Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 583
Filter
1.
Int J Mol Sci ; 22(23)2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34884850

ABSTRACT

SIMILAR TO RCD-ONEs (SROs) comprise a small plant-specific gene family which play important roles in regulating numerous growth and developmental processes and responses to environmental stresses. However, knowledge of SROs in sesame (Sesamum indicum L.) is limited. In this study, four SRO genes were identified in the sesame genome. Phylogenetic analysis showed that 64 SROs from 10 plant species were divided into two groups (Group I and II). Transcriptome data revealed different expression patterns of SiSROs over various tissues. Expression analysis showed that Group II SROs, especially SiSRO2b, exhibited a stronger response to various abiotic stresses and phytohormones than those in Group I, implying their crucial roles in response to environmental stimulus and hormone signals. In addition, the co-expression network and protein-protein interaction network indicated that SiSROs are associated with a wide range of stress responses. Moreover, transgenic yeast harboring SiSRO2b showed improved tolerance to salt, osmotic and oxidative stress, indicating SiSRO2b could confer multiple tolerances to transgenic yeast. Taken together, this study not only lays a foundation for further functional dissection of the SiSRO gene family, but also provides valuable gene candidates for genetic improvement of abiotic stress tolerance in sesame.


Subject(s)
Nuclear Proteins/metabolism , Plant Proteins/metabolism , Sesamum/metabolism , Stress, Physiological , Multigene Family , Nuclear Proteins/classification , Nuclear Proteins/genetics , Phylogeny , Plant Growth Regulators/pharmacology , Plant Proteins/classification , Plant Proteins/genetics , Promoter Regions, Genetic , Protein Interaction Maps/genetics , Response Elements/drug effects , Response Elements/genetics , Sesamum/genetics , Transcriptome/drug effects
2.
Int J Mol Sci ; 21(17)2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32825330

ABSTRACT

The estrogen receptor (ER) signaling regulates numerous physiological processes mainly through activation of gene transcription (genomic pathways). Caveolin1 (CAV1) is a membrane-resident protein that behaves as platform to enable different signaling molecules and receptors for membrane-initiated pathways. CAV1 directly interacts with ERs and allows their localization on membrane with consequent activation of ER-non-genomic pathways. Loss of CAV1 function is a common feature of different types of cancers, including breast cancer. Two protein isoforms, CAV1α and CAV1ß, derived from two alternative translation initiation sites, are commonly described for this gene. However, the exact transcriptional regulation underlying CAV1 expression pattern is poorly elucidated. In this study, we dissect the molecular mechanism involved in selective expression of CAV1ß isoform, induced by estrogens and downregulated in breast cancer. Luciferase assays and Chromatin immunoprecipitation demonstrate that transcriptional activation is triggered by estrogen-responsive elements embedded in CAV1 intragenic regions and DNA-binding of estrogen-ER complexes. This regulatory control is dynamically established by local chromatin changes, as proved by the occurrence of histone H3 methylation/demethylation events and association of modifier proteins as well as modification of H3 acetylation status. Thus, we demonstrate for the first time, an estrogen-ERs-dependent regulatory circuit sustaining selective CAV1ß expression.


Subject(s)
Breast Neoplasms/genetics , Caveolin 1/genetics , Response Elements , Adult , Aged , Cell Line, Tumor , Estradiol/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Histones/genetics , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Middle Aged , Receptors, Estrogen/genetics , Response Elements/drug effects , Response Elements/genetics
3.
Sci Rep ; 10(1): 13125, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753634

ABSTRACT

Regulated expression of genetic elements that either encode polypeptides or various types of functional RNA is a fundamental goal for gene therapy. Inducible expression may be preferred over constitutive promoters to allow clinician-based control of gene expression. Existing Tet-On systems represent one of the tightest rheostats for control of gene expression in mammals. However, basal expression in absence of tetracycline compromises the widespread application of Tet-controlled systems in gene therapy. We demonstrate that the order of P2A-linked genes of interest was critical for maximal response and tightness of a chimeric antigen receptor (CAR)-based construct. The introduction of G72V mutation in the activation region of the TetR component of the rtTA further improved the fold response. Although the G72V mutation resulted in a removal of a cryptic splice site within rtTA, additional removal of this splice site led to only a modest improvement in the fold-response. Selective removal of key promoter elements (namely the BRE, TATA box, DPE and the four predicted Inr) confirmed the suitability of the minimal CMV promoter and its downstream sequences for supporting inducible expression. The results demonstrate marked improvement of the rtTA based Tet-On system in Sleeping Beauty for applications such as CAR T cell therapy.


Subject(s)
DNA Transposable Elements/genetics , Receptors, Chimeric Antigen/genetics , Tetracycline/pharmacology , Amino Acid Sequence , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Conformation , Receptors, Chimeric Antigen/chemistry , Response Elements/drug effects
4.
Oncol Rep ; 44(3): 1013-1024, 2020 09.
Article in English | MEDLINE | ID: mdl-32705280

ABSTRACT

Neuroblastoma is the most common inheritable, solid neoplasm in children found under the age of 7 and accounts for approximately 7% of childhood cancers. A common treatment that has been prescribed for over a decade is retinoid therapy [using all­trans retinoic acid (RA)]. Treatment with this differentiating agent has been revealed to progress the cells from their stem­cell state to a mature neuronal state gaining classical neuronal characteristics, including the suppression of proliferation. However, the molecular mechanism underlying the action of RA treatment remains to be elucidated. In the present study, a novel mechanism of RA­induced differentiation via regulation of receptor tyrosine kinase­like orphan receptor 1 (ROR1) is reported. ROR1 is overexpressed in neuroblastoma but significantly downregulated in mature differentiated neurons. Hence, it was hypothesized that RA may modulate ROR1 leading to differentiation and termination of cancerous properties. Immunoblotting revealed that following RA treatment, ROR1 levels initially increased then sharply decreased by 96 h. This was paired with synaptophysin, a mature neuron marker, sharply increasing concurrently, providing evidence of differentiation by 96 h. Investigation of the ROR1 pathway confirmed ROR1­dependent downstream activation of the PI3K/AKT signaling axis, a growth pathway previously demonstrated to promote differentiation. Chromatin immunoprecipitation revealed an increase in RAR binding to the promoters of ROR1 and its endogenous ligand, Wnt5a. This research provided compelling evidence that RA is able to modulate the expression of ROR1 and Wnt5a to promote differentiation through the expression of synaptophysin. This data combined with the overarching data from the scientific community regarding proliferation and other proliferative factors in early­stage neurons provides a more in­depth model of the process of differentiation in neurons.


Subject(s)
Antineoplastic Agents/pharmacology , Neuroblastoma/drug therapy , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Child , Datasets as Topic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neurons/drug effects , Neurons/pathology , Promoter Regions, Genetic , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Response Elements/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Synaptophysin/genetics , Tretinoin/therapeutic use , Wnt-5a Protein/genetics
5.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32428200

ABSTRACT

Spatiotemporally regulated targeted gene manipulation is a common way to study the effect of gene variants on phenotypic traits, but the Cre/loxP and Tet-On/Tet-Off systems can affect whole-organism physiology and function due to off-target effects. We highlight some of these adverse effects, including whole-body endocrinology and disturbances in the gut microbiome and in mitochondrial and metabolic function.


Subject(s)
Artifacts , CRISPR-Cas Systems , Gene Editing/methods , Genome , Response Elements/drug effects , Animals , Doxycycline/adverse effects , Gene Expression Regulation , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/adverse effects , Tetracycline/adverse effects , Transfection/methods
6.
J Endocrinol Invest ; 43(9): 1317-1325, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32219690

ABSTRACT

PURPOSE: Wolfram syndrome (WS) is a rare disorder caused by mutations in WFS1 that is characterized by diabetes mellitus, optic atrophy, sensorineural deafness, diabetes insipidus, and neurodegeneration. This disease is usually inherited as an autosomal recessive trait, but an autosomal dominant form has been reported. WFS1 encodes a transmembrane protein, which is a maintenance component of endoplasmic homeostasis. These dominant mutations were thought to increase endoplasmic reticulum (ER) stress. Recent studies suggest that 4-phenylbutyrate (PBA) and valproate (VPA) reduce ER stress. The objective of this study was to analyze the effect of PBA and VPA on dominant WFS1 mutants in vitro. METHODS: We determined whether dominant WFS1 mutants (p.His313Tyr, p.Trp314Arg, p.Asp325_Ile328del, p.Glu809Lys, and p.Glu864Lys) have the dominant negative effect using a luciferase assay of ER stress response element marker as ER stress. Moreover, the rescue of cell apoptosis induced by dominant WFS1 mutants following treatment with PBA or VPA was determined by quantitative real-time PCR of C/EBP homologous protein (CHOP) mRNA expression. RESULTS: These mutants showed the dominant negative effect on the wild-type WFS1. In addition, the levels of ER stress and CHOP mRNA were significantly elevated by all dominant WFS1 mutants. After treatment with PBA or VPA, ER stress and cell apoptosis were reduced in each mutant. CONCLUSIONS: PBA and VPA could reduce the ER stress and cell apoptosis caused by dominant WFS1 mutants.


Subject(s)
Membrane Proteins/physiology , Phenylbutyrates/pharmacology , Valproic Acid/pharmacology , Wolfram Syndrome/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation/drug effects , Genes, Dominant/drug effects , Genes, Dominant/physiology , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation/physiology , Protein Transport/drug effects , Response Elements/drug effects , Tissue Distribution/drug effects , Transcription Factor CHOP/genetics , Transcriptional Activation/drug effects , Transfection
7.
Am J Physiol Endocrinol Metab ; 318(4): E441-E452, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31935111

ABSTRACT

During pregnancy, the uterus transitions from a quiescent state to an excitable, highly contractile state to deliver the fetus. Two important contributors essential for this transition are hormones and ion channels, both of which modulate myometrial smooth muscle cell (MSMC) excitability. Recently, the sodium (Na+) leak channel, nonselective (NALCN), was shown to contribute to a Na+ leak current in human MSMCs, and mice lacking NALCN in the uterus had dysfunctional labor. Microarray data suggested that the proquiescent hormone progesterone (P4) and the procontractile hormone estrogen (E2) regulated this channel. Here, we sought to determine whether P4 and E2 directly regulate NALCN. In human MSMCs, we found that NALCN mRNA expression decreased by 2.3-fold in the presence of E2 and increased by 5.6-fold in the presence of P4. Similarly, E2 treatment decreased, and P4 treatment restored NALCN protein expression. Additionally, E2 significantly inhibited, and P4 significantly enhanced an NALCN-dependent leak current in MSMCs. Finally, we identified estrogen response and progesterone response elements (EREs and PREs) in the NALCN promoter. With the use of luciferase assays, we showed that the PREs, but not the ERE, contributed to regulation of NALCN expression. Our findings reveal a new mechanism by which NALCN is regulated in the myometrium and suggest a novel role for NALCN in pregnancy.


Subject(s)
Estradiol/pharmacology , Ion Channels/biosynthesis , Ion Channels/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Myocytes, Smooth Muscle/metabolism , Myometrium/metabolism , Progesterone/pharmacology , Adult , Cell Line , Female , Humans , Mutation/genetics , Myocytes, Smooth Muscle/drug effects , Myometrium/drug effects , Pregnancy , RNA, Messenger/biosynthesis , Response Elements/drug effects
8.
Mol Cell Endocrinol ; 504: 110701, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31926189

ABSTRACT

Despite advances in early detection and treatment, invasion and metastasis of breast tumors remains a major hurdle. Cystatin A (CSTA, also called stefin A), an estrogen-regulated gene in breast cancer cells, is an inhibitor of cysteine cathepsins, and a purported tumor suppressor. Loss of CSTA expression in breast tumors evidently shifts the balance in favor of cysteine cathepsins, thereby promoting extracellular matrix remodeling, tumor invasion and metastasis. However, the underlying mechanism behind the loss of CSTA expression in breast tumors is not known. Here, we have analyzed CSTA expression, and methylation of upstream and intron-2 CpG sites within the CSTA locus in human breast cancer cell lines and breast tumors of the TCGA cohort. Results showed an inverse relationship between expression and methylation. Sequence analysis revealed a potential estrogen response element (ERE) in the intron-2. Analysis of ChIP-seq data (ERP000380) and our own ChIP experiments showed that 17ß-estradiol (E2) enhanced ERα binding to this ERE in MCF-7 cells. This ERE was located amidst the differentially methylated intron-2 CpG sites, which provoked us to examine the possible conflict between estrogen-regulation of CSTA and DNA methylation in the intron-2. We analyzed the expression of CSTA and its regulation by E2 in MDA-MB-231 and T47D cells subjected to global demethylation by 5-azacytidine (5-aza). 5-aza significantly demethylated intron-2 CpGs, and enhanced estrogen-induced ERα occupancy at the intron-2 ERE, leading to restoration of estrogen-regulation. Taken together, our results indicate that DNA methylation-dependent silencing could play a significant role in the loss of CSTA expression in breast tumors. The potential of DNA methylation as an indicator of CSTA expression or as a marker of tumor progression can be explored in future investigations. Furthermore, our results indicate the convergence of ERα-mediated estrogen regulation and DNA methylation in the intron-2, thereby offering a novel context to understand the role of estrogen-ERα signaling axis in breast tumor invasion and metastasis.


Subject(s)
Breast Neoplasms/genetics , Cystatin A/genetics , Cystatin A/metabolism , DNA Methylation , Estrogen Receptor alpha/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DNA Methylation/drug effects , Estradiol/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Promoter Regions, Genetic/drug effects , Protein Binding/drug effects , Response Elements/drug effects , Response Elements/genetics , Tumor Cells, Cultured
9.
Mol Cell Endocrinol ; 503: 110693, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31881246

ABSTRACT

Constitutively active estrogen receptor α (ERα) variants with C-terminal truncation are candidate molecules for gain of both endocrine- and chemotherapy-resistance in estrogen-sensitive tumors. Our previous reports using artificially truncated ERα constructs demonstrated that ERα variants encoded in 1-2-3-cryptic exon- and 1-2-3-4-cryptic exon-types of transcripts have potentials to display constitutive transactivation of an estrogen response element reporter. However, naturally occurring 1-2-3-cryptic exon-type ERα variants have not been cloned yet. Therefore, the present study was designed to identify naturally occurring ERα variants encoded in 1-2-3-cryptic exon-type ERα transcripts. We cloned a novel C-terminally truncated ERα variant (ERαi34) encoded in a 1-2-3-i34 transcript from MCF-7 cells and characterized its constitutive and ER antagonist-resistant transactivation in transfected cells. Stable transfection of the variant into MCF-7 cells augmented basal cell proliferation insensitive to fulvestrant. Collectively, we validated the structure-based mechanisms underlying constitutive and ER antagonist-resistant transactivation by C-terminally truncated ERα variants.


Subject(s)
Drug Resistance/genetics , Estrogen Receptor alpha/genetics , Transcriptional Activation/genetics , Alternative Splicing/genetics , Animals , COS Cells , Chlorocebus aethiops , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Female , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Protein Binding/genetics , Protein Domains/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Response Elements/drug effects , Response Elements/genetics
10.
Genes Genomics ; 41(4): 483-490, 2019 04.
Article in English | MEDLINE | ID: mdl-30656518

ABSTRACT

BACKGROUND: RNA interference (RNAi), defined as double-stranded, RNA-mediated gene silencing, is a useful tool for functional genomic studies. Along with increasing information about genomic sequences due to the innovative development of genome-sequencing technologies, functional genomic technologies are needed to annotate the genome and determine the processes by which each gene is regulated. Lentiviral vectors have been used to efficiently deliver reagents, such as small interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs), into cells and tissues for functional genomic analyses. OBJECTIVE: We developed a lentiviral vector that efficiently expresses intronic shRNA from the tetracycline regulatory element (TRE) promoter in a doxycycline-dependent manner. METHODS: We developed a lentiviral vector system that contains reverse tetracycline-controlled transactivator 3 (rtTA3) and the TRE promoter, which are necessary for the doxycycline-inducible expression of shRNAs that are expressed as intronic miR-30a precursors. We then measured the cross-talk between the cytomegalovirus (CMV) and TRE promoters in the vector. RESULTS: We found that nearby promoters influence each other and that the TRE promoter should be located far from other promoters, such as the CMV promoter, in a vector. The orientation of a promoter with respect to other promoters also influences its transcriptional activity. A head-to-head orientation of the CMV and TRE promoters maintains the lowest level of transcription from TRE in the absence of doxycycline, compared to the tail-to-tail and head-to-tail orientations. CONCLUSION: Based on these findings, we were able to construct a lentiviral vector that faithfully expresses intronic miR-30a shRNA precursors in a doxycycline-inducible manner.


Subject(s)
MicroRNAs/genetics , Response Elements/drug effects , Transcriptional Activation , Animals , CHO Cells , Cricetinae , Cricetulus , Genetic Vectors/genetics , HEK293 Cells , Humans , Introns , Lentivirus/genetics , MicroRNAs/metabolism , Tetracycline/pharmacology , Trans-Activators/metabolism
11.
Mol Biotechnol ; 61(5): 305-316, 2019 May.
Article in English | MEDLINE | ID: mdl-30656573

ABSTRACT

Microalgae are in the focus for the production of recombinant proteins in research and potential commercial application. Inducible promoters represent important tools that potentially allow the expression of recombinant proteins at higher rates. In general, they are used to separate the culture growth phase from the production phase by initiating product formation after high cell densities have been achieved. This potentially offers a higher space-time yield, consequently improving the economics of a process. In the case of the green micro alga Chlamydomonas reinhardtii, a controlled switch between activation and deactivation of gene expression is possible by changes in cultivation parameters. In this work, parameters of induction and deactivation of the iron-responsive Fea1 promoter were analyzed over time in C. reinhardtii. The results presented for the strain CC4351 validate our previous findings presented for strain CC 400. The Fea1 promoter was successfully deactivated upon transferring the cells to medium containing 10 and 20 µM Fe3+. Within 120 h, cells showed only 1.7-6% of the initial fluorescence. Activation of the Fea1 promoter occurred promptly and prominently when cells were transferred to iron-deplete medium. In general, both strains showed a pronounced difference between the active and the inactive states of the Fea1 promoter.


Subject(s)
Chlamydomonas reinhardtii/growth & development , Microbiological Techniques/methods , Protein Engineering/methods , Response Elements , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chlamydomonas reinhardtii/genetics , Culture Media/chemistry , Gene Expression Regulation, Fungal , Iron/pharmacology , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Response Elements/drug effects , Transgenes
12.
Biochim Biophys Acta Gen Subj ; 1863(3): 586-597, 2019 03.
Article in English | MEDLINE | ID: mdl-30611848

ABSTRACT

BACKGROUND: The nuclear Peroxisome Proliferator Activated Receptors (PPARs) are ligand-activated transcription factors playing a fundamental role in energy homeostasis and metabolism. Consequently, functional impairment or dysregulation of these receptors lead to a variety of metabolic diseases. While some phytocannabinoids (pCBs) are known to activate PPARγ, no data have been reported so far on their possible activity at PPARα. METHODS: The putative binding modes of pCBs into PPARα/γ Ligand Binding Domains were found and assessed by molecular docking and molecular dynamics. Luciferase assays validated in silico predictions whereas the biological effects of such PPARα/γ ligands were assessed in HepG2 and 3T3L1 cell cultures. RESULTS: The in silico study identified cannabigerolic acid (CBGA), cannabidiolic acid (CBDA) and cannabigerol (CBG) from C. sativa as PPARα/γ dual agonists, suggesting their binding modes toward PPARα/γ isoforms and predicting their activity as full or partial agonists. These predictions were confirmed by luciferase functional assays. The resulting effects on downstream gene transcription in adipocytes and hepatocytes were also observed, establishing their actions as functional dual agonists. CONCLUSIONS: Our work broadens the activity spectrum of CBDA, CBGA and CBG by providing evidence that these pCBs act as dual PPARα/γ agonists with the ability to modulate the lipid metabolism. GENERAL SIGNIFICANCE: Dual PPARα/γ agonists have emerged as an attractive alternative to selective PPAR agonists to treat metabolic disorders. We identified some pCBs as dual PPARα/γ agonists, potentially useful for the treatment of dyslipidemia and type 2 diabetes mellitus.


Subject(s)
Cannabinoids/analysis , Cannabinoids/isolation & purification , PPAR alpha/agonists , PPAR gamma/agonists , Phytochemicals , 3T3-L1 Cells , Animals , COS Cells , Cannabinoids/chemistry , Cannabinoids/pharmacology , Chlorocebus aethiops , Computational Biology , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Mice , Models, Molecular , Molecular Docking Simulation , PPAR alpha/chemistry , PPAR alpha/metabolism , PPAR gamma/chemistry , PPAR gamma/metabolism , Phytochemicals/analysis , Phytochemicals/chemistry , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Protein Binding , Response Elements/drug effects
13.
Bioorg Med Chem Lett ; 29(2): 339-341, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30477891

ABSTRACT

Synthetic neamine mimetics have been evaluated for binding to the HIV-1 Rev response element. Modified neamine derivatives, obtained from reductive amination of neamine, led to identification of new 6-amino modified neamine-type ligands with HIV-1 RRE binding affinity up to 20× that of neamine and up to 6× that of the more complex neomycin itself. This provides a noteworthy structure-activity increase and a useful lead to simplified, chemically accessible mimetics.


Subject(s)
Anti-HIV Agents/pharmacology , Framycetin/pharmacology , HIV-1/drug effects , Neomycin/pharmacology , RNA, Viral/drug effects , Response Elements/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Dose-Response Relationship, Drug , Framycetin/chemical synthesis , Framycetin/chemistry , Molecular Structure , Neomycin/analogs & derivatives , Neomycin/chemistry , Structure-Activity Relationship
14.
J Endocrinol ; 238(3): 165-176, 2018 09.
Article in English | MEDLINE | ID: mdl-30012715

ABSTRACT

Oestrogens are well-known proliferation and differentiation factors that play an essential role in the correct development of sex-related organs and behaviour in mammals. With the use of the ERE-Luc reporter mouse model, we show herein that throughout mouse development, oestrogen receptors (ERs) are active starting from day 12 post conception. Most interestingly, we show that prenatal luciferase expression in each organ is proportionally different in relation to the germ layer of the origin. The luciferase content is highest in ectoderm-derived organs (such as brain and skin) and is lowest in endoderm-derived organs (such as liver, lung, thymus and intestine). Consistent with the testosterone surge occurring in male mice at the end of pregnancy, in the first 2 days after birth, we observed a significant increase in the luciferase content in several organs, including the liver, bone, gonads and hindbrain. The results of the present study show a widespread transcriptional activity of ERs in developing embryos, pointing to the potential contribution of these receptors in the development of non-reproductive as well as reproductive organs. Consequently, the findings reported here might be relevant in explaining the significant differences in male and female physiopathology reported by a growing number of studies and may underline the necessity for more systematic analyses aimed at the identification of the prenatal effects of drugs interfering with ER signalling, such as aromatase inhibitors or endocrine disrupter chemicals.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , Receptors, Estrogen/physiology , Animals , Embryo, Mammalian , Embryonic Development/drug effects , Estrogens/pharmacology , Female , Fulvestrant/pharmacology , Gene Expression Regulation, Developmental/drug effects , Genes, Reporter/drug effects , Luciferases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Response Elements/drug effects , Response Elements/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
15.
Sci Rep ; 8(1): 7488, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29748590

ABSTRACT

Glucocorticoid receptor (GR) is a hormone-activated transcription regulatory protein involved in metabolism as well as adrenocortical responses to psychosocial stress. Ligand-activated GR localizes to the nucleus, where GR homodimers regulate gene transcription via direct binding to glucocorticoid response elements (GREs). The role of GR homodimers in transcriptional activation has not yet been elucidated. In this study, we determined the concentration of GR homodimer, and its dissociation constant (Kd), at the single-cell level, by using fluorescence correlation spectroscopy (FCS) combined with a microwell system. Results from dissociation constant analysis and diffusion analysis suggested that GR forms complexes with other proteins as well as homodimers. We determined the relationship between the concentration of GR homodimer and transcriptional activity using a triple-color FCS-microwell system-based fluorescent reporter assay. The binding affinity of GR to GREs was analyzed via fluorescence cross-correlation spectroscopy (FCCS). Our findings indicate that the GR homodimer is essential for activating target gene transcription.


Subject(s)
Gene Expression Regulation , Protein Multimerization/physiology , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/physiology , Dexamethasone/pharmacology , Dimerization , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , HeLa Cells , Humans , Microfluidic Analytical Techniques , Protein Binding/drug effects , Receptors, Glucocorticoid/analysis , Response Elements/drug effects , Spectrometry, Fluorescence/instrumentation , Spectrometry, Fluorescence/methods , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Tumor Cells, Cultured
16.
Eur J Pharmacol ; 832: 138-144, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-29782856

ABSTRACT

Mesangial proliferative glomerulonephritis (MsPGN) is characterized by glomerular mesangial cells proliferation and extracellular matrix deposition in mesangial area, which develop into glomerulosclerosis. Both silent information regulator 2-related protein 1 (Sirt1) and nuclear factor erythroid 2-related factor 2/anti-oxidant response element (Nrf2/ARE) pathway had remarkable renoprotective effects. However, whether Sirt1 and Nrf2/ARE pathway can regulate the pathological process of MsPGN remains unknown. Here, we found that Sirt1 activation by SRT1720 decreased mesangial hypercellularity and mesangial matrix areas, reduced renal Col4 and α-SMA expressions, lowered 24 h proteinuria, and eventually reduced FN and TGF-ß1 expressions in rats received anti-Thy 1.1 IgG. Further study showed that SRT1720 markedly enhanced the activity of Nrf2/ARE pathway including promoting the nuclear content and ARE-binding ability of Nrf2, elevating the protein levels of HO-1 and SOD1, two target genes of Nrf2, which eventually increased total SOD activity and decreased malondialdehyde level in the kidney tissues of experimental anti-Thy 1.1 MsPGN rats. Taken together, Sirt1 prevented the pathological process of experimental anti-Thy 1.1 MsPGN through promoting the activation of Nrf2/ARE pathway, which warrants further elucidation. Sirt1 might be a potential therapeutic target for treating MsPGN.


Subject(s)
Antioxidants/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/prevention & control , Mesangial Cells/pathology , NF-E2-Related Factor 2/metabolism , Response Elements/drug effects , Sirtuin 1/metabolism , Animals , Enzyme Activation/drug effects , Glomerulonephritis/enzymology , Male , Mesangial Cells/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar
17.
Carcinogenesis ; 39(6): 838-849, 2018 05 28.
Article in English | MEDLINE | ID: mdl-29668859

ABSTRACT

We hypothesized that octamer-binding transcription factor 4 (OCT4) inhibition would have therapeutic benefits in testicular germ cell tumors (TGCT). To identify inhibitors of OCT4, a chemical library was screened using a luciferase reporter system under the control of an OCT4 response element. A compound named KRIBB53 was identified based on its blocking of OCT4-dependent luciferase activation. When NCCIT cells were exposed to KRIBB53, the expression levels of OCT4 target genes, such as NANOG and USP44, were inhibited with an IC50 of 13 and 15 µM, respectively. In addition, the levels of OCT4 were decreased by exposing NCCIT cells to KRIBB53, and pretreating the cells with the proteasomal inhibitor MG132 reversed the KRIBB53-induced OCT4 degradation. Biotinyl-KRIBB53 was synthesized and showed comparable activity to KRIBB53 in OCT4 downregulation. Using affinity chromatography assay, KRIBB53 was shown to associate with OCT4 in vitro. Furthermore, the drug affinity responsive target stability (DARTS) assay confirmed unmodified KRIBB53 binding to OCT4. KRIBB53 selectively inhibited proliferation of TGCT cells such as NCCIT and Tera-1 cells but not that of immortalized normal cells. Finally, the administration of KRIBB53 at 30 mg/kg reduced tumor volumes by 77% in the mice xenografted with NCCIT cells relative to their vehicle-treated counterparts. Immunoblotting assays showed that expression of OCT4 was lower in KRIBB53-treated tumor tissues than in control tissues. We provide the first report, to our knowledge, of an OCT4 inhibitor that binds to OCT4 and induces its degradation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms, Germ Cell and Embryonal/drug therapy , Octamer Transcription Factor-3/metabolism , Proteasome Endopeptidase Complex/metabolism , Testicular Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Mice, Inbred NOD , Neoplasms, Germ Cell and Embryonal/metabolism , Response Elements/drug effects , Testicular Neoplasms/metabolism , Ubiquitin-Specific Proteases/metabolism
18.
PLoS Genet ; 14(3): e1007227, 2018 03.
Article in English | MEDLINE | ID: mdl-29538372

ABSTRACT

Esophageal cancer occurs as either squamous cell carcinoma (ESCC) or adenocarcinoma. ESCCs comprise almost 90% of cases worldwide, and recur with a less than 15% five-year survival rate despite available treatments. The identification of new ESCC drivers and therapeutic targets is critical for improving outcomes. Here we report that expression of the human DEK oncogene is strongly upregulated in esophageal SCC based on data in the cancer genome atlas (TCGA). DEK is a chromatin-associated protein with important roles in several nuclear processes including gene transcription, epigenetics, and DNA repair. Our previous data have utilized a murine knockout model to demonstrate that Dek expression is required for oral and esophageal SCC growth. Also, DEK overexpression in human keratinocytes, the cell of origin for SCC, was sufficient to cause hyperplasia in 3D organotypic raft cultures that mimic human skin, thus linking high DEK expression in keratinocytes to oncogenic phenotypes. However, the role of DEK over-expression in ESCC development remains unknown in human cells or genetic mouse models. To define the consequences of Dek overexpression in vivo, we generated and validated a tetracycline responsive Dek transgenic mouse model referred to as Bi-L-Dek. Dek overexpression was induced in the basal keratinocytes of stratified squamous epithelium by crossing Bi-L-Dek mice to keratin 5 tetracycline transactivator (K5-tTA) mice. Conditional transgene expression was validated in the resulting Bi-L-Dek_K5-tTA mice and was suppressed with doxycycline treatment in the tetracycline-off system. The mice were subjected to an established HNSCC and esophageal carcinogenesis protocol using the chemical carcinogen 4-nitroquinoline 1-oxide (4NQO). Dek overexpression stimulated gross esophageal tumor development, when compared to doxycycline treated control mice. Furthermore, high Dek expression caused a trend toward esophageal hyperplasia in 4NQO treated mice. Taken together, these data demonstrate that Dek overexpression in the cell of origin for SCC is sufficient to promote esophageal SCC development in vivo.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Oncogene Proteins/genetics , Poly-ADP-Ribose Binding Proteins/genetics , 4-Nitroquinoline-1-oxide/toxicity , Animals , Carcinoma, Squamous Cell/chemically induced , DNA-Binding Proteins/metabolism , Epithelium/pathology , Esophageal Neoplasms/chemically induced , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Keratinocytes/pathology , Mice, Transgenic , Oncogene Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Response Elements/drug effects , Response Elements/genetics , Tetracycline/pharmacology , Tongue/drug effects , Tongue/pathology , Transgenes
19.
Gene ; 658: 70-75, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29514046

ABSTRACT

The polyphenol resveratrol is found in many plant and fruits and is a constituent of our diet. Resveratrol has been proposed to have chemopreventive and anti-inflammatory activities. On the cellular level, resveratrol activates stimulus-regulated transcription factors. To identify resveratrol-responsive elements within a natural gene promoter, the molecular pathway leading to c-Fos gene expression by resveratrol was dissected. The c-Fos gene encodes a basic region leucine zipper transcription factor and is a prototype of an immediate-early gene that is regulated by a wide range of signaling molecules. We analyzed chromatin-integrated c-Fos promoter-luciferase reporter genes where transcription factor binding sites were destroyed by point mutations or deletion mutagenesis. The results show that mutation of the binding sites for serum response factor (SRF), activator protein-1 (AP-1) and cAMP response element binding protein (CREB) significantly reduced reporter gene transcription following stimulation of the cells with resveratrol. Inactivation of the binding sites for signal transducer and activator of transcription (STAT) or ternary complex factors did not influence resveratrol-regulated c-Fos promoter activity. Thus, the c-Fos promoter contains three resveratrol-responsive elements, the cAMP response element (CRE), and the binding sites for SRF and AP-1. Moreover, we show that the transcriptional activation potential of the c-Fos protein is increased in resveratrol-stimulated cells, indicating that the biological activity of c-Fos is elevated by resveratrol stimulation. Pharmacological and genetic experiments revealed that the protein kinase ERK1/2 is the signal transducer that connects resveratrol treatment with the c-Fos gene.


Subject(s)
Mitogen-Activated Protein Kinase 3/metabolism , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-fos/genetics , Stilbenes/pharmacology , Caco-2 Cells , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Genes, fos/drug effects , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Response Elements/drug effects , Resveratrol , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
20.
Eur J Pharmacol ; 822: 128-137, 2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29355553

ABSTRACT

A large population of drug candidates have failed "from bench to bed" due to unwanted toxicities. We intend to develop an alternative approach for drug discovery, that is, to seek candidates from "safe" compounds. Rebaudioside A (Reb-A) is an approved commercial sweetener from Stevia rebaudiana Bertoni. We found that Reb-A protects against carbon tetrachloride (CCl4)-induced oxidative injury in human liver hepatocellular carcinoma (HepG2) cells. Reb-A showed antioxidant activity on reducing cellular reactive oxygen species and malondialdehyde levels while increasing glutathione levels and superoxide dismutase and catalase activities. Reb-A treatment induced nuclear factor erythroid-derived 2-like 2 (Nrf2) activation and antioxidant response element activity, as well as the expression of heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1). Further mechanistic studies indicated that c-Jun N-terminal kinase (JNK), extracellular signal-regulated protein kinase (ERK), mitogen-active protein kinase (MAPK) and protein kinase C epsilon (PKCε) signaling was upregulated. Thus, the present in vitro study conclusively demonstrated that Reb-A is an activator of Nrf2 and is a potential candidate hepatoprotective agent. More importantly, the present study illustrated that seeking drug candidates from "safe" compounds is a promising strategy.


Subject(s)
Cytoprotection/drug effects , Diterpenes, Kaurane/pharmacology , Liver/cytology , Liver/drug effects , NF-E2-Related Factor 2/metabolism , Sweetening Agents/pharmacology , Active Transport, Cell Nucleus/drug effects , Antioxidants/metabolism , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , MAP Kinase Signaling System/drug effects , Oxidative Stress/drug effects , Response Elements/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...