Res ; 25(17): 5364-5375, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31126962

ABSTRACT

PURPOSE: Lung fibrosis is a major side effect experienced by patients after lung cancer radiotherapy. However, effective protection strategies and underlying treatment targets remain unclear. In an effort to improve clinical outcomes, pharmacologic treatment of fibrosis is becoming increasingly popular; however, no ideal therapeutic strategy is yet available. EXPERIMENTAL DESIGN: We used a mouse model to irradiate high focal (90 or 75 Gy) to 3-mm volume of the left lung. Lung tissues of mice were subjected to microarray, mRNA expression, and immunohistochemical analysis. Correlations of radiation (IR)-induced epithelial-mesenchymal transition (EMT) were validated in lung cell lines using appropriate treatments to activate or inhibit selected pathways. RESULTS: The expression of Hsp27 was increased during IR-induced lung fibrosis in a mouse model. Inhibition of functional Hsp27 using shRNA and a synthetic small molecule inhibitor (J2) in lung cells alleviated IR-mediated EMT. The activation of NFkB pathways via direct interaction between Hsp27 and IkBα resulted in increased expressions of Twist, IL-1ß, and IL-6 and facilitated IR-mediated EMT, which was identified as an underlying mechanism of Hsp27-mediated fibrosis after IR. J2 also inhibited IR-induced lung fibrosis in an orthotopic lung cancer model, and IR-induced lung fibrotic tissues from patients showed higher expression of Hsp27 than unirradiated lungs. CONCLUSIONS: Collectively, IkBα-NFkB signaling activation by Hsp27, which resulted in the facilitation of Twist, IL1ß, and IL6 expression, is involved in the EMT process that is tightly connected to the development of IR-induced lung fibrosis. Our findings also suggest that inhibition of Hsp27 has the potential to become a valuable therapeutic strategy for IR-induced lung fibrosis.


Subject(s)
Heat-Shock Proteins/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Pulmonary Fibrosis/pathology , Radiation Injuries/pathology , Animals , Disease Models, Animal , Epithelial-Mesenchymal Transition , Gamma Rays , Gene Expression Profiling , HSP27 Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Radiation Injuries/etiology , Radiation Injuries/metabolism , Signal Transduction , Tumor Cells, Cultured
6.
Sci Rep ; 7(1): 16946, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29208963

ABSTRACT

CD4+ T cells play key roles in the regulation of immune responses against pathogenic infectious antigens via development into effector T helper and induced regulatory T (iTreg) cells. Particularly, CD4+CD25+Foxp3+ iTreg cells are crucial for maintaining immune homeostasis and controlling inflammatory diseases. Anti-inflammatory drugs that enhance iTreg cell generation would be effective at preventing and treating inflammatory and autoimmune diseases. In this study, we examined whether anti-malarial and anti-arthritic amodiaquine (AQ) could affect iTreg cell development. Despite the anti-proliferative activity of AQ, AQ only moderately decreased iTreg cell proliferation but substantially increased IL-2 production by iTreg cells. Furthermore, AQ dose-dependently increased iTreg cell development and significantly upregulated iTreg cell markers including CD25. Interestingly, CD25 expression was decreased at later stages of iTreg cell development but was sustained in the presence of AQ, which was independent of IL-2 signaling pathway. AQ directly increased CD25 gene transcription by enhancing the DNA-binding and transcriptional activity of nuclear receptor 4 A. Most importantly, in vivo administration of AQ attenuated inflammatory colitis, resulted in the increased iTreg cells and decreased inflammatory cytokines. The ability of anti-malarial AQ to potentiate iTreg cell development makes it a promising drug for preventing and treating inflammatory and autoimmune diseases.


Subject(s)
Amodiaquine/pharmacology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology , Animals , Cell Proliferation/drug effects , Colitis/drug therapy , Colitis/etiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, Interleukin-2/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Transforming Growth Factor beta/pharmacology
7.
Sci Rep ; 7(1): 8455, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814721

ABSTRACT

Osteoblast differentiation is regulated through the successive activation of signaling molecules by a complex interplay of extracellular signals such as bone morphogenetic protein (BMP) and Wnt ligands. Numerous studies have identified natural as well as synthetic compounds with osteogenic activity through the regulation of either BMP/SMADs or the Wnt/ß-catenin pathway. Here, we attempted to isolate small molecules that concurrently activated both SMADs and ß-catenin, which led to the discovery of a novel potent osteogenic compound, DMP-PYT. Upon BMP2 stimulation, DMP-PYT substantially increased osteoblast differentiation featured by enhanced expression of osteoblast-specific genes and accelerated calcification through activation of BMPs expression. DMP-PYT promoted BMP2-induced SMAD1/5/8 phosphorylation and ß-catenin expression, the latter in a BMP2-independent manner. DMP-PYT alone enhanced nuclear localization of ß-catenin to promote the DNA-binding and transcriptional activity of T-cell factor, thereby resulting in increased osteoblast differentiation in the absence of BMP2. Most importantly, DMP-PYT advanced skeletal development and bone calcification in zebrafish larvae. Conclusively, DMP-PYT strongly stimulated osteoblast differentiation and bone formation in vitro and in vivo by potentiating BMP2-induced activation of SMADs and ß-catenin. These results suggest that DMP-PYT may have beneficial effects for preventing and for treating osteoporosis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Wnt Proteins/metabolism , Animals , Animals, Genetically Modified , Bone Morphogenetic Proteins/genetics , Cell Differentiation/genetics , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression/drug effects , Larva/genetics , Larva/metabolism , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Molecular Structure , Osteoblasts/cytology , Osteogenesis/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Small Molecule Libraries/chemistry , Wnt Proteins/genetics , Zebrafish/genetics , Zebrafish/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Red Fluorescent Protein
8.
Bioorg Med Chem ; 25(12): 3127-3134, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28442260

ABSTRACT

A growing number of studies have demonstrated that interleukin (IL)-6 plays pathological roles in the development of chronic inflammatory disease and autoimmune disease by activating innate immune cells and by stimulating adaptive inflammatory T cells. So, suppression of IL-6 function may be beneficial for prevention and treatment of chronic inflammatory disease. This study reports that a series of synthetic derivatives of benzoxazole have suppressive effects on IL-6-mediated signaling. Among 16 synthetic derivatives of benzoxazole, the compounds 4, 6, 11, 15, 17, and 19 showed a strong suppressive activity against IL-6-induced phosphorylation of signal transducer and activator of transcription (STAT) 3 by 80-90%. While the cell viability was strongly decreased by compounds 11, 17, 19, the compounds 4, 6, and 15 revealed less cytotoxicity. We then examined the effects of the compounds on inflammatory cytokine production by CD4+ T cells. CD4+ T cells were induced to differentiate into interferon (IFN)-γ-, IL-17-, or IL-4-producing effector T cells in the presence of either the compound 4 or the compound 7. While the inactive compound 7 had no significant effect on the cytokine production by effector T cells, the active compound 4 strongly suppressed the production of inflammatory cytokines IFN-γ and IL-17, and also inhibited allergic inflammatory cytokines IL-4, IL-5, and IL-13 produced by effector Th2 cells. These results suggest that a benzoxazole derivative, compound 4 effectively suppresses IL-6-STAT3 signaling and inflammatory cytokine production by T cells and provides a beneficial effect for treating chronic inflammatory and autoimmune disease.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Interleukin-6/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemical synthesis , Benzoxazoles/chemical synthesis , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Hep G2 Cells , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Interleukin-4/immunology , Interleukin-5/immunology , Interleukin-6/immunology , Mice, Inbred C57BL , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects
9.
Sci Rep ; 7: 42144, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28169371

ABSTRACT

Interferon-γ (IFN-γ), a critical inflammatory cytokine, is primarily produced by T helper 1 (Th1) cells and accelerates the pathogenesis of inflammatory colitis. Pharmacological suppression of IFN-γ production attenuates dysregulated inflammatory responses and may be beneficial for treating inflammatory disease. In this study, we aimed to discover potent anti-inflammatory compounds that suppress IFN-γ production and found that the novel benzoxazole derivatives, 2-((3,4-dichlorophenyl) amino) benzo[d]xazol-5-ol (DCPAB) and 2-((3,4-hydroxyphenyl) amino) benzo[d]xazol-5-ol (HPAB), suppressed IFN-γ production by T cells. Treatment of CD4+ T cells with DCPAB and HPAB selectively inhibited Th1 cell development, and DCPAB more potently suppressed IFN-γ than HPAB did. Interestingly, DCPAB and HPAB significantly suppressed the expression of T-box containing protein expressed in T cells (T-bet) that activates IFN-γ gene transcription. DCPAB additionally suppressed transcriptional activity of T-bet on IFN-γ gene promoter, whereas HPAB had no effect on T-bet activity. IFN-γ suppressive activity of DCPAB and HPAB was impaired in the absence of T-bet but was retrieved by the restoration of T-bet in T-bet-deficient T cells. Furthermore, DCPAB and HPAB attenuated inflammatory colitis development that was induced by CD4+ T cells in vivo. We suggest that the novel benzoxazole derivatives, DCPAB and HPAB, may have therapeutic effects on inflammatory colitis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzoxazoles/pharmacology , Colitis/drug therapy , Interferon-gamma/antagonists & inhibitors , T-Box Domain Proteins/immunology , Th1 Cells/drug effects , Adoptive Transfer , Animals , Anti-Inflammatory Agents/chemical synthesis , Antibodies/pharmacology , Benzoxazoles/chemical synthesis , CD3 Complex/genetics , CD3 Complex/immunology , Colitis/genetics , Colitis/immunology , Colitis/pathology , Disease Models, Animal , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Promoter Regions, Genetic , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Th1 Cells/transplantation
10.
Arch Pharm Res ; 39(11): 1530-1536, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27778276

ABSTRACT

Regulatory T (Treg) cells with high expression of both CD25 and Foxp3 are developed in the thymus and also peripheral tissues. Treg cells suppress the activation and functions of effector T cells raised against specific antigens and are crucial for maintaining immune homeostasis. Treg cell development is associated with the induction of and epigenetic alterations of forkhead transcription factor Foxp3. Foxp3 expression is increased by the activation of several transcription factors including nuclear factor-kappa B (NF-κB), nuclear factor of activated T cells (NFAT), and Smad3 in response to various signals such as TGFß, retinoic acid, and rapamycin. Recently, the orphan nuclear receptor 4A proteins (NR4As) including NR4A1 (Nur77), NR4A2 (Nurr1), and NR4A3 (Nor1) are reported to regulate Treg cell development through activation of Foxp3 and have therapeutic potentials in treating immune disorders. This review summarizes the function and regulatory mechanisms of Treg cells and also implicates current advances in immunomodulatory functions of NR4As and their therapeutic potentials in inflammation and cancer.


Subject(s)
Forkhead Transcription Factors/metabolism , Immunomodulation , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Humans , Inflammation/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
11.
Biochem Biophys Res Commun ; 474(2): 345-350, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27109480

ABSTRACT

Chloroquine (CQ) and amodiaquine (AQ) have been used for treating or preventing malaria for decades, and their application has expanded into treating inflammatory disease in humans. CQ and AQ are applicable for controlling rheumatoid arthritis, but their molecular mechanisms of anti-inflammatory activity remain to be elucidated. In this study, we examined the effects of CQ and AQ on T cell activation and T cell-mediated immune response. CQ had no significant effect on T cell numbers, but decreased the population of T cells with a high division rate. However, AQ treatment significantly increased the number of cells with low division rates and eliminated cells with high division rates, resulting in the inhibition of T cell proliferation triggered by T cell receptor stimulation, of which inhibition occurred in developing effector T helper and regulatory T cells, regardless of the different exogenous cytokines. Interestingly, the cyclin-dependent kinase inhibitor p21 was significantly and dose-dependently increased by CQ, and more potently by AQ, while other cell cycle regulators were unchanged. Both CQ and AQ elevated the transcription level of p21 though the activation of p53, but also blocked p21 protein degradation in the presence of cycloheximide, causing p21 protein accumulation mainly in the nucleus. Sustained treatment of developing T cells with either CQ or AQ suppressed IFN-γ production in a dose dependent manner and potently inhibited the differentiation of IFN-γ-producing Th1 cells. These results demonstrate that CQ and AQ increase the expression level of p21 and inhibit T cell proliferation and the development of IFN-γ-producing Th1 cells, thereby revealing beneficial roles in treating a wide range of chronic inflammatory diseases mediated by inflammatory T cells.


Subject(s)
Amodiaquine/administration & dosage , Chloroquine/administration & dosage , Cyclin-Dependent Kinase Inhibitor p21/metabolism , T-Lymphocytes/physiology , Th1 Cells/cytology , Th1 Cells/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , Th1 Cells/drug effects , Treatment Outcome
12.
Immune Netw ; 15(4): 199-205, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26330806

ABSTRACT

T-bet is a critical transcription factor that regulates differentiation of Th1 cells from CD4(+) precursor cells. Since T-bet directly binds to the promoter of the IFN-γ gene and activates its transcription, T-bet deficiency impairs IFN-γ production in Th1 cells. Interestingly, T-bet-deficient Th cells also display substantially augmented the production of IL-2, a T cell growth factor. Exogenous expression of T-bet in T-bet deficient Th cells rescued the IFN-γ production and suppressed IL-2 expression. IFN-γ and IL-2 reciprocally regulate Th cell proliferation following TCR stimulation. Therefore, we examined the effect of T-bet on Th cell proliferation and found that T-bet deficiency significantly enhanced Th cell proliferation under non-skewing, Th1-skewing, and Th2-skewing conditions. By using IFN-γ-null mice to eliminate the anti-proliferative effect of IFN-γ, T-bet deficiency still enhanced Th cell proliferation under both Th1- and Th2-skewing conditions. Since the anti-proliferative activity of T-bet may be influenced by IL-2 suppression in Th cells, we examined whether T-bet modulates IL-2-independent cell proliferation in a non-T cell population. We demonstrated that T-bet expression induced by ecdysone treatment in human embryonic kidney (HEK) cells increased IFN-γ promoter activity in a dose dependent manner, and sustained T-bet expression considerably decreased cell proliferation in HEK cells. Although the molecular mechanisms underlying anti-proliferative activity of T-bet remain to be elucidated, T-bet may directly suppress cell proliferation in an IFN-γ- or an IL-2-independent manner.

13.
J Immunol ; 191(8): 4029-37, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048895

ABSTRACT

Peroxiredoxin (Prx) II is an intracellular antioxidant molecule that eliminates hydrogen peroxide, employing a high substrate-binding affinity. PrxII deficiency increases the levels of intracellular reactive oxygen species in many types of cells, which may increase reactive oxygen species-mediated inflammation. In this study, we investigated the susceptibility of PrxII knockout (KO) mice to experimentally induced colitis and the effects of PrxII on the immune system. Wild-type mice displayed pronounced weight loss, high mortality, and colon shortening after dextran sulfate sodium administration, whereas colonic inflammation was significantly attenuated in PrxII KO mice. Although macrophages were hyperactivated in PrxII KO mice, the amount of IFN-γ and IL-17 produced by CD4(+) T cells was substantially reduced. Foxp3(+) regulatory T (Treg) cells were elevated, and Foxp3 protein expression was increased in the absence of PrxII in vitro and in vivo. Restoration of PrxII into KO cells suppressed the increased Foxp3 expression. Interestingly, endogenous PrxII was inactivated through hyperoxidation during Treg cell development. Furthermore, PrxII deficiency stabilized FoxO1 expression by reducing mouse double minute 2 homolog expression and subsequently activated FoxO1-mediated Foxp3 gene transcription. PrxII overexpression, in contrast, reduced FoxO1 and Foxp3 expression. More interestingly, adoptive transfer of naive CD4(+) T cells from PrxII KO mice into immune-deficient mice attenuated T cell-induced colitis, with a reduction in mouse double minute 2 homolog expression and an increase in FoxO1 and Foxp3 expression. These results suggest that inactivation of PrxII is important for the stability of FoxO1 protein, which subsequently mediates Foxp3(+) Treg cell development, thereby attenuating colonic inflammation.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/metabolism , Peroxiredoxins/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis/genetics , Dextran Sulfate , Forkhead Box Protein O1 , Interferon-gamma/metabolism , Interleukin-17/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxiredoxins/genetics , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes, Regulatory/metabolism
14.
J Bone Miner Res ; 28(3): 618-26, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23044658

ABSTRACT

We examined the effects that ovariectomy had on sclerostin mRNA and protein levels in the bones of 8-week-old mice that were either sham-operated (SHAM) or ovariectomized (OVX) and then euthanized 3 or 6 weeks later. In this model, bone loss occurred between 3 and 5 weeks postsurgery. In calvaria, ovariectomy significantly decreased sclerostin mRNA levels at 6 weeks postsurgery (by 52%) but had no significant effect at 3 weeks. In contrast, sclerostin mRNA levels were significantly lower in OVX femurs at 3 weeks postsurgery (by 53%) but equal to that of SHAM at 6 weeks. The effects of ovariectomy on sclerostin were not a global response of osteocytes because they were not mimicked by changes in the mRNA levels for two other relatively osteocyte-specific genes: DMP-1 and FGF-23. Sclerostin protein decreased by 83% and 60%, at 3 and 6 weeks postsurgery in calvaria, respectively, and by 38% in lumbar vertebrae at 6 weeks. We also detected decreases in sclerostin by immunohistochemistry in cortical osteocytes of the humerus at 3 weeks postsurgery. However, there were no significant effects of ovariectomy on sclerostin protein in femurs or on serum sclerostin at 3 and 6 weeks postsurgery. These results demonstrate that ovariectomy has variable effects on sclerostin mRNA and protein in mice, which are dependent on the bones examined and the time after surgery. Given the discrepancy between the effects of ovariectomy on serum sclerostin levels and sclerostin mRNA and protein levels in various bones, these results argue that, at least in mice, serum sclerostin levels may not accurately reflect changes in the local production of sclerostin in bones. Additional studies are needed to evaluate whether this is also the case in humans.


Subject(s)
Glycoproteins/metabolism , Ovariectomy , Adaptor Proteins, Signal Transducing , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factor-23 , Glycoproteins/blood , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Tomography, X-Ray Computed
15.
J Bone Miner Res ; 28(4): 948-59, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23044992

ABSTRACT

CD74 is a type II transmembrane protein that can act as a receptor for macrophage migration inhibitory factor (MIF) and plays a role in MIF-regulated responses. We reported that MIF inhibited osteoclast formation and MIF knockout (KO) mice had decreased bone mass. We therefore examined if CD74 was involved in the ability of MIF to alter osteoclastogenesis in cultured bone marrow (BM) from wild-type (WT) and CD74-deficient (KO) male mice. We also measured the bone phenotype of CD74 KO male mice. Bone mass in the femur of 8-week-old mice was measured by micro-computed tomography and histomorphometry. Bone marrow cells from CD74 KO mice formed 15% more osteoclast-like cells (OCLs) with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) (both at 30 ng/mL) compared to WT. Addition of MIF to WT cultures inhibited OCL formation by 16% but had no effect on CD74KO cultures. The number of colony forming unit granulocyte-macrophage (CFU-GM) in the bone marrow of CD74 KO mice was 26% greater than in WT controls. Trabecular bone volume (TBV) in the femurs of CD74 KO male mice was decreased by 26% compared to WT. In addition, cortical area and thickness were decreased by 14% and 11%, respectively. Histomorphometric analysis demonstrated that tartrate-resistant acid phosphatase (TRAP)(+) osteoclast number and area were significantly increased in CD74 KO by 35% and 43%, respectively compared to WT. Finally, we examined the effect of MIF on RANKL-induced-signaling pathways in bone marrow macrophage (BMM) cultures. MIF treatment decreased RANKL-induced nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) and c-Fos protein in BMM cultures by 70% and 41%, respectively. Our data demonstrate that CD74 is required for MIF to affect in vitro osteoclastogenesis. Further, the bone phenotype of CD74 KO mice is similar to that of MIF KO mice. MIF treatment of WT cultures suppressed RANKL-induced activator protein 1 (AP-1) expression, which resulted in decreased osteoclast differentiation in vitro. We propose that CD74 plays a critical role in the MIF inhibition of osteoclastogenesis.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Bone and Bones/pathology , Gene Deletion , Histocompatibility Antigens Class II/metabolism , Osteoclasts/metabolism , Osteogenesis , Receptors, Immunologic/metabolism , Acid Phosphatase/metabolism , Animals , Bone Marrow Cells/cytology , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Down-Regulation/drug effects , Humans , I-kappa B Proteins/metabolism , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha , NFATC Transcription Factors/metabolism , Organ Size/drug effects , Osteoclasts/drug effects , Phenotype , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/pharmacology , Signal Transduction/drug effects , Tartrate-Resistant Acid Phosphatase , Transcription Factor AP-1/metabolism , X-Ray Microtomography
16.
Immune Netw ; 11(3): 169-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21860610

ABSTRACT

BACKGROUND: NADPH oxidase (NOX) modulates cell proliferation, differentiation and immune response through generation of reactive oxygen species. Particularly, NOX2 is recently reported to be important for regulating Treg cell differentiation of CD4+ T cells. METHODS: We employed ovalbumin-induced airway inflammation in wild-type and NOX2-deficient mice and analyzed tissue histopathology and cytokine profiles. RESULTS: We investigated whether NOX2-deficiency affects T cell-mediated airway inflammation. Ovalbumin injection which activates T cell-mediated allergic response increased airway inflammation in wild-type mice, as evidenced by increased immune cell infiltration, allergic cytokine expression, and goblet cell hyperplasia in the lung. Interestingly, NOX2 knockout (KO) mice were more susceptible to allergen-induced lung inflammation compared to wild-type mice. Immune cells including neutrophils, lymphocytes, macrophages, and eosinophils were drastically infiltrated into the lung of NOX2 KO mice and mucus secretion was substantially increased in deficiency of NOX2. Furthermore, inflammatory allergic cytokines and eotaxin were significantly elevated in NOX2 KO mice, in accordance with enhanced generation of inflammatory cytokines interleukin-17 and interferon-γ by CD4+ T cells. CONCLUSION: These results indicate that NOX2 deficiency favorably produces inflammatory cytokines by T cells and thus increases the susceptibility to severe airway inflammation.

17.
Proc Natl Acad Sci U S A ; 108(23): 9548-53, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21593419

ABSTRACT

Emerging evidence indicates that NADPH oxidase (NOX) and its reactive oxygen species (ROS) products modulate a variety of cellular events, including proliferation, differentiation, and apoptosis. In this study, we investigated the functions of NOX2 and ROS in immune modulation using NOX2 knockout (KO) mice. Interestingly, NOX2 KO mice spontaneously developed arthritis with onset at 6-7 wk of age and high incidence (60%) at 15-18 wk of age. Arthritis severity in NOX2 KO mice was proportionally increased with age and higher in females than in males. Bone destruction was confirmed by microcomputed tomography scanning and histological analyses of joints. Inflammatory factors, including TNF-α, IL-1ß, and RANKL, and serum level of anti-type II collagen IgG were significantly increased in NOX2 KO mice. In addition, NOX2 deficiency perturbed the immune system upon aging. NOX2 KO mice demonstrated preferred development of CD11b+Gr-1+ myeloid cells with profound production of proinflammatory cytokines and augmented expression of IL-17 through the activation of STAT3 and RORγt in vivo. NOX2 deficiency increased differentiation of effector Th cells in vitro and decreased CD25+FoxP3+ Treg cells both in vitro and in vivo. Furthermore, adoptive transfer of NOX2-deficient CD4(+) T cells into RAG KO mice increased arthritic inflammation compared with WT cells. These results demonstrated that NOX2 deficiency affected the development of CD11b+ myeloid cells and Th17/Treg cells, and thus promoted inflammatory cytokine production and inflammatory arthritis development, strongly supporting a crucial role for ROS generation in the modulation of Th17/Treg cell development and its related inflammatory immune response upon aging.


Subject(s)
Aging/immunology , Arthritis/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Membrane Glycoproteins/immunology , NADPH Oxidases/immunology , Adoptive Transfer , Animals , Arthritis/genetics , Arthritis/metabolism , Bone Density , Bone and Bones/abnormalities , Bone and Bones/metabolism , CD11b Antigen/immunology , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cells, Cultured , Female , Flow Cytometry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , X-Ray Microtomography
18.
PLoS One ; 6(3): e18168, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21464945

ABSTRACT

Increasing evidence that decreased bone density and increased rates of bone fracture are associated with abnormal metabolic states such as hyperglycemia and insulin resistance indicates that diabetes is a risk factor for osteoporosis. In this study, we observed that TallyHo/JngJ (TH) mice, a polygenic model of type II diabetes, spontaneously developed bone deformities with osteoporotic features. Female and male TH mice significantly gained more body weight than control C57BL/6 mice upon aging. Interestingly, bone density was considerably decreased in male TH mice, which displayed hyperglycemia. The osteoblast-specific bone forming markers osteocalcin and osteoprotegerin were decreased in TH mice, whereas osteoclast-driven bone resorption markers such as IL-6 and RANKL were significantly elevated in the bone marrow and blood of TH mice. In addition, RANKL expression was prominently increased in CD4+ T cells of TH mice upon T cell receptor stimulation, which was in accordance with enhanced IL-17 production. IL-17 production in CD4+ T cells was directly promoted by treatment with leptin while IFN-γ production was not. Moreover, blockade of IFN-γ further increased RANKL expression and IL-17 production in TH-CD4+ T cells. In addition, the osteoporotic phenotype of TH mice was improved by treatment with alendronate. These results strongly indicate that increased leptin in TH mice may act in conjunction with IL-6 to preferentially stimulate IL-17 production in CD4+ T cells and induce RANKL-mediated osteoclastogenesis. Accordingly, we propose that TH mice could constitute a beneficial model for osteoporosis.


Subject(s)
Bone Resorption/metabolism , CD4-Positive T-Lymphocytes/metabolism , Interleukin-17/biosynthesis , RANK Ligand/metabolism , Alendronate/pharmacology , Animals , Biomarkers/metabolism , Bone Density/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Resorption/blood , Bone Resorption/physiopathology , CD4-Positive T-Lymphocytes/drug effects , Female , Interferon-gamma/blood , Interleukin-6/blood , Leptin/blood , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/blood , Osteogenesis/drug effects , Testosterone/pharmacology
19.
Antioxid Redox Signal ; 13(5): 575-87, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20367278

ABSTRACT

Engagement of T cell receptor (TCR) triggers signaling pathways that mediate activation, proliferation, and differentiation of T lymphocytes. Such signaling events are mediated by reactive oxygen species (ROS), including hydrogen peroxide and lipid peroxides, both of which are reduced by glutathione peroxidase 1 (GPx1). We have now examined the role of GPx1 in the activation, differentiation, and functions of CD4(+) T helper (Th) cells. TCR stimulation increased the intracellular ROS concentration in Th cells in a time-dependent manner, and such TCR-induced ROS generation was found to promote cell proliferation. GPx1-deficient Th cells produced higher levels of intracellular ROS and interleukin-2 than wild-type Th cells and proliferated at a faster rate than did wild-type cells. Moreover, differentiation of GPx1-deficient Th cells was biased toward Th1, and Th17 cell development was also impeded by GPx1 depletion. Consistent with these findings, GPx1-null mice were protected from the development of ovalbumin-induced allergic asthma. Eosinophil infiltration, goblet cell hyperplasia, collagen deposition, and airway hyperresponsiveness were thus all attenuated in the lungs of GPx1-null mice. These data indicate that GPx1-dependent control of intracellular ROS accumulation is important not only for regulation of Th cell proliferation but for modulation of differentiation into Th1, Th2, and Th17 cells.


Subject(s)
Allergens/immunology , Bronchial Hyperreactivity/immunology , Cell Differentiation/genetics , Glutathione Peroxidase/deficiency , T-Lymphocytes, Helper-Inducer/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antioxidants/pharmacology , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/physiopathology , CD3 Complex/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cells, Cultured , Collagen/metabolism , Cytokines/metabolism , Eosinophils/cytology , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression/immunology , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Goblet Cells/cytology , Goblet Cells/metabolism , Homeodomain Proteins/genetics , Interferon-gamma/genetics , Interleukin-2/genetics , Interleukin-2/metabolism , Interleukin-4/genetics , Lung/cytology , Lung/immunology , Lung/metabolism , Lymph Nodes/cytology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Reactive Oxygen Species/metabolism , Spleen/cytology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology
20.
Biochem Biophys Res Commun ; 394(3): 811-6, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20233578

ABSTRACT

G12 family have been known to modulate a variety of cellular events such as cell migration, B cell activation and maturation, cytokine production, and cell differentiation. In particular, Galpha12 modulates IgG production, thus induces IgG antibody-mediated immune responses. However, it is largely unknown whether Galpha12 is required for T cell-mediated immune functions. In this study, we investigated the effects of Galpha12 in the activation and differentiation of CD4+ T cells. While PMA plus ionomycin induced equal levels of IL-2 production in WT and Galpha12-deficient lymphocytes, TCR-triggered IL-2 production was significantly attenuated in Galpha12 KO lymphocytes. In particular, CD4+ T cells and effector Th cells lacking of Galpha12 revealed diminished IL-2 production, but not IFNgamma production, upon TCR stimulation. In addition, supplement of IL-2 preferentially induced Galpha12-deficient CD4+ T cells into Th2 and Th17 cells; however, the expression of specific transcription factors was unchanged in Galpha12 KO Th cells. While IL-2 expression was still diminished by the re-stimulation with anti-CD3, PMA plus ionomycin restored IL-2 production in Galpha12-deficient Th1 and Th2 cells. These results suggest that Galpha12 may be a critical signaling molecule in TCR-induced IL-2 production and also relay a signal to suppress Th2 and Th17 cell differentiation.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/physiology , Interleukin-17/immunology , Interleukin-2/biosynthesis , T-Lymphocytes, Helper-Inducer/immunology , Th2 Cells/immunology , Animals , CD4 Antigens/immunology , Cell Differentiation , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...