Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 451
Filtrar
1.
Sci Rep ; 10(1): 13125, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32753634

RESUMO

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.


Assuntos
Elementos de DNA Transponíveis/genética , Receptores de Antígenos Quiméricos/genética , Tetraciclina/farmacologia , Sequência de Aminoácidos , Células HEK293 , Humanos , Modelos Moleculares , Mutação , Conformação Proteica , Receptores de Antígenos Quiméricos/química , Elementos de Resposta/efeitos dos fármacos
2.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32825330

RESUMO

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.


Assuntos
Neoplasias da Mama/genética , Caveolina 1/genética , Elementos de Resposta , Adulto , Idoso , Linhagem Celular Tumoral , Estradiol/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histonas/genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Pessoa de Meia-Idade , Receptores de Estrogênio/genética , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética
3.
Oncol Rep ; 44(3): 1013-1024, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32705280

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Neuroblastoma/tratamento farmacológico , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores do Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Antineoplásicos/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Criança , Conjuntos de Dados como Assunto , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Neuroblastoma/genética , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Regiões Promotoras Genéticas , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sinaptofisina/genética , Tretinoína/uso terapêutico , Proteína Wnt-5a/genética
4.
J Cell Biol ; 219(7)2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32428200

RESUMO

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.


Assuntos
Artefatos , Sistemas CRISPR-Cas , Edição de Genes/métodos , Genoma , Elementos de Resposta/efeitos dos fármacos , Animais , Doxiciclina/efeitos adversos , Regulação da Expressão Gênica , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Tamoxifeno/efeitos adversos , Tetraciclina/efeitos adversos , Transfecção/métodos
5.
Am J Physiol Endocrinol Metab ; 318(4): E441-E452, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31935111

RESUMO

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.


Assuntos
Estradiol/farmacologia , Canais Iônicos/biossíntese , Canais Iônicos/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Miócitos de Músculo Liso/metabolismo , Miométrio/metabolismo , Progesterona/farmacologia , Adulto , Linhagem Celular , Feminino , Humanos , Mutação/genética , Miócitos de Músculo Liso/efeitos dos fármacos , Miométrio/efeitos dos fármacos , Gravidez , RNA Mensageiro/biossíntese , Elementos de Resposta/efeitos dos fármacos
6.
Mol Cell Endocrinol ; 504: 110701, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31926189

RESUMO

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.


Assuntos
Neoplasias da Mama/genética , Cistatina A/genética , Cistatina A/metabolismo , Metilação de DNA , Receptor alfa de Estrogênio/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Metilação de DNA/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Células Tumorais Cultivadas
7.
Mol Cell Endocrinol ; 503: 110693, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31881246

RESUMO

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.


Assuntos
Resistência a Medicamentos/genética , Receptor alfa de Estrogênio/genética , Ativação Transcricional/genética , Processamento Alternativo/genética , Animais , Células COS , Chlorocebus aethiops , Antagonistas do Receptor de Estrogênio/farmacologia , Antagonistas do Receptor de Estrogênio/uso terapêutico , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Feminino , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Células MCF-7 , Ligação Proteica/genética , Domínios Proteicos/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética
8.
Mol Biotechnol ; 61(5): 305-316, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30656573

RESUMO

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.


Assuntos
Chlamydomonas reinhardtii/crescimento & desenvolvimento , Técnicas Microbiológicas/métodos , Engenharia de Proteínas/métodos , Elementos de Resposta , Núcleo Celular/genética , Núcleo Celular/metabolismo , Chlamydomonas reinhardtii/genética , Meios de Cultura/química , Regulação Fúngica da Expressão Gênica , Ferro/farmacologia , Regiões Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Elementos de Resposta/efeitos dos fármacos , Transgenes
9.
Biochim Biophys Acta Gen Subj ; 1863(3): 586-597, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30611848

RESUMO

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.


Assuntos
Canabinoides/análise , Canabinoides/isolamento & purificação , PPAR alfa/agonistas , PPAR gama/agonistas , Compostos Fitoquímicos , Células 3T3-L1 , Animais , Células COS , Canabinoides/química , Canabinoides/farmacologia , Chlorocebus aethiops , Biologia Computacional , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Camundongos , Modelos Moleculares , Simulação de Acoplamento Molecular , PPAR alfa/química , PPAR alfa/metabolismo , PPAR gama/química , PPAR gama/metabolismo , Compostos Fitoquímicos/análise , Compostos Fitoquímicos/química , Compostos Fitoquímicos/isolamento & purificação , Compostos Fitoquímicos/farmacologia , Ligação Proteica , Elementos de Resposta/efeitos dos fármacos
10.
J Endocrinol ; 238(3): 165-176, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30012715

RESUMO

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.


Assuntos
Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Estrogênio/fisiologia , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/efeitos dos fármacos , Estrogênios/farmacologia , Feminino , Fulvestranto/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Luciferases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
11.
Sci Rep ; 8(1): 7488, 2018 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-29748590

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Multimerização Proteica/fisiologia , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/fisiologia , Dexametasona/farmacologia , Dimerização , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Células HeLa , Humanos , Técnicas Analíticas Microfluídicas , Ligação Proteica/efeitos dos fármacos , Receptores de Glucocorticoides/análise , Elementos de Resposta/efeitos dos fármacos , Espectrometria de Fluorescência/instrumentação , Espectrometria de Fluorescência/métodos , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Células Tumorais Cultivadas
12.
Carcinogenesis ; 39(6): 838-849, 2018 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-29668859

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Fator 3 de Transcrição de Octâmero/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Neoplasias Testiculares/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos NOD , Neoplasias Embrionárias de Células Germinativas/metabolismo , Elementos de Resposta/efeitos dos fármacos , Neoplasias Testiculares/metabolismo , Proteases Específicas de Ubiquitina/metabolismo
13.
PLoS Genet ; 14(3): e1007227, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29538372

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Proteínas de Ligação a DNA/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Proteínas Oncogênicas/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , 4-Nitroquinolina-1-Óxido/toxicidade , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Proteínas de Ligação a DNA/metabolismo , Epitélio/patologia , Neoplasias Esofágicas/induzido quimicamente , Carcinoma de Células Escamosas do Esôfago , Regulação Neoplásica da Expressão Gênica , Queratinócitos/patologia , Camundongos Transgênicos , Proteínas Oncogênicas/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Tetraciclina/farmacologia , Língua/efeitos dos fármacos , Língua/patologia , Transgenes
14.
Gene ; 658: 70-75, 2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29514046

RESUMO

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.


Assuntos
Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Estilbenos/farmacologia , Células CACO-2 , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes fos/efeitos dos fármacos , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Elementos de Resposta/efeitos dos fármacos , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
15.
Eur J Pharmacol ; 822: 128-137, 2018 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-29355553

RESUMO

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.


Assuntos
Citoproteção/efeitos dos fármacos , Diterpenos do Tipo Caurano/farmacologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Edulcorantes/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antioxidantes/metabolismo , Morte Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Heme Oxigenase-1/metabolismo , Células Hep G2 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Elementos de Resposta/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
16.
Eur J Pharmacol ; 818: 470-479, 2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29154837

RESUMO

The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway, which induces the production of antioxidant enzymes, is a possible therapeutic target for treating diseases related to oxidative stress. Nrf2 activators often exhibit cytotoxicity due to nonspecific electrophilic reactions with thiol groups. We screened a chemical library to explore Nrf2 activators with a wide safety margin. In at least in vitro experiments, TPNA10168, identified from the library, showed a higher efficacy in Nrf2 activation and a lower cytotoxicity than sulforaphane, a well-known Nrf2 activator. The present study demonstrated the protective effect of TPNA10168 against 6-hydroxydopamine-induced cytotoxicity. In PC12 cells, NAD(P)H:quinone oxidoreductase 1 was upregulated by TPNA10168 and participated in the protective effect. In primary mesencephalic cultures, heme oxygenase-1, upregulated by TPNA10168 in astrocytes, provided protection of dopaminergic neurons via a guanylate cyclase/protein kinase G signaling pathway via carbon monoxide. These results suggest that the compound identified from the chemical library may be suitable as a neuroprotective agent with the ability to induce antioxidant enzymes.


Assuntos
Antioxidantes/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/farmacologia , Elementos de Resposta/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/citologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Oxidopamina/toxicidade , Células PC12 , Ratos , Regulação para Cima/efeitos dos fármacos
17.
Biol Pharm Bull ; 40(11): 1909-1916, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29093337

RESUMO

Bisphenols are endocrine disruptors that are widely found in the environment. Accumulating experimental evidence suggests an adverse interaction between bisphenols and estrogen signaling. Most studies have performed experiments that focused on estrogen receptor (ER) engagement by bisphenols. Therefore, the effects of bisphenols on the expression of ERα (ESR1) and ERß (ESR2) remain largely unknown. In the present study, we examined the effects of four bisphenols: bisphenol A (BPA), bisphenol B (BPB), bisphenol S (BPS), and bisphenol AF (BPAF), on estrogen signaling in two human breast cancer cell lines (MCF-7 and SK-BR-3). Among these bisphenols, BPAF up-regulated the expression of ERß, and this was coupled with the abrogation of estrogen response element (ERE)-mediated transcriptional activities as well as the down-regulation of Cdc2 expression in MCF-7 cells, without influencing the expression of ERα. BPAF functioned as an agonist of ERα at lower concentrations (nanomolar order), but did not exhibit any modulatory action on ERα transiently expressed in SK-BR-3 cells in the presence or absence of 17ß-estradiol (E2) at higher concentrations (micromolar order). The introduction of ERß cDNA resulted in greater reductions in MCF-7 cell viability than with BPAF alone. Since ERß is a suppressive molecule of ERα function, these results provide rational evidence for BPAF functioning as an anti-estrogenic compound via the induction of ERß at higher concentrations.


Assuntos
Compostos Benzidrílicos/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Fenóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína Quinase CDC2/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Disruptores Endócrinos/farmacologia , Estradiol/metabolismo , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7 , Elementos de Resposta/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima
18.
J Biol Chem ; 292(33): 13521-13530, 2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-28679531

RESUMO

NK3 homeobox 1 (Nkx3.1), a transcription factor expressed in the prostate epithelium, is crucial for maintaining prostate cell fate and suppressing tumor initiation. Nkx3.1 is ubiquitously expressed in luminal cells of hormonally intact prostate but, upon androgen deprivation, exclusively labels a type of luminal stem cells named castration-resistant Nkx3.1-expressing cells (CARNs). During prostate cancer initiation, Nkx3.1 expression is frequently lost in both humans and mouse models. Therefore, investigating how Nkx3.1 expression is regulated in vivo is important for understanding the mechanisms of prostate stem cell specification and cancer initiation. Here, using a transgenic mouse line with destabilized GFP, we identified an 11-kb genomic region 3' of the Nkx3.1 transcription start site to be responsible for alterations in Nkx3.1 expression patterns under various physiological conditions. We found that androgen cell-autonomously activates Nkx3.1 expression through androgen receptor (AR) binding to the 11-kb region in both normal luminal cells and CARNs and discovered new androgen response elements in the Nkx3.1 3' UTR. In contrast, we found that, in Pten-/- prostate tumors, loss of Nkx3.1 expression is mediated at the transcriptional level through the 11-kb region despite functional AR in the nucleus. Importantly, the GFP reporter specifically labeled CARNs in the regressed prostate only in the presence of cell-autonomous AR, supporting a facultative model for CARN specification.


Assuntos
Regiões 3' não Traduzidas/efeitos dos fármacos , Células-Tronco Adultas/efeitos dos fármacos , Androgênios/farmacologia , Proteínas de Homeodomínio/metabolismo , Próstata/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/patologia , Androgênios/toxicidade , Animais , Carcinogênese/induzido quimicamente , Carcinógenos/toxicidade , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Orquiectomia , Próstata/citologia , Próstata/metabolismo , Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/induzido quimicamente , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Elementos de Resposta/efeitos dos fármacos , Fatores de Transcrição/genética
19.
J Biol Chem ; 292(34): 14165-14175, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28687631

RESUMO

Circadian clock and Smad2/3/4-mediated Nodal signaling regulate multiple physiological and pathological processes. However, it remains unknown whether Clock directly cross-talks with Nodal signaling and how this would regulate embryonic development. Here we show that Clock1a coordinated mesoderm development and primitive hematopoiesis in zebrafish embryos by directly up-regulating Nodal-Smad3 signaling. We found that Clock1a is expressed both maternally and zygotically throughout early zebrafish development. We also noted that Clock1a alterations produce embryonic defects with shortened body length, lack of the ventral tail fin, or partial defect of the eyes. Clock1a regulates the expression of the mesodermal markers ntl, gsc, and eve1 and of the hematopoietic markers scl, lmo2, and fli1a Biochemical analyses revealed that Clock1a stimulates Nodal signaling by increasing expression of Smad2/3/4. Mechanistically, Clock1a activates the smad3a promoter via its E-box1 element (CAGATG). Taken together, these findings provide mechanistic insight into the role of Clock1a in the regulation of mesoderm development and primitive hematopoiesis via modulation of Nodal-Smad3 signaling and indicate that Smad3a is directly controlled by the circadian clock in zebrafish.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário , Mesoderma/metabolismo , Proteína Nodal/agonistas , Transdução de Sinais , Proteína Smad3/agonistas , Proteínas de Peixe-Zebra/agonistas , Peixe-Zebra , Animais , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Embrião não Mamífero/anormalidades , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Hematopoese/efeitos dos fármacos , Humanos , Hibridização In Situ , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mesoderma/anormalidades , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Microinjeções , Microscopia de Fluorescência , Morfolinos/farmacologia , Mutação , Proteína Nodal/genética , Proteína Nodal/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
20.
J Biol Chem ; 292(34): 14270-14278, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28659340

RESUMO

Smooth muscle cell (SMC) differentiation is essential for vascular development, and TGF-ß signaling plays a critical role in this process. Although long non-coding RNAs (lncRNAs) regulate various cellular events, their functions in SMC differentiation remain largely unknown. Here, we demonstrate that the lncRNA growth arrest-specific 5 (GAS5) suppresses TGF-ß/Smad3 signaling in smooth muscle cell differentiation of mesenchymal progenitor cells. We found that forced expression of GAS5 blocked, but knockdown of GAS5 increased, the expression of SMC contractile proteins. Mechanistically, GAS5 competitively bound Smad3 protein via multiple RNA Smad-binding elements (rSBEs), which prevented Smad3 from binding to SBE DNA in TGF-ß-responsive SMC gene promoters, resulting in suppression of SMC marker gene transcription and, consequently, in inhibition of TGF-ß/Smad3-mediated SMC differentiation. Importantly, other lncRNAs or artificially synthesized RNA molecules that contained rSBEs also effectively inhibited TGF-ß/Smad3 signaling, suggesting that lncRNA-rSBE may be a general mechanism used by cells to fine-tune Smad3 activity in both basal and TGF-ß-stimulated states. Taken together, our results have uncovered an lncRNA-based mechanism that modulates TGF-ß/Smad3 signaling during SMC differentiation.


Assuntos
Músculo Liso Vascular/metabolismo , RNA Longo não Codificante/metabolismo , RNA/metabolismo , Elementos de Resposta , Transdução de Sinais , Proteína Smad3/antagonistas & inibidores , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Ligação Competitiva , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Hibridização in Situ Fluorescente , Isoquinolinas/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Proteínas Musculares/agonistas , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Conformação de Ácido Nucleico , Piridinas/farmacologia , Pirróis/farmacologia , Interferência de RNA , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/química , Elementos de Resposta/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/química , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/química , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA