Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 60
2.
PLoS One ; 14(10): e0223842, 2019.
Article En | MEDLINE | ID: mdl-31622977

The process by which fibroblasts are directly reprogrammed into cardiomyocytes involves two stages; initiation and maturation. Initiation represents the initial expression of factors that induce fibroblasts to transdifferentiate into cardiomyocytes. Following initiation, the cell undergoes a period of maturation before becoming a mature cardiomyocyte. We wanted to understand the role of cardiac development transcription factors in the maturation process. We directly reprogram fibroblasts into cardiomyocytes by a combination of miRNAs (miR combo). The ability of miR combo to induce cardiomyocyte-specific genes in fibroblasts was lost following the knockdown of the cardiac transcription factors Gata4, Mef2C, Tbx5 and Hand2 (GMTH). To further clarify the role of GMTH in miR combo reprogramming we utilized a modified CRISPR-Cas9 approach to activate endogenous GMTH genes. Importantly, both miR combo and the modified CRISPR-Cas9 approach induced comparable levels of GMTH expression. While miR combo was able to reprogram fibroblasts into cardiomyocyte-like cells, the modified CRISPR-Cas9 approach could not. Indeed, we found that cardiomyocyte maturation only occurred with very high levels of GMT expression. Taken together, our data indicates that while endogenous cardiac transcription factors are insufficient to reprogram fibroblasts into mature cardiomyocytes, endogenous cardiac transcription factors are necessary for expression of maturation genes.


GATA4 Transcription Factor/genetics , T-Box Domain Proteins/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Transdifferentiation , Cells, Cultured , Cellular Reprogramming , Fibroblasts/cytology , Fibroblasts/metabolism , GATA4 Transcription Factor/antagonists & inhibitors , GATA4 Transcription Factor/metabolism , Gene Editing , MEF2 Transcription Factors/antagonists & inhibitors , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering/metabolism , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/metabolism
3.
Mol Immunol ; 114: 561-570, 2019 10.
Article En | MEDLINE | ID: mdl-31522074

Autism is a neurodevelopmental disorder characterized by deficits and qualitative impairments in communication and implicit skill learning. Its prevalence is higher than previous estimates, and treatments have limited efficacy and are costly. Here, we assessed the therapeutic potential of JNJ77777120 (JNJ), a histamine-4 receptor (H4R) antagonist, using BTBR T+ Itpr3tf/J (BTBR) mice, a confirmed model of autism, and C57BL/6J (C57) mice, a commonly chosen reference strain. We first examined the effects of JNJ treatment on BTBR mice exposed to gamma-rays (irradiation-exposed) using a three-chambered apparatus. We further investigated the possible molecular mechanisms through which JNJ administration modulates IL-17A-, RORγT-, IL-22-, T-bet-, STAT3-, ICOS-, and Foxp3-producing CD8+ T cells in the spleens of irradiation-exposed BTBR mice. The effects of JNJ administration on the mRNA and protein expression of IL-17A, RORγT, IL-22, T-bet, STAT-3, pSTAT3, IL-10, and Foxp3 in brain tissue were also explored. Results showed that JNJ treatment with irradiation exposure increased social interactions in BTBR mice compared to that in irradiation-exposed BTBR mice. Additionally, JNJ-treated and irradiation-exposed BTBR mice exhibited decreases in IL-17A-, RORγT-, IL-22-, T-bet-, and STAT3-producing CD8+ T cells and increases in ICOS- and Foxp3-producing CD8+ T cells. Moreover, JNJ treatment and irradiation exposure in BTBR mice regulated the mRNA and protein expression levels of IL-17A, RORγT, IL-22, T-bet, STAT3, pSTAT-3, IL-10, and Foxp3 in the brain tissue. These results suggest that JNJ is useful for the treatment of autism, as this H4R antagonist could block inflammatory cytokine production and transcription factor signaling.


CD8-Positive T-Lymphocytes/drug effects , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Piperazines/pharmacology , Receptors, Histamine H4/antagonists & inhibitors , Signal Transduction/drug effects , T-Box Domain Proteins/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Gamma Rays/adverse effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteins/metabolism
4.
Lipids Health Dis ; 17(1): 147, 2018 Jun 23.
Article En | MEDLINE | ID: mdl-29935534

BACKGROUND: Among the eight stereoisomers of phytanic acid (PA), the 3RS, 7R, 11R-isomer is naturally occurring and is present in foods and the human body. PA is considered to have possible health benefits in the immune system. However, it remains undetermined whether these effects are elicited by the 3RS, 7R, 11R-PA isomer, because previous studies used a commercially available PA whose isomer configuration is unknown. In this study, we synthesized a preparation of 3RS, 7R, 11R-PA, and investigated its in vitro immunomodulatory effects, especially the T-cell production of interferon (IFN)-γ, which is associated with various autoimmune diseases. This study also investigated the effects of 3RS, 7R, 11R-PA on NF-κB activity in order to address the mechanism of its immunomodulatory effects. METHODS: Mouse splenocytes and purified T-cells were stimulated with T-cell mitogens and incubated with 3RS, 7R, 11R-PA, followed by evaluation of IFN-γ production. The effect of 3RS, 7R, 11R-PA on NF-κB activity was also investigated using an A549 cell line with stable expression of an NF-κB-dependent luciferase reporter gene. RESULTS: 3RS, 7R, 11R-PA significantly reduced in vitro IFN-γ production at both the protein and mRNA levels, and was accompanied by decreased expression of T-bet, a key regulator of Th1 cell differentiation. The results indicated that NF-κB-mediated transcriptional activity was significantly decreased by 3RS, 7R, 11R-PA and that GW6471, an antagonist of peroxisome proliferator activated receptor α (PPARα), abrogated the inhibitory effect of 3RS, 7R, 11R-PA on NF-κB activity. CONCLUSIONS: The present study suggests that 3RS, 7R, 11R-PA is a functional and bioactive fatty acid, and has a potentially beneficial effect for amelioration of T-cell mediated autoimmune diseases. This study also indicates that interference in the NF-κB pathway via PPARα activation is a potential mechanism of the immunomodulatory effects of 3RS, 7R, 11R-PA.


Immunologic Factors/pharmacology , Interferon-gamma/genetics , PPAR alpha/genetics , Phytanic Acid/pharmacology , RNA, Messenger/genetics , T-Lymphocytes/drug effects , A549 Cells , Animals , Cell Differentiation/drug effects , Female , Gene Expression Regulation , Genes, Reporter , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Luciferases/genetics , Luciferases/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/immunology , Oxazoles/pharmacology , PPAR alpha/agonists , PPAR alpha/antagonists & inhibitors , PPAR alpha/immunology , Phytohemagglutinins/antagonists & inhibitors , Phytohemagglutinins/pharmacology , Primary Cell Culture , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/immunology , Signal Transduction , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
5.
Front Immunol ; 9: 3132, 2018.
Article En | MEDLINE | ID: mdl-30687330

Susceptibility to ankylosing spondylitis (AS) is polygenic with more than 100 genes identified to date. These include HLA-B27 and the aminopeptidases (ERAP1, ERAP2, and LNPEPS), which are involved in antigen processing and presentation to T-cells, and several genes (IL23R, IL6R, STAT3, JAK2, IL1R1/2, IL12B, and IL7R) involved in IL23 driven pathways of inflammation. AS is also strongly associated with polymorphisms in two transcription factors, RUNX3 and T-bet (encoded by TBX21), which are important in T-cell development and function. The influence of these genes on the pathogenesis of AS and their potential for identifying drug targets is discussed here.


Core Binding Factor Alpha 3 Subunit/genetics , Gene Expression Regulation/immunology , Interleukin-23/metabolism , Spondylitis, Ankylosing/immunology , T-Box Domain Proteins/genetics , Aminopeptidases/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation/drug effects , HLA-B27 Antigen/genetics , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interleukin-23/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Molecular Targeted Therapy/methods , Polymorphism, Single Nucleotide , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Spondylitis, Ankylosing/genetics , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/metabolism
6.
Mol Cancer Ther ; 17(3): 603-613, 2018 03.
Article En | MEDLINE | ID: mdl-29237806

Chordomas are rare bone tumors with no approved therapy. These tumors express several activated tyrosine kinase receptors, which prompted attempts to treat patients with tyrosine kinase inhibitors. Although clinical benefit was observed in phase II clinical trials with imatinib and sorafenib, and sporadically also with EGFR inhibitors, therapies evaluated to date have shown modest activity. With the goal of identifying new drugs with immediate therapeutic potential for chordoma patients, we collected clinically approved drugs and other advanced inhibitors of MET, PDGFRß, and EGFR tyrosine kinases, and assessed their antiproliferative activity against a panel of chordoma cell lines. Chordoma cell lines were not responsive to MET and PDGFRß inhibitors. U-CH1 and UM-Chor1 were sensitive to all EGFR inhibitors, whereas the remaining cell lines were generally insensitive to these drugs. Afatinib was the only EGFR inhibitor with activity across the chordoma panel. We then investigated the molecular mechanisms behind the responses observed and found that the antiproliferative IC50s correlate with the unique ability of afatinib to promote degradation of EGFR and brachyury, an embryonic transcription factor considered a key driver of chordoma. Afatinib displayed potent antitumor efficacy in U-CH1, SF8894, CF322, and CF365 chordoma tumor models in vivo In the panel analyzed, high EGFR phosphorylation and low AXL and STK33 expression correlated with higher sensitivity to afatinib and deserve further investigation as potential biomarkers of response. These data support the use of afatinib in clinical trials and provide the rationale for the upcoming European phase II study on afatinib in advanced chordoma. Mol Cancer Ther; 17(3); 603-13. ©2017 AACR.


Afatinib/therapeutic use , Bone Neoplasms/drug therapy , Chordoma/drug therapy , ErbB Receptors/antagonists & inhibitors , Fetal Proteins/antagonists & inhibitors , T-Box Domain Proteins/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chordoma/genetics , Chordoma/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Nude , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
7.
Mol Med Rep ; 16(5): 6050-6058, 2017 Nov.
Article En | MEDLINE | ID: mdl-28849151

T­Box (TBX)­2 is a member of the T­box gene family, which is aberrantly expressed in numerous types of malignant tumors, and has previously been demonstrated to be conducive to tumor progression by acting as a transcription factor. However, specific information regarding the expression and function of TBX2 in prostate cancer cells remains to be elucidated. The present study demonstrated that silencing of TBX2 by TBX2 small interfering RNA inhibited cell proliferation and promoted cell senescence. It was demonstrated that knockdown of TBX2 inhibited cell metastatic abilities by upregulating E­cadherin and downregulating N­cadherin, Vimentin and fibronectin. In addition, the expression of TBX2 in prostate cancer tissues and tumor adjacent tissues was detected by immunohistochemistry. The results indicated that the expression rates of TBX2 were significantly increased in the cancerous tissues, compared with the healthy tumor adjacent tissue, and TBX2 increased staining was associated with the clinical stage and pathological grade. The findings of the present study therefore suggest that TBX2 expression is markedly increased in prostate cancer and TBX2 may act as a potential beneficial therapeutic target for the future treatment of prostate cancer.


Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/genetics , Prostatic Neoplasms/genetics , RNA, Small Interfering/genetics , T-Box Domain Proteins/genetics , Aged , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/genetics , Cadherins/agonists , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Fibronectins/genetics , Fibronectins/metabolism , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neovascularization, Pathologic/diagnosis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/metabolism , Signal Transduction , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/metabolism , Vimentin/genetics , Vimentin/metabolism
8.
Mol Med Rep ; 16(5): 6368-6375, 2017 Nov.
Article En | MEDLINE | ID: mdl-28849203

Abnormal immune response resulting from disordered T helper (Th)1/Th2 and Th17/regulatory T cells (Treg) cytokine expression has been demonstrated to serve an important role in the pathogenesis of preeclampsia (PE). However, the role of transcription factors regulating Th cell differentiation contributing to PE remain unclear. To determine whether a decrease in the expression of the T cell lineage transcription factor T­bet can restore immune balance and alleviate the systemic inflammatory response present in PE, 30 patients diagnosed with PE were assessed and compared with healthy pregnant controls. The expression of the transcription factors T­bet and retinoic acid receptor­related orphan receptor (ROR)γt were increased in the peripheral blood mononuclear cells of PE patients compared with controls, consistent with the presence of abnormally high T­bet:GATA3 and RORγt:forkhead box (FOX) P3 ratios. The present study additionally identified a high­efficiency, specific small interfering (si)RNA that can downregulate RORγt and T­bet mRNA levels and inhibit protein expression. This effective siRNA was transfected into activated CD4+ T cells derived from patients with PE to observe the changes to transcription factor expression and attempt to elucidate the regulatory mechanism of T cell subsets. It was identified that knockdown of RORγt induced increased expression of FOXP3 and that the ratios of RORγt:FOXP3 and interleukin (IL)­17A:IL­10 were subsequently decreased. The results suggested that siRNA­mediated knockdown of T­bet regulated the immune balance of Th17/Tregs via changes to RORγt and FOXP3. When siRNA against RORγt and T­bet were used in combination, a stronger ability to regulate immune balance was observed. These results imply that Th1­ and Th17­type immunity is dominant in PE and that the siRNA­mediated knockdown of certain Th1 and Th17 cell transcription factors may be an effective therapeutic target for promoting immune balance in CD4+ T cell subgroups and ameliorating local and generalized inflammation in PE.


Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Pre-Eclampsia/genetics , RNA, Small Interfering/genetics , T-Box Domain Proteins/genetics , T-Lymphocytes, Helper-Inducer/immunology , Case-Control Studies , Cell Differentiation , Cell Lineage , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Gene Expression Regulation , Gestational Age , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Pre-Eclampsia/immunology , Pre-Eclampsia/pathology , Pregnancy , Primary Cell Culture , RNA, Small Interfering/immunology , Signal Transduction , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/immunology , T-Lymphocytes, Helper-Inducer/classification , T-Lymphocytes, Helper-Inducer/pathology
9.
Cancer Res ; 77(6): 1331-1344, 2017 03 15.
Article En | MEDLINE | ID: mdl-28108510

Identification of factors that mediate visceral and bone metastatic spread and subsequent bone remodeling events is highly relevant to successful therapeutic intervention in advanced human prostate cancer. TBX2, a T-box family transcription factor that negatively regulates cell-cycle inhibitor p21, plays critical roles during embryonic development, and recent studies have highlighted its role in cancer. Here, we report that TBX2 is overexpressed in human prostate cancer specimens and bone metastases from xenograft mouse models of human prostate cancer. Blocking endogenous TBX2 expression in PC3 and ARCaPM prostate cancer cell models using a dominant-negative construct resulted in decreased tumor cell proliferation, colony formation, and invasion in vitro Blocking endogenous TBX2 in human prostate cancer mouse xenografts decreased invasion and abrogation of bone and soft tissue metastasis. Furthermore, blocking endogenous TBX2 in prostate cancer cells dramatically reduced bone-colonizing capability through reduced tumor cell growth and bone remodeling in an intratibial mouse model. TBX2 acted in trans by promoting transcription of the canonical WNT (WNT3A) promoter. Genetically rescuing WNT3A levels in prostate cancer cells with endogenously blocked TBX2 partially restored the TBX2-induced prostate cancer metastatic capability in mice. Conversely, WNT3A-neutralizing antibodies or WNT antagonist SFRP-2 blocked TBX2-induced invasion. Our findings highlight TBX2 as a novel therapeutic target upstream of WNT3A, where WNT3A antagonists could be novel agents for the treatment of metastasis and for skeletal complications in prostate cancer patients. Cancer Res; 77(6); 1331-44. ©2017 AACR.


Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/metabolism , Bone Neoplasms/prevention & control , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/prevention & control , T-Box Domain Proteins/antagonists & inhibitors , Wnt3A Protein/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, SCID , Molecular Targeted Therapy , Neoplasm Grading , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tumor Cells, Cultured , Wnt3A Protein/genetics , Wnt3A Protein/metabolism , Xenograft Model Antitumor Assays
10.
BMC Cancer ; 16(1): 671, 2016 08 23.
Article En | MEDLINE | ID: mdl-27553211

BACKGROUND: TBX3 is a T-box transcription factor repressor that is elevated in metastatic breast cancer and is believed to promote malignancy of tumor cells, possibly by promoting cell survival and epithelial-mesenchymal transition. METHODS: The relative expression of TBX3 was assessed in the 21T cell lines which were derived from an individual patient and represent three distinct stages of breast cancer progression: 21PT, atypical ductal hyperplasia; 21NT, ductal carcinoma in situ; and 21MT-1, invasive mammary carcinoma. Two different isoforms of TBX3 (TBX3iso1 and TBX3iso2) were overexpressed to evaluate cell survival/colony forming ability, growth, and invasion in the ductal carcinoma in situ-like 21NT cell line using an in vitro Matrigel model of cancer progression. In addition, TBX3 expression was knocked down to evaluate the effects of downregulating TBX3 on the invasive mammary carcinoma-like 21MT-1 cell line. Finally, PCR array profiling was used to assess alterations in gene expression due to TBX3 overexpression in the 21NT cells. RESULTS: TBX3 is abundant in the invasive 21MT-1 cell line, while being minimally expressed in the non-invasive 21NT and 21PT cell lines. Overexpression of either TBX3iso1 or TBX3iso2 in 21NT cells resulted in increased cell survival/colony forming ability, growth vs. apoptosis and invasion in Matrigel. In contrast, short hairpin RNA-mediated knockdown of TBX3 in the 21MT-1 cells resulted in smaller colonies, with a more regular, less dispersed (less infiltrative) morphology. Array profiling of the 21NT TBX3 iso1 and iso2 transfectants showed that there are common alterations in expression of several genes involved in signal transduction, cell cycle control/cell survival, epithelial-mesenchymal transition and invasiveness. CONCLUSIONS: Overall, these results indicate that TBX3 (isoform 1 or 2) expression can promote progression in a model of early breast cancer by altering cell properties involved in cell survival/colony formation and invasiveness, as well as key regulatory and EMT/invasiveness-related gene expressions.


Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Gene Expression Regulation, Neoplastic , Hyperplasia/pathology , T-Box Domain Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Collagen , Disease Progression , Drug Combinations , Epithelial-Mesenchymal Transition , Female , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Laminin , Neoplasm Invasiveness , Protein Isoforms , Proteoglycans , RNA, Small Interfering/genetics , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , Tumor Cells, Cultured
11.
Scand J Immunol ; 83(4): 288-96, 2016 Apr.
Article En | MEDLINE | ID: mdl-27028319

Interleukin (IL)-10 has been implicated in persistence of pathogens in a number of chronic infections. Infected CD4+ cells upon reactivation with HIV antigens were also shown to produce IL-10, which might contribute to their persistence. Hence, it is crucial to determine mechanisms regulating IL-10 production after activation with HIV antigens for devising effective blocking strategies. In this study, ERK-, T-bet- and FoxP3-dependent pathways were evaluated for their possible roles in IL-10 production by infected CD4+ cells after reactivation with HIV Env. Intracellular and secreted IL-10 levels were determined by flow cytometry and Bioplex assay after treating PBMCs with PD98059, tipifarnib and cyclosporin A for blocking of ERK-, T-bet-and FoxP3-dependent pathways, respectively. Baseline levels of T-bet, pERK were higher in P24+ CD4+ cells as compared to uninfected CD4+ cells, which increased further after activation with Env. Inhibition of T-bet resulted in 2.3-fold reduction of IL-10 expression whereas ERK and FoxP3 inhibition failed to cause suppression of IL-10 expression. Conversely, IL-10 secreted by PBMCs was inhibited maximally after ERK inhibition suggesting its role in regulation of cytokine secretory pathway. IFN-γ was found to be suppressed after treatment with inhibitors of all these pathways. Thus, the study highlighted need for IL-10 blockade along with the use of antigens for therapeutic vaccinations or latency reversal and identified the T-bet-dependent pathway as an important pathway regulating IL-10 production by infected CD4+ cells. However, simultaneous blockade of IFN-γ precludes use of inhibitor of this pathway as an IL-10 blocking strategy.


CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Interleukin-10/biosynthesis , T-Box Domain Proteins/metabolism , Adult , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Cyclosporine/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Flavonoids/pharmacology , Forkhead Transcription Factors/antagonists & inhibitors , HIV Core Protein p24/metabolism , HIV Infections/virology , Humans , Interferon-gamma/biosynthesis , Interleukin-10/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , Quinolones/pharmacology , T-Box Domain Proteins/antagonists & inhibitors , Young Adult
12.
Immunology ; 147(4): 367-76, 2016 Apr.
Article En | MEDLINE | ID: mdl-26726991

Initially understood to be a key regulator of interferon-γ-producing helper T cells, our knowledge of T-bet's functional roles has expanded to encompass a growing range of cellular lineages. In addition to regulating other interferon-γ-producing adaptive immune cells, it is now clear that T-bet plays a fundamental role in the regulation of innate immune responses across mucosal surfaces. This homeostatic role is demonstrated by the spontaneous colitis that occurs when T-bet is deleted from innate immune cells in RAG(-/-) mice. Using this model as a focal point, we review our understanding of T-bet's regulation of adaptive and innate immune systems, focusing particularly on mucosal populations including innate lymphoid cells, dendritic cells and intraepithelial lymphocytes. With the increasingly diverse effects of T-bet on different lineages, the classical binding-centric paradigm of T-bet's molecular functionality has increasingly struggled to account for the versatility of T-bet's biological effects. Recent recognition of the synergistic interactions between T-bet and other canonical transcription factors has led to a co-operative paradigm that has provided greater explanatory power. Synthesizing insights from ChIP-seq and comparative biology, we expand the co-operative paradigm further and suggest a network approach as a powerful way to understand and model T-bet's diverse functionality.


Immunity, Mucosal , Immunomodulation , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Adaptive Immunity , Animals , Drug Discovery , Gene Expression Regulation , Homeostasis/immunology , Humans , Mucous Membrane/cytology , Mucous Membrane/immunology , Mucous Membrane/metabolism , T-Box Domain Proteins/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transcription, Genetic
13.
Mol Med Rep ; 12(1): 995-1001, 2015 Jul.
Article En | MEDLINE | ID: mdl-25683840

The objective of the current study was to investigate the impact of Brachyury on epithelial-mesenchymal transitions and chemosensitivity in non-small cell lung cancer (NSCLC). In 115 archived NSCLC tissue samples, the expression of Brachyury was observed to be significantly higher than that in adjacent normal lung tissues. In addition, the current study demonstrated that the expression of Brachyury is closely associated with TNM staging, lymph node metastasis and the prognosis of NSCLC, although not with patient age, gender or tumor differentiation. Brachyury expression is also accompanied by the downregulation of E-cadherin and the upregulation of N-cadherin. Brachyury may promote lung cancer through induction of epithelial-mesenchymal transition, which leads to metastasis and consequent poor prognosis in patients with lung cancer. Furthermore, the present study observed that interfering with Brachyury increases the sensitivity of cells to chemotherapeutic treatment with cisplatin. These results, in combination with those of additional studies, suggest that Brachyury may be used as a novel target for the prevention and treatment of lung cancer.


Carcinoma, Non-Small-Cell Lung/genetics , Epithelial-Mesenchymal Transition/genetics , Fetal Proteins/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , T-Box Domain Proteins/genetics , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Fetal Proteins/antagonists & inhibitors , Fetal Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/metabolism
14.
J Mol Cell Biol ; 6(4): 272-85, 2014 Aug.
Article En | MEDLINE | ID: mdl-24924767

Development of animal embryos before zygotic genome activation at the midblastula transition (MBT) is essentially supported by egg-derived maternal products. Nodal proteins are crucial signals for mesoderm and endoderm induction after the MBT. It remains unclear which maternal factors activate zygotic expression of nodal genes in the ventrolateral blastodermal margin of the zebrafish blastulas. In this study, we show that loss of maternal Eomesodermin a (Eomesa), a T-box transcription factor, impairs zygotic expression of the nodal genes ndr1 and ndr2 as well as mesodermal and endodermal markers, indicating an involvement in mesendoderm induction. Maternal Eomesa is also required for timely zygotic expression of the transcription factor gene mxtx2, a regulator of nodal gene expression. Eomesa directly binds to the Eomes-binding sites in the promoter or enhancer of ndr1, ndr2, and mxtx2 to activate their transcription. Furthermore, human and mouse Nodal genes are also regulated by Eomes. Transfection of zebrafish eomesa into murine embryonic stem cells promotes mesendodermal differentiation with constant higher levels of endogenous Nodal expression, suggesting a conserved function of Eomes. Taken together, our findings reveal a conserved role of maternal T-box transcription factors in regulating nodal gene expression and mesendoderm induction in vertebrate embryos.


Embryo, Nonmammalian/cytology , Endoderm/cytology , Gene Expression Regulation, Developmental , Mesoderm/cytology , Nodal Protein/metabolism , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Zygote/metabolism , Animals , Cell Differentiation , Chromatin Immunoprecipitation , Embryo, Nonmammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mesoderm/metabolism , Mice , Nodal Protein/genetics , Nodal Signaling Ligands/genetics , Nodal Signaling Ligands/metabolism , Oocytes/cytology , Oocytes/metabolism , RNA, Small Interfering/genetics , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zygote/cytology
15.
Biochem Biophys Res Commun ; 449(3): 289-94, 2014 Jul 04.
Article En | MEDLINE | ID: mdl-24845384

The T-box transcriptional factor T-bet is crucial in the development, differentiation and function of Th1 cells. It drives Th1 immune response primarily through promoting expression of Th1 hallmark cytokine IFN-γ. Although T-bet was found associated with many immune-mediated diseases such as asthma and systemic sclerosis, little is known about the regulation of T-bet stability and function. Here we identified USP10, a carboxyl-terminal ubiquitin-processing protease, could interact with T-bet in the nucleus. Overexpression of USP10 directly inhibited T-bet ubiquitination and increased the expression of T-bet. We further confirmed Quercetin, a reported inhibitor of T-bet, could target USP10. Quercetin treatment downregulated USP10 and promoted T-bet degradation in a proteasome dependent way. Moreover, we found USP10 expression was upregulated in asthmatic patient PBMC, suggesting USP10 may maintain high level of T-bet and IFN-γ to fight against Th2-dominated inflammation.


T-Box Domain Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination/physiology , Asthma/immunology , Cell Nucleus/metabolism , Down-Regulation , HEK293 Cells , Humans , Inflammation , Protein Stability , Proteolysis , Quercetin/pharmacology , T-Box Domain Proteins/antagonists & inhibitors , Th1 Cells/immunology , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitination/drug effects
16.
Oncotarget ; 5(6): 1609-20, 2014 Mar 30.
Article En | MEDLINE | ID: mdl-24742492

TBX2 is an oncogenic transcription factor known to drive breast cancer proliferation. We have identified the cysteine protease inhibitor Cystatin 6 (CST6) as a consistently repressed TBX2 target gene, co-repressed through a mechanism involving Early Growth Response 1 (EGR1). Exogenous expression of CST6 in TBX2-expressing breast cancer cells resulted in significant apoptosis whilst non-tumorigenic breast cells remained unaffected. CST6 is an important tumor suppressor in multiple tissues, acting as a dual protease inhibitor of both papain-like cathepsins and asparaginyl endopeptidases (AEPs) such as Legumain (LGMN). Mutation of the CST6 LGMN-inhibitory domain completely abrogated its ability to induce apoptosis in TBX2-expressing breast cancer cells, whilst mutation of the cathepsin-inhibitory domain or treatment with a pan-cathepsin inhibitor had no effect, suggesting that LGMN is the key oncogenic driver enzyme. LGMN activity assays confirmed the observed growth inhibitory effects were consistent with CST6 inhibition of LGMN. Knockdown of LGMN and the only other known AEP enzyme (GPI8) by siRNA confirmed that LGMN was the enzyme responsible for maintaining breast cancer proliferation. CST6 did not require secretion or glycosylation to elicit its cell killing effects, suggesting an intracellular mode of action. Finally, we show that TBX2 and CST6 displayed reciprocal expression in a cohort of primary breast cancers with increased TBX2 expression associating with increased metastases. We have also noted that tumors with altered TBX2/CST6 expression show poor overall survival. This novel TBX2-CST6-LGMN signaling pathway, therefore, represents an exciting opportunity for the development of novel therapies to target TBX2 driven breast cancers.


Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cystatin M/genetics , Cysteine Endopeptidases/metabolism , T-Box Domain Proteins/metabolism , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Chromatin Immunoprecipitation , Cystatin M/metabolism , Cysteine Endopeptidases/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glycosylation , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , Tumor Cells, Cultured
17.
Open Biol ; 4: 140014, 2014 Apr 23.
Article En | MEDLINE | ID: mdl-24759614

The transcription factor Tbx5 is expressed in the developing heart, eyes and anterior appendages. Mutations in human TBX5 cause Holt-Oram syndrome, a condition characterized by heart and upper limb malformations. Tbx5-knockout mouse embryos have severely impaired forelimb and heart morphogenesis from the earliest stages of their development. However, zebrafish embryos with compromised tbx5 function show a complete absence of pectoral fins, while heart development is disturbed at significantly later developmental stages and eye development remains to be thoroughly analysed. We identified a novel tbx5 gene in zebrafish--tbx5b--that is co-expressed with its paralogue, tbx5a, in the developing eye and heart and hypothesized that functional redundancy could be occurring in these organs in embryos with impaired tbx5a function. We have now investigated the consequences of tbx5a and/or tbx5b downregulation in zebrafish to reveal that tbx5 genes have essential roles in the establishment of cardiac laterality, dorsoventral retina axis organization and pectoral fin development. Our data show that distinct relationships between tbx5 paralogues are required in a tissue-specific manner to ensure the proper morphogenesis of the three organs in which they are expressed. Furthermore, we uncover a novel role for tbx5 genes in the establishment of correct heart asymmetry in zebrafish embryos.


Gene Expression Regulation, Developmental , T-Box Domain Proteins/metabolism , Zebrafish/metabolism , 5' Untranslated Regions , Animal Fins/growth & development , Animal Fins/metabolism , Animals , Down-Regulation , Embryo, Nonmammalian/metabolism , Gene Knockdown Techniques , Heart/growth & development , Left-Right Determination Factors/genetics , Left-Right Determination Factors/metabolism , Morpholinos/metabolism , Mutation , Myocardium/metabolism , Phenotype , Retina/growth & development , Retina/metabolism , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
18.
Mol Biol Cell ; 25(10): 1586-93, 2014 May.
Article En | MEDLINE | ID: mdl-24623725

During wound healing and cancer metastasis, cells are frequently observed to migrate in collective groups. This mode of migration relies on the cooperative guidance of leader and follower cells throughout the collective group. The upstream determinants and molecular mechanisms behind such cellular guidance remain poorly understood. We use live-cell imaging to track the behavior of epithelial sheets of keratinocytes in response to transforming growth factor ß (TGFß), which stimulates collective migration primarily through extracellular regulated kinase 1/2 (Erk1/2) activation. TGFß-treated sheets display a spatial pattern of Erk1/2 activation in which the highest levels of Erk1/2 activity are concentrated toward the leading edge of a sheet. We show that Erk1/2 activity is modulated by cellular density and that this functional relationship drives the formation of patterns of Erk1/2 activity throughout sheets. In addition, we determine that a spatially constrained pattern of Erk1/2 activity results in collective migration that is primarily wound directed. Conversely, global elevation of Erk1/2 throughout sheets leads to stochastically directed collective migration throughout sheets. Our study highlights how the spatial patterning of leader cells (cells with elevated Erk1/2 activity) can influence the guidance of a collective group of cells during wound healing.


Cell Movement/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratinocytes/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing/physiology , Cell Line , Epithelial Cells , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , RNA Interference , RNA, Small Interfering , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Smad2 Protein/genetics , Smad3 Protein/genetics , T-Box Domain Proteins/antagonists & inhibitors
19.
Mol Med Rep ; 9(3): 899-903, 2014 Mar.
Article En | MEDLINE | ID: mdl-24425064

Th1 and Th17 cells are involved in the pathogenesis of rheumatoid arthritis (RA). T-bet, a Th1-specific transcription factor, appears to drive the maturation of Th1 and IFN-γ secretion. In the present study, we established the T-bet shRNA recombinant plasmid (p-T-shRNA) and explored its possible anti-inflammatory effect in a collagen-induced arthritis (CIA) model by local injection of plasmid vectors. For the initiation of CIA, DBA/1J mice were immunized with type II collagen (CII) in Freund's adjuvant and the CII-immunized mice were treated with p-T-shRNA. Levels of T-bet, IFN-γ, IL-17 and RORγt mRNA in splenocytes and synovial joints were measured by quantitative real-time PCR and T-bet expression in joint tissue was detected by immunohistochemistry staining. The intracellular IFN-γ and IL-17 were analyzed by flow cytometry (FCM). The results demonstrated that therapeutic administration on the local joints with p-T-shRNA significantly suppressed IFN-γ and IL-17 gene expression and improved the pathogenesis of arthritis in CIA mice, while administration of a plasmid expressing T-bet (pIRES-T-bet) accelerated the disease onset. Our study suggests that T-bet may be developed as a potential target for arthritis therapy.


Arthritis, Rheumatoid/metabolism , Down-Regulation , Interferon-gamma/genetics , Interleukin-17/genetics , RNA, Small Interfering/metabolism , T-Box Domain Proteins/metabolism , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/pathology , Collagen Type II , Immunohistochemistry , Inflammation/physiopathology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Joints/metabolism , Joints/pathology , Lymph Nodes/metabolism , Male , Mice , Mice, Inbred DBA , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , RNA Interference , Spleen/metabolism , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , Th1 Cells/immunology , Th1 Cells/metabolism
20.
Carcinogenesis ; 35(7): 1491-9, 2014 Jul.
Article En | MEDLINE | ID: mdl-24445144

Recent evidence suggests that the expression of brachyury is necessary for chordoma growth. However, the mechanism associated with brachyury-regulated cell growth is poorly understood. Fibroblast growth factor (FGF), a regulator of brachyury expression in normal tissue, may also play an important role in chordoma pathophysiology. Using a panel of chordoma cell lines, we explored the role of FGF signaling and brachyury in cell growth and survival. Western blots showed that all chordoma cell lines expressed fibroblast growth factor receptor 2 (FGFR2), FGFR3, mitogen-activated protein kinase kinase (MEK) and extracellular signal-regulated kinase (ERK), whereas no cell lines expressed FGFR1 and FGFR4. Results of enzyme-linked immunosorbent assay indicated that chordoma cells produced FGF2. Neutralization of FGF2 inhibited MEK/ERK phosphorylation, decreased brachyury expression and induced apoptosis while reducing cell growth. Activation of the FGFR/MEK/ERK/brachyury pathway by FGF2-initiated phosphorylation of FGFR substrate 2 (FRS2)-α (Tyr196) prevented apoptosis while promoting cell growth and epithelial-mesenchymal transition (EMT). Immunofluorescence staining showed that FGF2 promoted the translocation of phosphorylated ERK to the nucleus and increased brachyury expression. The selective inhibition of FGFR, MEK and ERK phosphorylation by PD173074, PD0325901 and PD184352, respectively, decreased brachyury expression, induced apoptosis, and inhibited cell growth and EMT. Moreover, knockdown of brachyury by small hairpin RNA reduced FGF2 secretion, inhibited FGFR/MEK/ERK phosphorylation and blocked the effects of FGF2 on cell growth, apoptosis and EMT. Those findings highlight that FGFR/MEK/ERK/brachyury pathway coordinately regulates chordoma cell growth and survival and may represent a novel chemotherapeutic target for chordoma.


Apoptosis , Cell Proliferation , Chordoma/pathology , Fetal Proteins/metabolism , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , T-Box Domain Proteins/metabolism , Blotting, Western , Cell Movement , Cells, Cultured , Chordoma/genetics , Chordoma/metabolism , Epithelial-Mesenchymal Transition , Fetal Proteins/antagonists & inhibitors , Fetal Proteins/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics
...