Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Mol Oncol ; 17(10): 2090-2108, 2023 10.
Article En | MEDLINE | ID: mdl-37518985

Promyelocytic leukemia protein (PML) modulates diverse cell functions that contribute to both tumor suppressor and pro-oncogenic effects, depending on the cellular context. We show here that PML knockdown (KD) in MDA-MB-231, but not MCF7, breast cancer cells, prolonged stem-cell-like survival, and increased cell proliferation and migration, which is in line with gene-enrichment results from their RNA sequencing analysis. Of note, increased migration was accompanied by higher levels of the epithelial-mesenchymal transition (EMT) regulator Twist-related protein 2 (TWIST2). We showed here that PML binds to TWIST2 via its basic helix-loop-helix (bHLH) region and functionally interferes with the suppression of the epithelial target of TWIST2, CD24. In addition, PML ablation in MDA-MB-231 cells led to higher protein levels of hypoxia-inducible factor 1-alpha (HIF1a), resulting in a higher cell hypoxic response. Functionally, PML directly suppressed the induction of the HIF1a target gene vascular endothelial growth factor A (VEGFa). In line with these results, tumor xenografts of MDA-MB-231 PML-KD cells had enhanced aggressive properties, including higher microvessel density, faster local growth, and higher metastatic ability, with a preference for lung. Collectively, PML suppresses the cancer aggressive behavior by multiple mechanisms that impede both the HIF-hypoxia-angiogenic and EMT pathways.


Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Promyelocytic Leukemia Protein/genetics , Vascular Endothelial Growth Factor A , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Transcription Factors/genetics , Cell Movement
2.
Biomolecules ; 12(2)2022 02 15.
Article En | MEDLINE | ID: mdl-35204809

Protein-based carriers are promising vehicles for the intracellular delivery of therapeutics. In this study, we designed and studied adenovirus protein fiber constructs with potential applications as carriers for the delivery of protein and nanoparticle cargoes. We used as a basic structural framework the fibrous shaft segment of the adenovirus fiber protein comprising of residues 61-392, connected to the fibritin foldon trimerization motif at the C-terminal end. A fourteen-amino-acid biotinylation sequence was inserted immediately after the N-terminal, His-tagged end of the construct in order to enable the attachment of a biotin moiety in vivo. We report herein that this His-tag biotinylated construct folds into thermally and protease-stable fibrous nanorods that can be internalized into cells and are not cytotoxic. Moreover, they can bind to proteins and nanoparticles through the biotin-streptavidin interaction and mediate their delivery to cells. We demonstrate that streptavidin-conjugated gold nanoparticles can be transported into NIH3T3 fibroblast and HeLa cancer cell lines. Furthermore, two streptavidin-conjugated model proteins, alkaline phosphatase and horseradish peroxidase can be delivered into the cell cytoplasm in their enzymatically active form. This work is aimed at establishing the proof-of-principle for the rational engineering of diverse functionalities onto the initial protein structural framework and the use of adenovirus fiber-based proteins as nanorods for the delivery of nanoparticles and model proteins. These constructs could constitute a stepping stone for the development of multifunctional and modular fibrous nanorod platforms that can be tailored to applications at the sequence level.


Nanoparticle Drug Delivery System , Viral Proteins , Adenoviridae/chemistry , Animals , Biotin/chemistry , Biotin/metabolism , Gold/chemistry , HeLa Cells , Humans , Metal Nanoparticles/chemistry , Mice , NIH 3T3 Cells , Streptavidin/chemistry , Viral Proteins/chemistry
3.
Int J Dev Biol ; 66(1-2-3): 85-95, 2022.
Article En | MEDLINE | ID: mdl-34881785

The promyelocytic leukemia protein (PML) is the core organizer of cognate nuclear bodies (PML-NBs). Through physical interaction or modification of diverse protein clients, PML-NBs regulate a multitude of - often antithetical- biological processes such as antiviral and stress response, inhibition of cell proliferation and autophagy, and promotion of apoptosis or senescence. Although PML was originally recognized as a tumor-suppressive factor, more recent studies have revealed a "double-faced" agent role for PML. Indeed, PML displayed tumor cell pro-survival and pro-migratory functions via inhibition of migration suppressing molecules or promotion of transforming growth factor beta (TGF-ß) mediated Epithelial-Mesenchymal Transition (EMT) that may promote cancer cell dissemination. In this line, PML was found to correlate with poor patient prognosis in distinct tumor contexts. Furthermore, in the last decade, a number of publications have implicated PML in the physiology of normal or cancer stem cells (CSCs). Promyelocytic leukemia protein activates fatty acid oxidation (FAO), a metabolic mechanism required for the asymmetric divisions and maintenance of hematopoietic stem cells (HSCs). In embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), PML is required for maintenance of the naïve and acquisition of the induced pluripotency state, respectively. Correspondingly, PML ablation causes significant morphological gene expression and lineage choice changes. In this review, we focus on the mechanisms orchestrated by PML and PML-NBs in cancer and healthy stem cells, from cell physiology to the regulation of chromatin dynamics.


Neoplasms , Promyelocytic Leukemia Protein , Transcription Factors , Autophagy , Humans , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
4.
Biomolecules ; 10(1)2019 12 18.
Article En | MEDLINE | ID: mdl-31861408

Cell-penetrating peptides are used extensively to deliver molecules into cells due to their unique characteristics such as rapid internalization, charge, and non-cytotoxicity. Amyloid fibril biomaterials were reported as gene transfer or retroviral infection enhancers; no cell internalization of the peptides themselves is reported so far. In this study, we focus on two rationally and computationally designed peptides comprised of ß-sheet cores derived from naturally occurring protein sequences and designed positively charged and aromatic residues exposed at key residue positions. The ß-sheet cores bestow the designed peptides with the ability to self-assemble into amyloid fibrils. The introduction of positively charged and aromatic residues additionally promotes DNA condensation and cell internalization by the self-assembled material formed by the designed peptides. Our results demonstrate that these designer peptide fibrils can efficiently enter mammalian cells while carrying packaged luciferase-encoding plasmid DNA, and they can act as a protein expression enhancer. Interestingly, the peptides additionally exhibited strong antimicrobial activity against the enterobacterium Escherichia coli.


Amyloid/chemistry , Cell-Penetrating Peptides/chemistry , Gene Transfer Techniques , Amyloid/metabolism , Amyloid/pharmacology , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Plasmids/genetics , Plasmids/metabolism , Protein Conformation, beta-Strand
5.
Mol Oncol ; 13(6): 1369-1387, 2019 06.
Article En | MEDLINE | ID: mdl-30927552

The multitasking promyelocytic leukemia (PML) protein was originally recognized as a tumor-suppressive factor, but more recent evidence has implicated PML in tumor cell prosurvival actions and poor patient prognosis in specific cancer settings. Here, we report that inducible PMLIV expression inhibits cell proliferation as well as self-renewal and impairs cell cycle progression of breast cancer cell lines in a reversible manner. Transcriptomic profiling identified a large number of PML-deregulated genes associated with various cell processes. Among them, cell cycle- and division-related genes and their cognitive regulators are highly ranked. In this study, we focused on previously unknown PML targets, namely the Forkhead transcription factors. PML suppresses the Forkhead box subclass M1 (FOXM1) transcription factor at both the RNA and protein levels, along with many of its gene targets. We show that FOXM1 interacts with PMLIV primarily via its DNA-binding domain and dynamically colocalizes in PML nuclear bodies. In parallel, PML modulates the activity of Forkhead box O3 (FOXO3), a factor opposing certain FOXM1 activities, to promote cell survival and stress resistance. Thus, PMLIV affects the balance of FOXO3 and FOXM1 transcriptional programs by acting on discrete gene subsets to favor both growth inhibition and survival. Interestingly, PMLIV-specific knockdown mimicked ectopic expression vis-à-vis loss of proliferative ability and self-renewal, but also led to loss of survival ability as shown by increased apoptosis. We propose that divergent or similar effects on cell physiology may be elicited by high or low PMLIV levels dictated by other concurrent genetic or epigenetic cancer cell states that may additionally account for its disparate effects in various cancer types.


Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Promyelocytic Leukemia Protein/metabolism , Blotting, Western , Breast Neoplasms/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Fluorescent Antibody Technique , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Forkhead Transcription Factors/genetics , HEK293 Cells , Humans , Immunoprecipitation , MCF-7 Cells , Oligonucleotide Array Sequence Analysis , Promyelocytic Leukemia Protein/genetics
6.
Sci Rep ; 8(1): 13790, 2018 09 13.
Article En | MEDLINE | ID: mdl-30214018

Groucho related gene 5 (GRG5) is a multifunctional protein that has been implicated in late embryonic and postnatal mouse development. Here, we describe a previously unknown role of GRG5 in early developmental stages by analyzing its function in stem cell fate decisions. By both loss and gain of function approaches we demonstrate that ablation of GRG5 deregulates the Embryonic Stem Cell (ESC) pluripotent state whereas its overexpression leads to enhanced self-renewal and acquisition of cancer cell-like properties. The malignant characteristics of teratomas generated by ESCs that overexpress GRG5 reveal its pro-oncogenic potential. Furthermore, transcriptomic analysis and cell differentiation approaches underline GRG5 as a multifaceted signaling regulator that represses mesendodermal-related genes. When ESCs exit pluripotency, GRG5 promotes neuroectodermal specification via Wnt and BMP signaling suppression. Moreover, GRG5 promotes the neuronal reprogramming of fibroblasts and maintains the self-renewal of Neural Stem Cells (NSCs) by sustaining the activity of Notch/Hes and Stat3 signaling pathways. In summary, our results demonstrate that GRG5 has pleiotropic roles in stem cell biology functioning as a stemness factor and a neural fate specifier.


Cell Transformation, Neoplastic/pathology , Embryonic Stem Cells/cytology , Neural Plate/embryology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Co-Repressor Proteins , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Mice , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/genetics , Wnt Signaling Pathway
7.
Stem Cell Reports ; 8(5): 1366-1378, 2017 05 09.
Article En | MEDLINE | ID: mdl-28392218

Promyelocytic leukemia protein (PML), the main constituent of PML nuclear bodies, regulates various physiological processes in different cell types. However, little is known about its functions in embryonic stem cells (ESC). Here, we report that PML contributes to ESC self-renewal maintenance by controlling cell-cycle progression and sustaining the expression of crucial pluripotency factors. Transcriptomic analysis and gain- or loss-of-function approaches showed that PML-deficient ESC exhibit morphological, metabolic, and growth properties distinct to naive and closer to the primed pluripotent state. During differentiation of embryoid bodies, PML influences cell-fate decisions between mesoderm and endoderm by controlling the expression of Tbx3. PML loss compromises the reprogramming ability of embryonic fibroblasts to induced pluripotent stem cells by inhibiting the transforming growth factor ß pathway at the very early stages. Collectively, these results designate PML as a member of the regulatory network for ESC naive pluripotency and somatic cell reprogramming.


Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Promyelocytic Leukemia Protein/metabolism , Animals , Cell Line , Cells, Cultured , Ectoderm/metabolism , Induced Pluripotent Stem Cells/cytology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Promyelocytic Leukemia Protein/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism
8.
Stem Cell Reports ; 6(3): 292-301, 2016 Mar 08.
Article En | MEDLINE | ID: mdl-26876669

Over the past years, microRNAs (miRNAs) have emerged as crucial factors that regulate self-renewal and differentiation of embryonic stem cells (ESCs). Although much is known about their role in maintaining ESC pluripotency, the mechanisms by which they affect cell fate decisions remain poorly understood. By performing deep sequencing to profile miRNA expression in mouse ESCs (mESCs) and differentiated embryoid bodies (EBs), we identified four differentially expressed miRNAs. Among them, miR-191 and miR-16-1 are highly expressed in ESCs and repress Smad2, the most essential mediator of Activin-Nodal signaling, resulting in the inhibition of mesendoderm formation. miR-23a, which is also down-regulated in the differentiated state, suppresses differentiation toward the endoderm and ectoderm lineages. We further identified miR-421 as a differentiation-associated regulator through the direct repression of the core pluripotency transcription factor Oct4 and the bone morphogenetic protein (BMP)-signaling components, Smad5 and Id2. Collectively, our findings uncover a regulatory network between the studied miRNAs and both branches of TGF-ß/BMP-signaling pathways, revealing their importance for ESC lineage decisions.


Cell Lineage , Embryonic Stem Cells/metabolism , MicroRNAs/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Inhibitor of Differentiation Protein 2/metabolism , Mice , Octamer Transcription Factor-3/metabolism , Smad2 Protein/metabolism , Smad5 Protein/metabolism
9.
World J Stem Cells ; 7(9): 1150-84, 2015 Oct 26.
Article En | MEDLINE | ID: mdl-26516408

Pluripotency of embryonic stem cells (ESCs) and induced pluripotent stem cells is regulated by a well characterized gene transcription circuitry. The circuitry is assembled by ESC specific transcription factors, signal transducing molecules and epigenetic regulators. Growing understanding of stem-like cells, albeit of more complex phenotypes, present in tumors (cancer stem cells), provides a common conceptual and research framework for basic and applied stem cell biology. In this review, we highlight current results on biomarkers, gene signatures, signaling pathways and epigenetic regulators that are common in embryonic and cancer stem cells. We discuss their role in determining the cell phenotype and finally, their potential use to design next generation biological and pharmaceutical approaches for regenerative medicine and cancer therapies.

10.
Stem Cells Int ; 2012: 184154, 2012.
Article En | MEDLINE | ID: mdl-22550500

Histone deacetylase inhibitors (HDACi) are small molecules that have important and pleiotropic effects on cell homeostasis. Under distinct developmental conditions, they can promote either self-renewal or differentiation of embryonic stem cells. In addition, they can promote directed differentiation of embryonic and tissue-specific stem cells along the neuronal, cardiomyocytic, and hepatic lineages. They have been used to facilitate embryo development following somatic cell nuclear transfer and induced pluripotent stem cell derivation by ectopic expression of pluripotency factors. In the latter method, these molecules not only increase effectiveness, but can also render the induction independent of the oncogenes c-Myc and Klf4. Here we review the molecular pathways that are involved in the functions of HDAC inhibitors on stem cell differentiation and reprogramming of somatic cells into pluripotency. Deciphering the mechanisms of HDAC inhibitor actions is very important to enable their exploitation for efficient and simple tissue regeneration therapies.

11.
J Biol Chem ; 286(2): 1037-45, 2011 Jan 14.
Article En | MEDLINE | ID: mdl-21062744

Sall1 is a multi-zinc finger transcription factor that regulates kidney organogenesis. It is considered to be a transcriptional repressor, preferentially localized on heterochromatin. Mutations or deletions of the human SALL1 gene are associated with the Townes-Brocks syndrome. Despite its high expression, no function was yet assigned for Sall1 in embryonic stem (ES) cells. In the present study, we show that Sall1 is expressed in a differentiation-dependent manner and physically interacts with Nanog and Sox2, two components of the core pluripotency network. Genome-wide mapping of Sall1-binding loci has identified 591 genes, 80% of which are also targeted by Nanog. A large proportion of these genes are related to self-renewal and differentiation. Sall1 positively regulates and synergizes with Nanog for gene transcriptional regulation. In addition, our data show that Sall1 suppresses the ectodermal and mesodermal differentiation. Specifically, the induction of the gastrulation markers T brachyury, Goosecoid, and Dkk1 and the neuroectodermal markers Otx2 and Hand1 was inhibited by Sall1 overexpression during embryoid body differentiation. These data demonstrate a novel role for Sall1 as a member of the transcriptional network that regulates stem cell pluripotency.


Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Cell Differentiation/physiology , Chromatin/physiology , Gene Expression Regulation, Developmental , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Nanog Homeobox Protein , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology
12.
PLoS One ; 4(9): e6804, 2009 Sep 03.
Article En | MEDLINE | ID: mdl-19727443

Embryonic stem (ES) cells have high self-renewal capacity and the potential to differentiate into a large variety of cell types. To investigate gene networks operating in pluripotent ES cells and their derivatives, the "Functional Genomics in Embryonic Stem Cells" consortium (FunGenES) has analyzed the transcriptome of mouse ES cells in eleven diverse settings representing sixty-seven experimental conditions. To better illustrate gene expression profiles in mouse ES cells, we have organized the results in an interactive database with a number of features and tools. Specifically, we have generated clusters of transcripts that behave the same way under the entire spectrum of the sixty-seven experimental conditions; we have assembled genes in groups according to their time of expression during successive days of ES cell differentiation; we have included expression profiles of specific gene classes such as transcription regulatory factors and Expressed Sequence Tags; transcripts have been arranged in "Expression Waves" and juxtaposed to genes with opposite or complementary expression patterns; we have designed search engines to display the expression profile of any transcript during ES cell differentiation; gene expression data have been organized in animated graphs of KEGG signaling and metabolic pathways; and finally, we have incorporated advanced functional annotations for individual genes or gene clusters of interest and links to microarray and genomic resources. The FunGenES database provides a comprehensive resource for studies into the biology of ES cells.


Databases, Genetic , Genomics , Stem Cells/cytology , Animals , Cell Differentiation , Cell Line , Cluster Analysis , Expressed Sequence Tags , Gene Expression Profiling , Mice , Multigene Family , Oligonucleotide Array Sequence Analysis , Signal Transduction , Software
13.
Genome Biol ; 9(4): R65, 2008 Apr 04.
Article En | MEDLINE | ID: mdl-18394158

BACKGROUND: Epigenetic mechanisms regulate gene expression patterns affecting cell function and differentiation. In this report, we examine the role of histone acetylation in gene expression regulation in mouse embryonic stem cells employing transcriptomic and epigenetic analysis. RESULTS: Embryonic stem cells treated with the histone deacetylase inhibitor Trichostatin A (TSA), undergo morphological and gene expression changes indicative of differentiation. Gene profiling utilizing Affymetrix microarrays revealed the suppression of important pluripotency factors, including Nanog, a master regulator of stem cell identity, and the activation of differentiation-related genes. Transcriptional and epigenetic changes induced after 6-12 hours of TSA treatment mimic those that appear during embryoid body differentiation. We show here that the early steps of stem cell differentiation are marked by the enhancement of bulk activatory histone modifications. At the individual gene level, we found that transcriptional reprogramming triggered by histone deacetylase inhibition correlates with rapid changes in activating K4 trimethylation and repressive K27 trimethylation of histone H3. The establishment of H3K27 trimethylation is required for stable gene suppression whereas in its absence, genes can be reactivated upon TSA removal. CONCLUSION: Our data suggest that inhibition of histone deacetylases accelerates the early events of differentiation by regulating the expression of pluripotency- and differentiation-associated genes in an opposite manner. This analysis provides information about genes that are important for embryonic stem cell function and the epigenetic mechanisms that regulate their expression.


Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Histone Deacetylase Inhibitors/pharmacology , Acetylation , Animals , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Histones/metabolism , Hydroxamic Acids/pharmacology , Mice
14.
Nucleic Acids Res ; 34(3): 765-72, 2006.
Article En | MEDLINE | ID: mdl-16452299

The deacetylase inhibitor Trichostatin A (TSA) induces the transcription of the Major Histocompatibility Class II (MHC II) DRA gene in a way independent of the master coactivator CIITA. To analyze the molecular mechanisms by which this epigenetic regulator stimulates MHC II expression, we used chromatin immunoprecipitation (ChIP) assays to monitor the alterations in histone modifications that correlate with DRA transcription after TSA treatment. We found that a dramatic increase in promoter linked histone acetylation is followed by an increase in Histone H3 lysine 4 methylation and a decrease of lysine 9 methylation. Fluorescence recovery after photobleaching (FRAP) experiments showed that TSA increases the mobility of HDAC while decreasing the mobility of the class II enhanceosome factor RFX5. These data, in combination with ChIP experiments, indicate that the TSA-mediated induction of DRA transcription involves HDAC relocation and enhanceosome stabilization. In order to gain a genome-wide view of the genes responding to inhibition of deacetylases, we compared the transcriptome of B cells before and after TSA treatment using Affymetrix microarrays. This analysis showed that in addition to the DRA gene, the entire MHC II family and the adjacent histone cluster that are located in chromosome 6p21-22 locus are strongly induced by TSA. A complex pattern of gene reprogramming by TSA involves immune recognition, antiviral, apoptotic and inflammatory pathways and extends the rationale for using Histone Deacetylase Inhibitors (HDACi) to modulate the immune response.


Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Genes, MHC Class II , Histone Deacetylase Inhibitors , Histones/metabolism , Hydroxamic Acids/pharmacology , Transcriptional Activation , Acetylation/drug effects , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Profiling , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/genetics , HLA-DR alpha-Chains , HeLa Cells , Humans , Regulatory Factor X Transcription Factors , Transcription Factors/metabolism
15.
J Biol Chem ; 279(39): 40529-35, 2004 Sep 24.
Article En | MEDLINE | ID: mdl-15271997

Tightly regulated expression of major histocompatibility complex (MHC) class II genes is critical for the immune system. A conserved regulatory module consisting of four cis-acting elements, the W, X, X2 and Y boxes, controls transcription of MHC class II genes. The X, X2, and Y boxes are bound, respectively, by RFX, CREB, and NF-Y to form a MHC class II-specific enhanceosome complex. The latter constitutes a landing pad for recruitment of the transcriptional co-activator CIITA. In contrast to the well defined roles of the X, X2, and Y boxes, the role of the W region has remained controversial. In vitro binding studies have suggested that it might contain a second RFX-binding site. We demonstrate here by means of promoter pull-down assays that the most conserved subsequence within the W region, called the S box, is a critical determinant for tethering of CIITA to the enhanceosome complex. Binding of CIITA to the enhanceosome requires both integrity of the S box and a remarkably stringent spacing between the S and X boxes. Even a 1-2-base pair change in the native S-X distance is detrimental for CIITA recruitment and promoter function. In contrast to current models, binding of RFX to a putative duplicated binding site in the W box is thus not required for either CIITA recruitment or promoter activity. This paves the way for the identification of novel factors mediating the contribution of the S box to the activation of MHC class II promoters.


Genes, MHC Class II , Nuclear Proteins/genetics , Promoter Regions, Genetic , Trans-Activators/genetics , Transcription, Genetic , Base Sequence , Binding Sites , Cell Line , Humans , Immunoblotting , Luciferases/metabolism , Major Histocompatibility Complex , Molecular Sequence Data , Mutation , Protein Binding , Transcriptional Activation
17.
EMBO J ; 22(19): 5125-36, 2003 Oct 01.
Article En | MEDLINE | ID: mdl-14517250

We describe the temporal order of recruitment of transcription factors, cofactors and basal transcriptional components and the consequent biochemical events that lead to activation of the major histocompatibility class II (MHCII) DRA gene transcription by IFN-gamma. We found that the gene is 'poised' for activation since both the activators and a fraction of the basal transcriptional machinery are pre-assembled at the enhancer and promoter prior to IFN-gamma treatment. The class II transactivator is synthesized following IFN-gamma treatment and it is recruited to the enhanceosome leading to the subsequent recruitment of the CBP and GCN5 coactivators. This is followed by histone acetylation and recruitment of the SWI/SNF chromatin remodeling complex. CIITA also recruits the CDK7 and CDK9 kinases and enhances the ability of CDK7 to phosphorylate Pol II at Ser5 leading to initiation of mRNA synthesis. Thus, the gene-specific class II transactivator selects the target genes for expression by coordinating a multiple set of biochemical activities ranging from chromatin alterations and pre-initiation complex assembly to promoter clearance.


Nuclear Proteins , RNA Polymerase II/metabolism , Trans-Activators/metabolism , Animals , COS Cells , HeLa Cells , Humans , Phosphorylation , Promoter Regions, Genetic , Serine/metabolism , Transcription, Genetic/physiology
18.
Mol Endocrinol ; 17(12): 2509-18, 2003 Dec.
Article En | MEDLINE | ID: mdl-12933903

We show here that steroid receptor coactivator 1 (SRC-1) is a coactivator of MHC class II genes that stimulates their interferon gamma (IFNgamma) and class II transactivator (CIITA)-mediated expression. SRC-1 interacts physically with the N-terminal activation domain of CIITA through two regions: one central [extending from amino acids (aa) 360-839] that contains the nuclear receptors binding region and one C-terminal (aa 1138-1441) that contains the activation domain 2. Using chromatin immunoprecipitation assays we show that SRC-1 recruitment on the class II promoter is enhanced upon IFNgamma stimulation. Most importantly, SRC-1 relieves the inhibitory action of estrogens on the IFNgamma-mediated induction of class II genes in transient transfection assays. We provide evidence that inhibition by estradiol is due to multiple events such as slightly reduced recruitment of CIITA and SRC-1 and severely inhibited assembly of the preinitiation complex.


Interferon-gamma/physiology , Steroids/physiology , Transcription Factors/physiology , Animals , Base Sequence , Breast Neoplasms , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , DNA Primers , Genes, MHC Class II , Genes, Reporter , HeLa Cells , Histocompatibility Antigens Class II/immunology , Histone Acetyltransferases , Humans , Luciferases/genetics , Mice , Nuclear Receptor Coactivator 1 , Polymerase Chain Reaction , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transfection
19.
Cancer Immunol Immunother ; 52(8): 513-22, 2003 Aug.
Article En | MEDLINE | ID: mdl-12715240

To broaden the applicability of adoptive cellular immunotherapy against HER-2/neu overexpressing human cancers, we constructed a chimeric scFv/gamma gene composed of the variable regions of a HER-2/neu specific monoclonal antibody (mAb) joined to the signaling gamma-chain of the Fc(epsilon)RI receptor. The scFv(anti-HER-2/neu)/gamma chimeric gene was successfully expressed as functional surface receptor in the MD.45 cytolytic T-cell (CTL) hybridoma (MD.45-HER/gamma). Expression of the chimeric protein triggered IL-2 and IFN-gamma secretion in vitro upon encountering cell surface HER-2/neu and mediated non-major-histocompatibility-complex (MHC)-restricted HER-2/neu-specific target cell lysis. We also examined the in vivo activity of the MD.45-HER/gamma transduced cells. Severe combined immunodeficiency disease (SCID) mice that were given HER-2/neu positive (+) human tumor cell lines had significantly increased survival compared to mice treated with saline only, or with MD.45 cells transduced with a control anti-trinitrophenyl (anti-TNP) chimeric receptor gene (MD.45-TNP/gamma). These results demonstrate the feasibility of redirecting MD.45 CTL to react in vitro and in vivo with a variety of HER-2/neu(+) tumor cells by our gene transduction protocol. Moreover, they open the possibility of using the same chimeric gene for transducing primary lymphocytes and thus allowing adoptive immunotherapy against HER-2/neu(+) cancers.


Immunoglobulin Fragments/genetics , Immunoglobulin gamma-Chains/genetics , Melanoma, Experimental/therapy , Receptor, ErbB-2/immunology , Receptors, IgE/immunology , Recombinant Fusion Proteins/genetics , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Base Sequence , Cytotoxicity, Immunologic , Hybridomas/immunology , Immunotherapy , Interferon-gamma/metabolism , Interleukin-2/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Molecular Sequence Data , Skin Neoplasms/genetics , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Transfection , Tumor Cells, Cultured
...