Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Immunol Cell Biol ; 102(3): 211-224, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38288547

ABSTRACT

CD4+ forkhead box P3 (FOXP3)+ regulatory T cells (Tregs) are essential in maintaining immune tolerance and suppressing excessive immune responses. Tregs also contribute to tissue repair processes distinct from their roles in immune suppression. For these reasons, Tregs are candidates for targeted therapies for inflammatory and autoimmune diseases, and in diseases where tissue damage occurs. MT-2 cells, an immortalized Treg-like cell line, offer a model to study Treg biology and their therapeutic potential. In the present study, we use clustered regularly interspaced palindromic repeats (CRISPR)-mediated knockdown of FOXP3 in MT-2 cells to understand the transcriptional and functional changes that occur when FOXP3 is lost and to compare MT-2 cells with primary human Tregs. We demonstrate that loss of FOXP3 affects the transcriptome of MT-2 cells and that FOXP3's potential downstream targets include a wide range of transcripts that participate in the cell cycle, promote growth and contribute to inflammatory processes, but do not wholly simulate previously reported human primary Treg transcriptional changes in the absence of FOXP3. We also demonstrate that FOXP3 regulates cell cycling and proliferation, expression of molecules crucial to Treg function and MT-2 cell-suppressive activities. Thus, MT-2 cells offer opportunities to address regulatory T-cell functions in vitro.


Subject(s)
Immunosuppression Therapy , T-Lymphocytes, Regulatory , Humans , Cell Line , Immune Tolerance , Forkhead Transcription Factors/metabolism
2.
Mol Syst Biol ; 14(9): e8140, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30177503

ABSTRACT

It is well known that clonal cells can make different fate decisions, but it is unclear whether these decisions are determined during, or before, a cell's own lifetime. Here, we engineered an endogenous fluorescent reporter for the pluripotency factor OCT4 to study the timing of differentiation decisions in human embryonic stem cells. By tracking single-cell OCT4 levels over multiple cell cycle generations, we found that the decision to differentiate is largely determined before the differentiation stimulus is presented and can be predicted by a cell's preexisting OCT4 signaling patterns. We further quantified how maternal OCT4 levels were transmitted to, and distributed between, daughter cells. As mother cells underwent division, newly established OCT4 levels in daughter cells rapidly became more predictive of final OCT4 expression status. These results imply that the choice between developmental cell fates can be largely predetermined at the time of cell birth through inheritance of a pluripotency factor.


Subject(s)
Cell Differentiation/genetics , Cell Tracking/methods , Human Embryonic Stem Cells/metabolism , Inheritance Patterns , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/metabolism , Bone Morphogenetic Protein 4/pharmacology , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , CRISPR-Cas Systems , Cell Cycle/genetics , Gene Expression Regulation , Genes, Reporter , Human Embryonic Stem Cells/cytology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Protein Engineering/methods , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Single-Cell Analysis/methods , Red Fluorescent Protein
3.
Nucleic Acids Res ; 40(14): 6725-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22561374

ABSTRACT

The transcription factor (TF) SOX2 is essential for the maintenance of pluripotency and self-renewal in embryonic stem cells. In addition to its normal stem cell function, SOX2 over-expression is associated with cancer development. The ability to selectively target this and other oncogenic TFs in cells, however, remains a significant challenge due to the 'undruggable' characteristics of these molecules. Here, we employ a zinc finger (ZF)-based artificial TF (ATF) approach to selectively suppress SOX2 gene expression in cancer cells. We engineered four different proteins each composed of 6ZF arrays designed to bind 18 bp sites in the SOX2 promoter and enhancer region, which controls SOX2 methylation. The 6ZF domains were linked to the Kruppel Associated Box (SKD) repressor domain. Three engineered proteins were able to bind their endogenous target sites and effectively suppress SOX2 expression (up to 95% repression efficiencies) in breast cancer cells. Targeted down-regulation of SOX2 expression resulted in decreased tumor cell proliferation and colony formation in these cells. Furthermore, induced expression of an ATF in a mouse model inhibited breast cancer cell growth. Collectively, these findings demonstrate the effectiveness and therapeutic potential of engineered ATFs to mediate potent and long-lasting down-regulation of oncogenic TF expression in cancer cells.


Subject(s)
Breast Neoplasms/therapy , Gene Silencing , SOXB1 Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , DNA/metabolism , Female , Humans , Mice , Protein Engineering , SOXB1 Transcription Factors/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
4.
Metabolites ; 14(2)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38392976

ABSTRACT

The role of the sodium citrate transporter (NaCT) SLC13A5 is multifaceted and context-dependent. While aberrant dysfunction leads to neonatal epilepsy, its therapeutic inhibition protects against metabolic disease. Notably, insights regarding the cellular and molecular mechanisms underlying these phenomena are limited due to the intricacy and complexity of the latent human physiology, which is poorly captured by existing animal models. This review explores innovative technologies aimed at bridging such a knowledge gap. First, I provide an overview of SLC13A5 variants in the context of human disease and the specific cell types where the expression of the transporter has been observed. Next, I discuss current technologies for generating patient-specific induced pluripotent stem cells (iPSCs) and their inherent advantages and limitations, followed by a summary of the methods for differentiating iPSCs into neurons, hepatocytes, and organoids. Finally, I explore the relevance of these cellular models as platforms for delving into the intricate molecular and cellular mechanisms underlying SLC13A5-related disorders.

5.
Nat Commun ; 15(1): 2765, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553455

ABSTRACT

Single-cell technologies can measure the expression of thousands of molecular features in individual cells undergoing dynamic biological processes. While examining cells along a computationally-ordered pseudotime trajectory can reveal how changes in gene or protein expression impact cell fate, identifying such dynamic features is challenging due to the inherent noise in single-cell data. Here, we present DELVE, an unsupervised feature selection method for identifying a representative subset of molecular features which robustly recapitulate cellular trajectories. In contrast to previous work, DELVE uses a bottom-up approach to mitigate the effects of confounding sources of variation, and instead models cell states from dynamic gene or protein modules based on core regulatory complexes. Using simulations, single-cell RNA sequencing, and iterative immunofluorescence imaging data in the context of cell cycle and cellular differentiation, we demonstrate how DELVE selects features that better define cell-types and cell-type transitions. DELVE is available as an open-source python package: https://github.com/jranek/delve .


Subject(s)
Gene Expression Profiling , Software , Gene Expression Profiling/methods , Single-Cell Analysis/methods , Cell Differentiation , Cell Cycle/genetics , Sequence Analysis, RNA/methods
6.
J Biol Chem ; 287(35): 29873-86, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22782891

ABSTRACT

Ovarian cancer is the leading cause of death among gynecological malignancies. It is detected at late stages when the disease is spread through the abdominal cavity in a condition known as peritoneal carcinomatosis. Thus, there is an urgent need to develop novel therapeutic interventions to target advanced stages of ovarian cancer. Mammary serine protease inhibitor (Maspin) represents an important metastasis suppressor initially identified in breast cancer. Herein we have generated a sequence-specific zinc finger artificial transcription factor (ATF) to up-regulate the Maspin promoter in aggressive ovarian cancer cell lines and to interrogate the therapeutic potential of Maspin in ovarian cancer. We found that although Maspin was expressed in some primary ovarian tumors, the promoter was epigenetically silenced in cell lines derived from ascites. Transduction of the ATF in MOVCAR 5009 cells derived from ascitic cultures of a TgMISIIR-TAg mouse model of ovarian cancer resulted in tumor cell growth inhibition, impaired cell invasion, and severe disruption of actin cytoskeleton. Systemic delivery of lipid-protamine-RNA nanoparticles encapsulating a chemically modified ATF mRNA resulted in inhibition of ovarian cancer cell growth in nude mice accompanied with Maspin re-expression in the treated tumors. Gene expression microarrays of ATF-transduced cells revealed an exceptional specificity for the Maspin promoter. These analyses identified novel targets co-regulated with Maspin in human short-term cultures derived from ascites, such as TSPAN12, that could mediate the anti-metastatic phenotype of the ATF. Our work outlined the first targeted, non-viral delivery of ATFs into tumors with potential clinical applications for metastatic ovarian cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms, Experimental/metabolism , Ovarian Neoplasms/metabolism , Transcription Factors/biosynthesis , Zinc Fingers , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Cell Line, Tumor , Female , Mice , Mice, Transgenic , Neoplasm Invasiveness , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Serpins/biosynthesis , Tetraspanins/genetics , Tetraspanins/metabolism , Transcription Factors/genetics , Transcriptome
7.
Sci Rep ; 13(1): 6118, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37059819

ABSTRACT

Tau tubulin kinase 1 and 2 (TTBK1/2) are highly homologous kinases that are expressed and mediate disease-relevant pathways predominantly in the brain. Distinct roles for TTBK1 and TTBK2 have been delineated. While efforts have been devoted to characterizing the impact of TTBK1 inhibition in diseases like Alzheimer's disease and amyotrophic lateral sclerosis, TTBK2 inhibition has been less explored. TTBK2 serves a critical function during cilia assembly. Given the biological importance of these kinases, we designed a targeted library from which we identified several chemical tools that engage TTBK1 and TTBK2 in cells and inhibit their downstream signaling. Indolyl pyrimidinamine 10 significantly reduced the expression of primary cilia on the surface of human induced pluripotent stem cells (iPSCs). Furthermore, analog 10 phenocopies TTBK2 knockout in iPSCs, confirming a role for TTBK2 in ciliogenesis.


Subject(s)
Induced Pluripotent Stem Cells , Tubulin , Humans , Tubulin/metabolism , Induced Pluripotent Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
8.
Noncoding RNA ; 9(6)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37987362

ABSTRACT

BACKGROUND: Currently, most of the research on breast cancer has been carried out in conventional two-dimensional (2D) cell cultures due to its practical benefits, however, the three-dimensional (3D) cell culture is becoming the model of choice in cancer research because it allows cell-cell and cell-extracellular matrix (ECM) interactions, mimicking the native microenvironment of tumors in vivo. METHODS: In this work, we evaluated the effect of 3D cell organization on the expression pattern of miRNAs (by Small-RNAseq) and mRNAs (by microarrays) in the breast cancer SKBR3 cell line and analyzed the biological processes and signaling pathways regulated by the differentially expressed protein-coding genes (DE-mRNAs) and miRNAs (DE-microRNAs) found in the organoids. RESULTS: We obtained well-defined cell-aggregated organoids with a grape cluster-like morphology with a size up to 9.2 × 105 µm3. The transcriptomic assays showed that cell growth in organoids significantly affected (all p < 0.01) the gene expression patterns of both miRNAs, and mRNAs, finding 20 upregulated and 19 downregulated DE-microRNAs, as well as 49 upregulated and 123 downregulated DE-mRNAs. In silico analysis showed that a subset of 11 upregulated DE-microRNAs target 70 downregulated DE-mRNAs. These genes are involved in 150 gene ontology (GO) biological processes such as regulation of cell morphogenesis, regulation of cell shape, regulation of canonical Wnt signaling pathway, morphogenesis of epithelium, regulation of cytoskeleton organization, as well as in the MAPK and AGE-RAGE signaling KEGG-pathways. Interestingly, hsa-mir-122-5p (Fold Change (FC) = 15.4), hsa-mir-369-3p (FC = 11.4), and hsa-mir-10b-5p (FC = 20.1) regulated up to 81% of the 70 downregulated DE-mRNAs. CONCLUSION: The organotypic 3D cell-organization architecture of breast cancer SKBR3 cells impacts the expression pattern of the miRNAs-mRNAs network mainly through overexpression of hsa-mir-122-5p, hsa-mir-369-3p, and hsa-mir-10b-5p. All these findings suggest that the interaction between cell-cell and cell-ECM as well as the change in the culture architecture impacts gene expression, and, therefore, support the pertinence of migrating breast cancer research from conventional cultures to 3D models.

9.
Elife ; 122023 10 11.
Article in English | MEDLINE | ID: mdl-37819053

ABSTRACT

TDP-43 proteinopathies including frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS) are neurodegenerative disorders characterized by aggregation and mislocalization of the nucleic acid-binding protein TDP-43 and subsequent neuronal dysfunction. Here, we developed endogenous models of sporadic TDP-43 proteinopathy based on the principle that disease-associated TDP-43 acetylation at lysine 145 (K145) alters TDP-43 conformation, impairs RNA-binding capacity, and induces downstream mis-regulation of target genes. Expression of acetylation-mimic TDP-43K145Q resulted in stress-induced nuclear TDP-43 foci and loss of TDP-43 function in primary mouse and human-induced pluripotent stem cell (hiPSC)-derived cortical neurons. Mice harboring the TDP-43K145Q mutation recapitulated key hallmarks of FTLD, including progressive TDP-43 phosphorylation and insolubility, TDP-43 mis-localization, transcriptomic and splicing alterations, and cognitive dysfunction. Our study supports a model in which TDP-43 acetylation drives neuronal dysfunction and cognitive decline through aberrant splicing and transcription of critical genes that regulate synaptic plasticity and stress response signaling. The neurodegenerative cascade initiated by TDP-43 acetylation recapitulates many aspects of human FTLD and provides a new paradigm to further interrogate TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis , Cognitive Dysfunction , Frontotemporal Dementia , Frontotemporal Lobar Degeneration , TDP-43 Proteinopathies , Humans , Animals , Mice , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Disease Models, Animal , RNA
10.
J Gastrointest Oncol ; 14(4): 1735-1745, 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37720443

ABSTRACT

Background: Colorectal cancer (CRC) is a leading cause of death worldwide. SRY-box transcription factor 9 (SOX9) participates in organogenesis and cell differentiation in normal tissues but has been involved in carcinogenesis development. Cancer stem cells (CSCs) are a small population of cells present in solid tumors that contribute to increased tumor heterogeneity, metastasis, chemoresistance, and relapse. CSCs have properties such as self-renewal and differentiation, which can be modulated by many factors. Currently, the role of SOX9 in the maintenance of the stem phenotype has not been well elucidated, thus, in this work we evaluated the effect of the absence of SOX9 in the stem phenotype of CRC cells. Methods: We knockout (KO) SOX9 in the undifferentiated CRC cell line HCT116 and evaluated their stemness properties using sphere formation assay, differentiation assay, and immunophenotyping. Results: SOX9-KO affected the epithelial morphology of HCT116 cells and stemness characteristics such as its pluripotency signature with the increase of SOX2 as a compensatory mechanism to induce SOX9 expression, the increase of KLF4 as a differentiation feature, as well as the inhibition of the stem cell markers CD44 and CD73. In addition, SOX9-KO cells gain the epithelial-mesenchymal transition (EMT) phenotype with a significant upregulation of CDH2. Furthermore, our results showed a remarkable effect on first- and second-sphere formation, being SOX9-KO cells less capable of forming high-size-resistant spheres. Nevertheless, CSCs surface markers were not affected during the differentiation assay. Conclusions: Collectively, our findings supply evidence that SOX9 promotes the maintenance of stemness properties in CRC-CSCs.

11.
Toxicol In Vitro ; 83: 105412, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35688329

ABSTRACT

The liver is a pivotal organ regulating critical developmental stages of fetal metabolism and detoxification. Though numerous studies have evaluated links between prenatal/perinatal exposures and adverse health outcomes in the developing fetus, the central role of liver to health disruptions resulting from these exposures remains understudied, especially concerning early development and later-in-life health outcomes. While numerous in vitro methods for evaluating liver toxicity have been established, the use of iPSC-derived hepatocytes appears to be particularly well suited to contribute to this critical research gap due to their potential to model a diverse range of disease phenotypes and different stages of liver development. The following key aspects are reviewed: (1) an introduction to developmental liver toxicity; (2) an introduction to embryonic and induced pluripotent stem cell models; (3) methods and challenges for deriving liver cells from stem cells; and (4) applications for iPSC-derived hepatocytes to evaluate liver developmental stages and their associated responses to insults. We conclude that iPSC-derived hepatocytes have great potential for informing liver toxicity and underlying disease mechanisms via the generation of patient-specific iPSCs; implementing large-scale drug and chemical screening; evaluating general biological responses as a potential surrogate target cell; and evaluating inter-individual disease susceptibility and response variability.


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Female , Hepatocytes/drug effects , Humans , Induced Pluripotent Stem Cells/metabolism , Liver/drug effects , Liver/growth & development , Pregnancy
12.
Breast Cancer Res ; 13(5): R94, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21952072

ABSTRACT

INTRODUCTION: Tumor-initiating cells (TIC) are being extensively studied for their role in tumor etiology, maintenance and resistance to treatment. The isolation of TICs has been limited by the scarcity of this population in the tissue of origin and because the molecular signatures that characterize these cells are not well understood. Herein, we describe the generation of TIC-like cell lines by ectopic expression of the OCT4 transcription factor (TF) in primary breast cell preparations. METHODS: OCT4 cDNA was over-expressed in four different primary human mammary epithelial (HMEC) breast cell preparations from reduction mammoplasty donors. OCT4-transduced breast cells (OTBCs) generated colonies (frequency ~0.01%) in self-renewal conditions (feeder cultures in human embryonic stem cell media). Differentiation assays, immunofluorescence, immunohistochemistry, and flow cytometry were performed to investigate the cell of origin of OTBCs. Serial dilutions of OTBCs were injected in nude mice to address their tumorigenic capabilities. Gene expression microarrays were performed in OTBCs, and the role of downstream targets of OCT4 in maintaining self-renewal was investigated by knock-down experiments. RESULTS: OTBCs overcame senescence, overexpressed telomerase, and down-regulated p16INK4A. In differentiation conditions, OTBCs generated populations of both myoepithelial and luminal cells at low frequency, suggesting that the cell of origin of some OTBCs was a bi-potent stem cell. Injection of OTBCs in nude mice generated poorly differentiated breast carcinomas with colonization capabilities. Gene expression microarrays of OTBC lines revealed a gene signature that was over-represented in the claudin-low molecular subtype of breast cancer. Lastly, siRNA-mediated knockdown of OCT4 or downstream embryonic targets of OCT4, such as NANOG and ZIC1, suppressed the ability of OTBCs to self-renew. CONCLUSIONS: Transduction of OCT4 in normal breast preparations led to the generation of cell lines possessing tumor-initiating and colonization capabilities. These cells developed high-grade, poorly differentiated breast carcinomas in nude mice. Genome-wide analysis of OTBCs outlined an embryonic TF circuitry that could be operative in TICs, resulting in up-regulation of oncogenes and loss of tumor suppressive functions. These OTBCs represent a patient-specific model system for the discovery of novel oncogenic targets in claudin-low tumors.


Subject(s)
Cell Transformation, Neoplastic/pathology , Mammary Glands, Human/pathology , Octamer Transcription Factor-3/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Cell Differentiation/genetics , Cell Lineage , Cell Transformation, Neoplastic/genetics , Cellular Senescence/genetics , Epithelial Cells , Female , Gene Expression Regulation , Genes, Tumor Suppressor , Homeodomain Proteins/genetics , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mesoderm/physiology , Mice , Mice, Nude , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering , SOXB1 Transcription Factors/genetics , Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic , Tumor Cells, Cultured
13.
Stem Cell Res ; 54: 102421, 2021 07.
Article in English | MEDLINE | ID: mdl-34130156

ABSTRACT

The generation of induced pluripotent stem cells (iPSCs) from healthy individuals is an invaluable resource as reference control in disease modeling and drug discovery. This paper details the reprogramming of peripheral blood mononuclear cells (PBMCs) isolated from a healthy 27 years-old male using non-integration technology. The derived iPSCs displayed typical pluripotent stem cell morphology, the capacity to differentiate into the three germ layers, and normal karyotype. This iPSC line will be used as a reference control to study the Cerebral Cavernous Malformation disease mechanism.


Subject(s)
Hemangioma, Cavernous, Central Nervous System , Induced Pluripotent Stem Cells , Adult , Cell Differentiation , Cellular Reprogramming , Germ Layers , Hemangioma, Cavernous, Central Nervous System/genetics , Humans , Leukocytes, Mononuclear , Male
14.
Stem Cell Res ; 49: 102015, 2020 12.
Article in English | MEDLINE | ID: mdl-33038744

ABSTRACT

Induced pluripotent stem cells (iPSCs) generated from young, healthy individuals are valuable tools for investigating molecular disease mechanisms during the early development of the brain vasculature. We generated an iPSC line from peripheral blood mononuclear cells (PBMCs) isolated from a healthy 13-yeard old female donor using the Sendai virus. The iPSCs differentiated into endothelial cells, astrocytes, and neurons. This iPSC line can serve as a healthy reference control for comparative studies in drug development and modeling the early onset of Cerebral Cavernous Malformation (CCM).


Subject(s)
Induced Pluripotent Stem Cells , Cell Differentiation , Endothelial Cells , Female , Humans , Leukocytes, Mononuclear , Sendai virus/genetics
15.
Bio Protoc ; 10(20): e3788, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33659443

ABSTRACT

Induced pluripotent stem cells (iPSCs) are genetically reprogrammed somatic cells that exhibit features identical to those of embryonic stem cells (ESCs). Multiple approaches are available to derive iPSCs, among which the Sendai virus is the most effective at reprogramming different cell types. Here we describe a rapid, efficient, safe, and reliable approach to reprogram human fibroblasts into iPSCs that are compatible with future iPSCs uses such as genome editing and differentiation to a transplantable cell type.

16.
Genes (Basel) ; 11(11)2020 11 12.
Article in English | MEDLINE | ID: mdl-33198240

ABSTRACT

DNA methyltransferases (DNMTs) play an essential role in DNA methylation and transcriptional regulation in the genome. DNMTs, along with other poorly studied elements, modulate the dynamic DNA methylation patterns of embryonic and adult cells. We summarize the current knowledge on the molecular mechanism of DNMTs' functional targeting to maintain genome-wide DNA methylation patterns. We focus on DNMTs' intrinsic characteristics, transcriptional regulation, and post-transcriptional modifications. Furthermore, we focus special attention on the DNMTs' specificity for target sites, including key cis-regulatory factors such as CpG content, common motifs, transcription factors (TF) binding sites, lncRNAs, and histone marks to regulate DNA methylation. We also review how complexes of DNMTs/TFs or DNMTs/lncRNAs are involved in DNA methylation in specific genome regions. Understanding these processes is essential because the spatiotemporal regulation of DNA methylation modulates gene expression in health and disease.


Subject(s)
DNA Methylation , DNA Modification Methylases/genetics , Histone Code/genetics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , 5-Methylcytosine/metabolism , Animals , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Modification Methylases/metabolism , Humans , Promoter Regions, Genetic
17.
Mol Cancer Ther ; 7(5): 1080-90, 2008 May.
Article in English | MEDLINE | ID: mdl-18483297

ABSTRACT

Mammary serine protease inhibitor (maspin) is an important tumor suppressor gene whose expression is associated not only with tumor growth inhibition but also with decreased angiogenesis and metastasis. Maspin expression is down-regulated in metastatic tumors by epigenetic mechanisms, including aberrant promoter hypermethylation. We have constructed artificial transcription factors (ATFs) as novel therapeutic effectors able to bind 18-bp sites in the maspin promoter and reactivate maspin expression in cell lines that harbor an epigenetically silenced promoter. In this article, we have investigated the influence of epigenetic modifications on ATF-mediated regulation of maspin by challenging MDA-MB-231 breast cancer cells, comprising a methylated maspin promoter, with different doses of ATFs and chromatin remodeling drugs: the methyltransferase inhibitor 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor suberoylanilide hydroxamic acid. We found that the ATFs synergized with both inhibitors in reactivating endogenous maspin expression. The strongest synergy was observed with the triple treatment ATF-126 + 5-aza-2'-deoxycytidine + suberoylanilide hydroxamic acid, in which the tumor suppressor was reactivated by 600-fold. Furthermore, this combination inhibited tumor cell proliferation by 95%. Our data suggest that ATFs enhance the efficiency of chromatin remodeling drugs in reactivating silenced tumor suppressors. Our results document the power of a novel therapeutic approach that combines both epigenetic and genetic (sequence-specific ATFs) strategies to reactivate specifically silenced regions of the genome and reprogram cellular phenotypes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chromatin Assembly and Disassembly/drug effects , Gene Silencing , Genes, Tumor Suppressor , Serine Proteinase Inhibitors/genetics , Serpins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Azacitidine/analogs & derivatives , Azacitidine/metabolism , Decitabine , Humans , Hydroxamic Acids/metabolism , Models, Biological , Serine Proteinase Inhibitors/metabolism , Serpins/metabolism , Tumor Cells, Cultured , Vorinostat
18.
Mol Cancer Res ; 17(7): 1503-1518, 2019 07.
Article in English | MEDLINE | ID: mdl-31000582

ABSTRACT

Screening of an inhibitor library targeting kinases and epigenetic regulators identified several molecules having antiproliferative synergy with extraterminal domain (BET) bromodomain (BD) inhibitors (JQ1, OTX015) in triple-negative breast cancer (TNBC). GSK2801, an inhibitor of BAZ2A/B BDs, of the imitation switch chromatin remodeling complexes, and BRD9, of the SWI/SNF complex, demonstrated synergy independent of BRD4 control of P-TEFb-mediated pause-release of RNA polymerase II. GSK2801 or RNAi knockdown of BAZ2A/B with JQ1 selectively displaced BRD2 at promoters/enhancers of ETS-regulated genes. Additional displacement of BRD2 from rDNA in the nucleolus coincided with decreased 45S rRNA, revealing a function of BRD2 in regulating RNA polymerase I transcription. In 2D cultures, enhanced displacement of BRD2 from chromatin by combination drug treatment induced senescence. In spheroid cultures, combination treatment induced cleaved caspase-3 and cleaved PARP characteristic of apoptosis in tumor cells. Thus, GSK2801 blocks BRD2-driven transcription in combination with BET inhibitor and induces apoptosis of TNBC. IMPLICATIONS: Synergistic inhibition of BDs encoded in BAZ2A/B, BRD9, and BET proteins induces apoptosis of TNBC by a combinatorial suppression of ribosomal DNA transcription and ETS-regulated genes.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Azepines/pharmacology , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Drug Synergism , Female , Humans , Indolizines/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Promoter Regions, Genetic/drug effects , RNA Polymerase II/genetics , RNA, Ribosomal/genetics , Receptors, Cell Surface/antagonists & inhibitors , Sulfones/pharmacology , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
19.
Elife ; 82019 11 04.
Article in English | MEDLINE | ID: mdl-31682229

ABSTRACT

Alexander disease (AxD) is a fatal neurodegenerative disorder caused by mutations in glial fibrillary acidic protein (GFAP), which supports the structural integrity of astrocytes. Over 70 GFAP missense mutations cause AxD, but the mechanism linking different mutations to disease-relevant phenotypes remains unknown. We used AxD patient brain tissue and induced pluripotent stem cell (iPSC)-derived astrocytes to investigate the hypothesis that AxD-causing mutations perturb key post-translational modifications (PTMs) on GFAP. Our findings reveal selective phosphorylation of GFAP-Ser13 in patients who died young, independently of the mutation they carried. AxD iPSC-astrocytes accumulated pSer13-GFAP in cytoplasmic aggregates within deep nuclear invaginations, resembling the hallmark Rosenthal fibers observed in vivo. Ser13 phosphorylation facilitated GFAP aggregation and was associated with increased GFAP proteolysis by caspase-6. Furthermore, caspase-6 was selectively expressed in young AxD patients, and correlated with the presence of cleaved GFAP. We reveal a novel PTM signature linking different GFAP mutations in infantile AxD.


Subject(s)
Alexander Disease/metabolism , Biomarkers/metabolism , Caspases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Adult , Alexander Disease/diagnosis , Alexander Disease/genetics , Astrocytes/metabolism , Binding Sites/genetics , Brain/metabolism , Brain/pathology , Cell Line , Glial Fibrillary Acidic Protein/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Infant , Intermediate Filaments/metabolism , Mutation , Phosphorylation , Proteolysis , Severity of Illness Index
20.
Comb Chem High Throughput Screen ; 11(2): 146-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18336208

ABSTRACT

Artificial Transcription Factors (ATFs) are engineered DNA-binding proteins designed to bind specific sequences of DNA. ATFs made of Zinc Finger (ZF) domains have been developed to regulate specific genes and phenotypes both in cells and whole organisms. Recently, an emerging application of engineered DNA-binding domains include the specific editing of the genome, the ability to specifically cut, recombine, modify DNA and image protein-nucleic acid interactions in living cells. In this review we will summarize the techniques used for the rational design, screening and functional selection of ZF proteins in mammalian cell systems and their applications in areas of biotechnology, functional genomics and molecular therapeutics. The in vivo specificity of the engineered ATFs will be discussed, with particular emphasis on epigenetic modifications influencing ATF-DNA interactions.


Subject(s)
Chromatin/genetics , Gene Expression Regulation/genetics , Gene Targeting , Transcription Factors/genetics , Animals , DNA/genetics , Drug Design , Humans , Nucleic Acid Conformation , Phenotype , Protein Engineering , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL