Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.693
Filter
1.
Genes (Basel) ; 14(10)2023 09 30.
Article in English | MEDLINE | ID: mdl-37895245

ABSTRACT

Alternative RNA splicing, a ubiquitous mechanism of gene regulation in eukaryotes, expands genome coding capacity and proteomic diversity. It has essential roles in all aspects of human physiology, including immunity. This review highlights the importance of RNA alternative splicing in regulating immune T cell function. We discuss how mutations that affect the alternative splicing of T cell factors can contribute to abnormal T cell function and ultimately lead to autoimmune diseases. We also explore the potential applications of strategies that target the alternative splicing changes of T cell factors. These strategies could help design therapeutic approaches to treat autoimmune disorders and improve immunotherapy.


Subject(s)
Alternative Splicing , RNA , Humans , Alternative Splicing/genetics , Proteomics , T-Lymphocytes , TCF Transcription Factors/genetics
2.
Exp Cell Res ; 429(2): 113646, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37271249

ABSTRACT

Wnt signaling drives nuclear translocation of ß-catenin and its subsequent association with the DNA-bound TCF/LEF transcription factors, which dictate target gene specificity by recognizing Wnt responsive elements across the genome. ß-Catenin target genes are therefore thought to be collectively activated upon Wnt pathway stimulation. However, this appears in contrast with the non-overlapping patterns of Wnt target gene expression in several contexts, including early mammalian embryogenesis. Here we followed Wnt target gene expression in human embryonic stem cells after Wnt pathway stimulation at a single-cell resolution. Cells changed gene expression program over time consistent with three key developmental events: i) loss of pluripotency, ii) induction of Wnt target genes, and iii) mesoderm specification. Contrary to our expectation, not all cells displayed equal amplitude of Wnt target gene activation; rather, they distributed in a continuum from strong to weak responders when ranked based on the expression of the target AXIN2. Moreover, high AXIN2 did not always correspond to elevated expression of other Wnt targets, which were activated in different proportions in individual cells. The uncoupling of Wnt target gene expression was also identified in single cell transcriptomics profiling of other Wnt-responding cell types, including HEK293T, murine developing forelimbs, and human colorectal cancer. Our finding underlines the necessity to identify additional mechanisms that explain the heterogeneity of the Wnt/ß-catenin-mediated transcriptional outputs in single cells.


Subject(s)
Wnt Signaling Pathway , beta Catenin , Mice , Humans , Animals , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism , HEK293 Cells , TCF Transcription Factors/metabolism , Gene Expression , Mammals/genetics
3.
Bioorg Med Chem ; 84: 117264, 2023 04 15.
Article in English | MEDLINE | ID: mdl-37003158

ABSTRACT

The Wnt/ß-catenin signaling pathway causes transcriptional activation through the interaction between ß-catenin and T cell-specific transcription factor (TCF) and regulates a wide variety of cellular responses, including proliferation, differentiation and cell motility. Excessive transcriptional activation of the Wnt/ß-catenin pathway is implicated in developing or exacerbating various cancers. We have recently reported that liver receptor homolog-1 (LRH-1)-derived peptides inhibit the ß-catenin/TCF interaction. In addition, we developed a cell-penetrating peptide (CPP)-conjugated LRH-1-derived peptide that inhibits the growth of colon cancer cells and specifically inhibits the Wnt/ß-catenin pathway. Nonetheless, the inhibitory activity of the CPP-conjugated LRH-1-derived peptide was unsatisfactory (ca. 20 µM), and improving the bioactivity of peptide inhibitors is required for their in vivo applications. In this study, we optimized the LRH-1-derived peptide using in silico design to enhance its activity further. The newly designed peptides showed binding affinity toward ß-catenin comparable to the parent peptide. In addition, the CPP-conjugated stapled peptide, Penetratin-st6, showed excellent inhibition (ca. 5 µM). Thus, the combination of in silico design by MOE and MD calculations has revealed that logical molecular design of PPI inhibitory peptides targeting ß-catenin is possible. This method can be also applied to the rational design of peptide-based inhibitors targeting other proteins.


Subject(s)
Cell-Penetrating Peptides , Wnt Signaling Pathway , beta Catenin , beta Catenin/metabolism , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Transcriptional Activation , Wnt Proteins/metabolism , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Computer Simulation
4.
J Biol Chem ; 299(1): 102735, 2023 01.
Article in English | MEDLINE | ID: mdl-36423688

ABSTRACT

Activation of the Wnt/ß-catenin pathway regulates gene expression by promoting the formation of a ß-catenin-T-cell factor (TCF) complex on target enhancers. In addition to TCFs, other transcription factors interact with the Wnt/ß-catenin pathway at different levels to produce tissue-specific patterns of Wnt target gene expression. The transcription factor SOX9 potently represses many Wnt target genes by downregulating ß-catenin protein levels. Here, we find using colony formation and cell growth assays that SOX9 surprisingly promotes the proliferation of Wnt-driven colorectal cancer (CRC) cells. In contrast to how it indirectly represses Wnt targets, SOX9 directly co-occupies and activates multiple Wnt-responsive enhancers in CRC cells. Our examination of the binding site grammar of these enhancers shows the presence of TCF and SOX9 binding sites that are necessary for transcriptional activation. In addition, we identify a physical interaction between the DNA-binding domains of TCFs and SOX9 and show that TCF-SOX9 interactions are important for target gene regulation and CRC cell growth. Our work demonstrates a highly context-dependent effect of SOX9 on Wnt targets, with the presence or absence of SOX9-binding sites on Wnt-regulated enhancers determining whether they are directly activated or indirectly repressed by SOX9.


Subject(s)
Colorectal Neoplasms , SOX9 Transcription Factor , TCF Transcription Factors , Wnt Signaling Pathway , Humans , beta Catenin/genetics , beta Catenin/metabolism , Colorectal Neoplasms/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , TCF Transcription Factors/metabolism , Transcriptional Activation
5.
Development ; 149(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36355069

ABSTRACT

Upon WNT/ß-catenin pathway activation, stabilized ß-catenin travels to the nucleus where it associates with the TCF/LEF transcription factors, constitutively bound to genomic Wnt-responsive elements (WREs), to activate target gene transcription. Discovering the binding profile of ß-catenin is therefore required to unambiguously assign direct targets of WNT signaling. Cleavage under targets and release using nuclease (CUT&RUN) has emerged as prime technique for mapping the binding profile of DNA-interacting proteins. Here, we present a modified version of CUT&RUN, named LoV-U (low volume and urea), that enables the robust and reproducible generation of ß-catenin binding profiles, uncovering direct WNT/ß-catenin target genes in human cells, as well as in cells isolated from developing mouse tissues. CUT&RUN-LoV-U outperforms original CUT&RUN when targeting co-factors that do not bind the DNA, can profile all classes of chromatin regulators and is well suited for simultaneous processing of several samples. We believe that the application of our protocol will allow the detection of the complex system of tissue-specific WNT/ß-catenin target genes, together with other non-DNA-binding transcriptional regulators that act downstream of ontogenetically fundamental signaling cascades.


Subject(s)
Transcription Factors , beta Catenin , Humans , Mice , Animals , beta Catenin/genetics , beta Catenin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , TCF Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , Endonucleases/metabolism , Genomics , Urea , Transcriptional Activation
6.
Sci Signal ; 15(757): eabn9009, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36282910

ABSTRACT

Neural crest cells (NCCs) are multipotent stem cells that can differentiate into multiple cell types, including the osteoblasts and chondrocytes, and constitute most of the craniofacial skeleton. Here, we show through in vitro and in vivo studies that the transcriptional regulators Yap and Taz have redundant functions as key determinants of the specification and differentiation of NCCs into osteoblasts or chondrocytes. Primary and cultured NCCs deficient in Yap and Taz switched from osteogenesis to chondrogenesis, and NCC-specific deficiency for Yap and Taz resulted in bone loss and ectopic cartilage in mice. Yap bound to the regulatory elements of key genes that govern osteogenesis and chondrogenesis in NCCs and directly regulated the expression of these genes, some of which also contained binding sites for the TCF/LEF transcription factors that interact with the Wnt effector ß-catenin. During differentiation of NCCs in vitro and NCC-derived osteogenesis in vivo, Yap and Taz promoted the expression of osteogenic genes such as Runx2 and Sp7 but repressed the expression of chondrogenic genes such as Sox9 and Col2a1. Furthermore, Yap and Taz interacted with ß-catenin in NCCs to coordinately promote osteoblast differentiation and repress chondrogenesis. Together, our data indicate that Yap and Taz promote osteogenesis in NCCs and prevent chondrogenesis, partly through interactions with the Wnt-ß-catenin pathway.


Subject(s)
Chondrogenesis , Osteogenesis , Animals , Mice , beta Catenin/genetics , Cell Differentiation , Chondrogenesis/genetics , Core Binding Factor Alpha 1 Subunit , Neural Crest , Osteogenesis/genetics , TCF Transcription Factors , YAP-Signaling Proteins/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
7.
ChemMedChem ; 17(24): e202200456, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36194001

ABSTRACT

The glycogen synthase kinase 3ß (GSK-3ß) is a ubiquitous enzyme that is a validated target for the development of potential therapeutics useful in several diseases including retinal degeneration. Aiming at developing an innovative class of allosteric inhibitors of GSK-3ß potentially useful for retinal degeneration, we explored the class of squaramides. The developed compounds (6 a-l) were obtained through a nontoxic one-pot synthetic protocol, which employs low-cost goods and avoids any purification step. Ethanol was used as the reaction solvent, simultaneously allowing the pure reaction products' recovery (by precipitation). Out of this set of squaramides, 6 j stood out, from computational and enzymatic converging data, as an ATP non-competitive inhibitor of GSK-3ß of micromolar potency. When engaged in cellular studies using retinal pigment epithelial cells (ARPE-19) transfected with a luciferase reporter gene under the control of T-cell factor/lymphoid enhancer factor (TCF/LEF) binding sites, 6 j was able to dose-dependently induce ß-catenin nuclear accumulation, as shown by the increased luciferase activity at a concentration of 2.5 µM.


Subject(s)
Epithelial Cells , Glycogen Synthase Kinase 3 beta , Quinine , Retinal Degeneration , TCF Transcription Factors , Humans , beta Catenin/metabolism , Epithelial Cells/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Luciferases/metabolism , Signal Transduction , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Quinine/analogs & derivatives , Quinine/chemical synthesis , Retinal Pigment Epithelium
8.
Bioorg Med Chem ; 73: 117021, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36198218

ABSTRACT

Wnt/ß-catenin pathway triggers the formation of a complex between ß-catenin and T cell-specific transcription factor (TCF), which induces transcriptional activation. Excessive transcriptional activation of this pathway is associated with the development, cause, and deterioration of various cancers. Therefore, the Wnt/ß-catenin pathway is an attractive drug target for cancer therapeutics and small molecule- and peptide-based protein-protein interaction (PPI) inhibitors have been developed. However, peptide-based PPI inhibitors generally have low cell-membrane permeability because of their large molecular size. To improve cell-membrane permeability, conjugating cell-penetrating peptides (CPPs) to PPI-inhibiting peptides is a useful method for developing intracellularly targeted PPI inhibitors. In this study, we focused on the interaction between ß-catenin and liver receptor homologue-1 (LRH-1) and designed and synthesized a series of LRH-1-derived peptides to develop inhibitors against Wnt/ß-catenin signaling. The results showed that a penetratin-conjugated LRH-1-derived peptide (Penetratin-st7) predominantly inhibited DLD-1 cell growth at 20 µM treatment via inhibition of the Wnt signaling pathway. This result suggests that Penetratin-st7 is one of promising PPI inhibitors between TCF and ß-catenin.


Subject(s)
Cell-Penetrating Peptides , Neoplasms , Cell-Penetrating Peptides/pharmacology , Humans , TCF Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
9.
Diagn Pathol ; 17(1): 69, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36096860

ABSTRACT

OBJECTIVES: To investigate lymphoid enhancer factor 1 (LEF-1) protein expression in medulloblastomas (MBs) and its correlation with molecular grouping of MBs. METHODS: Expressions of LEF-1 and ß-catenin were detected by immunohistochemistry, and molecular grouping was performed based on the NanoString and sequencing techniques for 30 MBs. RESULTS: By genetic defining, 3 MBs were WNT-activated, 11 were SHH-activated, 3 were in Group 3 and 13 in Group 4 respectively. Nuclear LEF-1 staining was found in 8 MBs using immunohistochemical method. Three out of 8 showed diffuse and strong nuclear LEF-1 staining which were proved to be WNT-activated genetically, while the other 5 MBs with focal staining were SHH-activated genetically. The expression of LEF-1 protein was significantly correlated with genetically defined WNT-activated MBs (P < 0.0001). We also found focal nuclear ß-catenin expression ( less than 1% of tumor cells) in 5 MBs. LEF-1 positivity was significantly correlated nuclear ß-catenin expression (p < 0.001). CONCLUSIONS: Immunohistochemical staining of LEF-1 can be used as a supplement for ß-catenin to diagnosis WNT-activated Medulloblastomas, when ß-catenin is difficult to recognize for its cytoplasm/membrane staining background. Diffuse nuclear staining of LEF-1 indicates WNT-activated MB.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Biomarkers , Cerebellar Neoplasms/diagnosis , Humans , Medulloblastoma/diagnosis , Medulloblastoma/genetics , Medulloblastoma/metabolism , Staining and Labeling , TCF Transcription Factors , beta Catenin/metabolism
10.
Birth Defects Res ; 114(16): 1056-1074, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36164276

ABSTRACT

BACKGROUND: Diabetes, which is characterized by an increase in blood glucose concentration, is accompanied by low bone turnover, increased fracture risk, and the formation of embryonic skeletal malformations. Yet, there are few studies elucidating the underlying alterations in signaling pathways leading to these osteogenic defects. We hypothesized here that bone formation deficiencies in a high glucose environment result from altered activity of beta-catenin (CTNNB1), a key contributor to osteogenic differentiation, dysregulation of which has also been implicated in the development of diabetes. METHODS: To test this hypothesis, we used a previously established embryonic stem cell (ESC) model of differentiation that mimics the diabetic environment of the developing embryo. We differentiated murine ESCs within osteogenic-inducing media containing either high (diabetic) or low (physiological) levels of D-glucose and performed time course analyses to study the influence of high glucose on early and late bone cell differentiation. RESULTS: Endpoint measures for osteogenic differentiation were reduced in a glucose-dependent manner and expression of precursor-specific markers altered at multiple time points. Furthermore, transcriptional activity of the lymphoid enhancer factor (LEF)/T cell factor (TCF) transcription factors during precursor formation stages was significantly elevated while levels of CTNNB1 complexed with Forkhead box O 3a (FOXO3a) declined. Modulation of AKT, a known upstream regulator of both LEF/TCF and FOXO3a, as well as CTNNB1 rescued some of the reductions in osteogenic output seen in the high glucose condition. CONCLUSIONS: Within our in vitro model, we found a clear involvement of LEF/TCF and FOXO3a signaling pathways in the regulation of osteogenic differentiation, which may account for the skeletal deficiencies found in newborns of diabetic mothers.


Subject(s)
Diabetes Mellitus , beta Catenin , Animals , Blood Glucose , Cell Differentiation , Embryonic Stem Cells/metabolism , Mice , Osteogenesis , Proto-Oncogene Proteins c-akt , TCF Transcription Factors/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism
11.
J Clin Lab Anal ; 36(10): e24678, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36036768

ABSTRACT

BACKGROUND: The inhibitor of ß-catenin and T-cell factor (ICAT) is a direct negative regulator of the canonical Wnt signaling pathway, which is an attractive therapeutic target for colorectal cancer (CRC). Accumulating evidence suggests that ICAT interacts with other proteins to exert additional functions, which are not yet fully elucidated. METHODS: The overexpression of ICAT of CRC cells was conducted by lentivirus infection and plasmids transfection and verified by quantitative real-time reverse transcription-polymerase chain reaction (real-time RT-PCR) and Western blotting. The effect of ICAT on the mobility of CRC cells was assessed by wound healing assay and transwell assay in vitro and lung metastasis in vivo. New candidate ICAT-interacting proteins were explored and verified using the STRING database, silver staining, co-immunoprecipitation mass spectrometry analysis (Co-IP/MS), and immunofluorescence (IF) staining analysis. RESULT: Inhibitor of ß-catenin and T-cell factor overexpression promoted in vitro cell migration and invasion and tumor metastasis in vivo. Co-IP/MS analysis and STRING database analyses revealed that junction plakoglobin (JUP), a homolog of ß-catenin, was involved in a novel protein interaction with ICAT. Furthermore, JUP downregulation impaired ICAT-induced migration and invasion of CRC cells. In addition, ICAT overexpression activated the NF-κB signaling pathway, which led to enhanced CRC cell migration and invasion. CONCLUSION: Inhibitor of ß-catenin and T-cell factor promoted CRC cell migration and invasion by interacting with JUP and the NF-κB signaling pathway. Thus, ICAT could be considered a protein diagnostic biomarker for predicting the metastatic ability of CRC.


Subject(s)
Colorectal Neoplasms , beta Catenin , Adaptor Proteins, Signal Transducing , Biomarkers , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/metabolism , Neoplasm Metastasis , TCF Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism , gamma Catenin/metabolism
12.
Cell Rep ; 40(8): 111247, 2022 08 23.
Article in English | MEDLINE | ID: mdl-36001974

ABSTRACT

WNT/ß-catenin signaling controls gene expression across biological contexts from development and stem cell homeostasis to diseases including cancer. How ß-catenin is recruited to distinct enhancers to activate context-specific transcription is unclear, given that most WNT/ß-catenin-responsive transcription is thought to be mediated by TCF/LEF transcription factors (TFs). With time-resolved multi-omic analyses, we show that SOX TFs can direct lineage-specific WNT-responsive transcription during the differentiation of human pluripotent stem cells (hPSCs) into definitive endoderm and neuromesodermal progenitors. We demonstrate that SOX17 and SOX2 are required to recruit ß-catenin to lineage-specific WNT-responsive enhancers, many of which are not occupied by TCFs. At TCF-independent enhancers, SOX TFs establish a permissive chromatin landscape and recruit a WNT-enhanceosome complex to activate SOX/ß-catenin-dependent transcription. Given that SOX TFs and the WNT pathway are critical for specification of most cell types, these results have broad mechanistic implications for the specificity of WNT responses across developmental and disease contexts.


Subject(s)
Pluripotent Stem Cells , beta Catenin , Humans , Pluripotent Stem Cells/metabolism , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , TCF Transcription Factors/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
13.
Eur J Haematol ; 109(5): 513-518, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35871391

ABSTRACT

PURPOSE: Extranodal natural killer (NK)/T-cell lymphoma (ENKTL) is not a uniform entity but consists of various disease subtypes associated with Epstein-Barr virus (EBV) infection. Lymphoid enhancer binding factor-1 (LEF-1), a member of the T-cell factor/LEF family of transcription factors, plays a significant role in NK-cell and T-cell development. We aimed to explore the expression of LEF-1 in ENKTL and evaluate the applicability of LEF-1 in the diagnosis of ENKTL. METHODS: The expression of LEF-1 was investigated in tissue samples harvested from patients with ENKTL by immunohistochemistry. RESULTS: LEF-1 staining was not observed in 85 of 88 ENKTL cases (97%). Eighty-six of the 88 cases (98%) were positive for CD56, whereas all the tested cases were negative for both CD16 and CD27. Of the cytotoxic T-cell-like features studied, 81 cases (92%) were negative for CD8, 85 of 88 cases (97%) were immunoreactive for the recombinant linker for activation of T cells (LAT), all (100%) were immunoreactive for TIA-1, 85 (97%) were immunoreactive for granzyme B and 65 (74%) were immunoreactive for perforin. CONCLUSION: Loss of LEF-1 expression is a highly specific diagnostic indicator of ENKTL.


Subject(s)
Epstein-Barr Virus Infections , Lymphoid Enhancer-Binding Factor 1/metabolism , Lymphoma, Extranodal NK-T-Cell , Epstein-Barr Virus Infections/complications , Granzymes , Herpesvirus 4, Human/genetics , Humans , Lymphoma, Extranodal NK-T-Cell/diagnosis , Lymphoma, Extranodal NK-T-Cell/pathology , Perforin , TCF Transcription Factors , Transcription Factors
14.
Elife ; 112022 07 14.
Article in English | MEDLINE | ID: mdl-35833630

ABSTRACT

The Wnt/ß-catenin signaling pathway is highly conserved throughout evolution, playing crucial roles in several developmental and pathological processes. Wnt ligands can act at a considerable distance from their sources and it is therefore necessary to examine not only the Wnt-producing but also the Wnt-receiving cells and tissues to fully appreciate the many functions of this pathway. To monitor Wnt activity, multiple tools have been designed which consist of multimerized Wnt signaling response elements (TCF/LEF binding sites) driving the expression of fluorescent reporter proteins (e.g. GFP, RFP) or of LacZ. The high stability of those reporters leads to a considerable accumulation in cells activating the pathway, thereby making them easily detectable. However, this makes them unsuitable to follow temporal changes of the pathway's activity during dynamic biological events. Even though fluorescent transcriptional reporters can be destabilized to shorten their half-lives, this dramatically reduces signal intensities, particularly when applied in vivo. To alleviate these issues, we developed two transgenic quail lines in which high copy number (12× or 16×) of the TCF/LEF binding sites drive the expression of destabilized GFP variants. Translational enhancer sequences derived from viral mRNAs were used to increase signal intensity and specificity. This resulted in transgenic lines efficient for the characterization of TCF/ß-catenin transcriptional dynamic activities during embryogenesis, including using in vivo imaging. Our analyses demonstrate the use of this transcriptional reporter to unveil novel aspects of Wnt signaling, thus opening new routes of investigation into the role of this pathway during amniote embryonic development.


Subject(s)
TCF Transcription Factors , beta Catenin , Animals , Animals, Genetically Modified , Embryonic Development , Quail/metabolism , TCF Transcription Factors/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
15.
Bioorg Med Chem ; 70: 116920, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35841828

ABSTRACT

The Wnt/ß-catenin signaling pathway is crucially involved in embryonic development, stem cell maintenance and tissue renewal. Hyperactivation of this pathway is associated with the development and progression of various types of cancers. The transcriptional coactivator ß-catenin represents a pivotal component of the pathway and its interaction with transcription factors of the TCF/LEF family is central to pathway activation. Inhibition of this crucial protein-protein interaction via direct targeting of ß-catenin is considered a promising strategy for the inactivation of oncogenic Wnt signaling. This review summarizes advances in the development of Wnt antagonists that have been shown to directly bind ß-catenin.


Subject(s)
TCF Transcription Factors , beta Catenin , Carcinogenesis , Humans , TCF Transcription Factors/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
16.
Biochem Biophys Res Commun ; 621: 88-93, 2022 09 17.
Article in English | MEDLINE | ID: mdl-35810596

ABSTRACT

The basic helix-loop-helix (bHLH) family is one of the most conserved transcription factor families that plays an important role in regulating cell growth, differentiation and tissue development. Typically, members of this family form homo- or heterodimers to recognize specific motifs and activate transcription. MyoD is a vital transcription factor that regulates muscle cell differentiation. However, it is necessary for MyoD to form a heterodimer with E-proteins to activate transcription. Even though the crystal structure of the MyoD homodimer has been determined, the structure of the MyoD heterodimer in complex with the E-box protein remains unclear. In this study, we determined the crystal structure of the bHLH domain of the MyoD-E47 heterodimer at 2.05 Å. Our structural analysis revealed that MyoD interacts with E47 through a hydrophobic interface. Moreover, we confirmed that heterodimerization could enhance the binding affinity of MyoD to E-box sequences. Our results provide new structural insights into the heterodimer of MyoD and E-box protein, suggesting the molecular mechanism of transcription activation of MyoD upon binding to E-box protein.


Subject(s)
DNA-Binding Proteins , MyoD Protein , DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , MyoD Protein/metabolism , Protein Binding , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factors/metabolism
17.
Cell Rep ; 39(7): 110826, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584684

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a global health concern, yearly resulting in 10 million new cases of active TB. Immunologic investigation of lung granulomas is essential for understanding host control of bacterial replication. Here, we identify and compare the pathological, cellular, and functional differences in granulomas at 4, 12, and 20 weeks post-infection in Chinese cynomolgus macaques. Original granulomas differ in transcription-factor expression within adaptive lymphocytes, with those at 12 weeks showing higher frequencies of CD8+T-bet+ T cells, while CD4+T-bet+ T cells increase at 20 weeks post-infection. The appearance of T-bet+ adaptive T cells at 12 and 20 weeks is coincident with a reduction in bacterial burden, suggesting their critical role in Mtb control. This study highlights the evolution of T cell responses within lung granulomas, suggesting that vaccines promoting the development and migration of T-bet+ T cells would enhance mycobacterial control.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , CD4-Positive T-Lymphocytes , Granuloma/pathology , Macaca fascicularis , T-Lymphocytes , TCF Transcription Factors
18.
Cells ; 11(9)2022 04 20.
Article in English | MEDLINE | ID: mdl-35563702

ABSTRACT

The transcription factors of the nuclear factor of activated T cell (NFAT) family play a crucial role in multiple aspects of T cell function. It has recently been reported that NFATs play an important role in the suppressive function of CD4+CD25+Foxp3+ regulatory T (Treg) cells. In this study, we have investigated the role of NFATs in the thymic development of Treg cells in mice. We show that NFAT factors are dispensable for the development of Foxp3+ Treg cells in the thymus but are critical for the maintenance of both the phenotype and survival of Treg cells in the thymus as well as in peripheral lymphoid organs. Specifically, the homeostasis of CD4+CD25+Foxp3+ but not the CD4+CD25-Foxp3+ fraction is severely perturbed when NFAT signaling is blocked, leading to a strongly reduced Treg population. We underscored this intriguing effect of NFAT on CD4+CD25+Foxp3+ Treg cells to the disruption of survival signals provided by interleukin 2 (IL-2). Accordingly, blocking Treg cell death by abolishing the activity of pro-apoptotic Bcl-2 family member Bim, compensated for the survival defects induced due to a lack of NFAT-IL-2-IL-2R signaling. Inhibition of NFAT activity led to a strong reduction in the number of Foxp3+ Treg cells; however, it did not influence the level of Foxp3 expression on an individual cell basis. In addition, we show a differential effect of IL-2 and IL-7 signaling on Foxp3+ Treg versus CD4+CD25- T cell development, again underlining the dispensability of NFAT signaling in the development, but not in the maintenance of Foxp3+ Treg cells.


Subject(s)
Interleukin-2 , T-Lymphocytes, Regulatory , Animals , Forkhead Transcription Factors/metabolism , Interleukin-2/metabolism , Mice , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , T-Lymphocytes, Regulatory/metabolism , TCF Transcription Factors/metabolism
19.
Front Immunol ; 13: 845488, 2022.
Article in English | MEDLINE | ID: mdl-35371057

ABSTRACT

T lymphocyte acute lymphoblastic leukemia (T-ALL) is a heterogeneous disease affecting T cells at multiple stages of their development and is characterized by frequent genomic alterations. The transcription factor LEF1 is inactivated through mutation in a subset of T-ALL cases but elevated LEF1 expression and activating mutations have also been identified in this disease. Here we show, in a murine model of T-ALL arising due to E2a inactivation, that the developmental timing of Lef1 mutation impacts its ability to function as a cooperative tumor suppressor or oncogene. T cell transformation in the presence of LEF1 allows leukemic cells to become addicted to its presence. In contrast, deletion prior to transformation both accelerates leukemogenesis and results in leukemic cells with altered expression of genes controlling receptor-signaling pathways. Our data demonstrate that the developmental timing of Lef1 mutations impact its apparent oncogenic or tumor suppressive characteristics and demonstrate the utility of mouse models for understanding the cooperation and consequence of mutational order in leukemogenesis.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Mice , Oncogenes , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , TCF Transcription Factors/genetics , Transcription Factors/metabolism
20.
Mol Med Rep ; 25(6)2022 Jun.
Article in English | MEDLINE | ID: mdl-35419615

ABSTRACT

The liver is a crucial digestive organ of humans and in charge of detoxification. Acute hepatic injury is an aggressive type of hepatic disease and its harmful effect cannot be ignored. The present study examined the role and mechanism of stress­associated endoplasmic reticulum protein 1 (SERP1) in acute hepatic injury. Mice were injected intraperitoneally with D­galactosamine/lipopolysaccharide (LPS) and rat hepatocytes were induced by LPS to establish an acute hepatic injury model. Tissue lesions were observed by H&E staining, and biomarkers of hepatic injury in the serum were examined. Western blotting, immunohistochemistry and reverse transcription­quantitative PCR were performed to assess SERP1 expression in tissues and hepatocytes. A SERP1 overexpression plasmid was constructed to evaluate the role of SERP1 in inflammation, apoptosis, endoplasmic reticulum stress (ERS) and the GSK3ß/ß­catenin/T­cell factor (TCF)/lymphoid enhancing factor (LEF) signaling pathway. In addition, a GSK3ß overexpression plasmid was constructed to investigate the role of GSK3ß/ß­catenin signal activation. Additionally, the present study investigated whether SERP1 regulated the endoplasmic reticulum via this pathway. In the present study, reliable animal and cellular hepatic injury models were established and verified. SERP1 overexpression reduced the expression of inflammatory factors, apoptosis­related proteins and ERS­related proteins, as well as the expression of proteins related to GSK3ß/ß­catenin/TCF/LEF signaling pathways. A GSK3ß overexpression plasmid was constructed and it was revealed that GSK3ß overexpression could reverse the effects of SERP1 overexpression in aforementioned aspects. This suggested that the activation of the GSK3ß/ß­catenin/TCF/LEF signaling pathway may be required for the regulation of SERP1. In conclusion, SERP1 regulated ERS via the GSK3ß/ß­catenin/TCF/LEF signaling pathway, thereby reducing inchoate acute hepatic injury.


Subject(s)
Endoplasmic Reticulum Stress , Membrane Proteins , beta Catenin , Animals , Apoptosis/genetics , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Lipopolysaccharides/pharmacology , Liver/metabolism , Membrane Proteins/genetics , Mice , Rats , Signal Transduction , TCF Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...