Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 983: 1-20, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28639188

RESUMEN

Small RNA partnering with Argonaute (Ago) proteins plays important roles in diverse biological processes mainly by suppressing the expression of cognate target sequences. Mounting evidence reveals that the small RNA-Ago pathway can also positively regulate gene expression, a phenomenon termed as RNA activation (RNAa), which is evolutionarily conserved from Caenorhabditis elegans to human. In this chapter, I provide a general overview of mammalian RNAa phenomena and their basic characteristics and discuss recent advances toward understanding the nature of the molecular machinery responsible for RNAa and the development of RNAa-based research tools and therapeutics.


Asunto(s)
Proteínas Argonautas/genética , ARN Pequeño no Traducido/genética , Activación Transcripcional , Animales , Humanos , Regiones Promotoras Genéticas
2.
Adv Exp Med Biol ; 983: 217-229, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28639203

RESUMEN

Small activating RNAs (saRNAs) are a class of artificially designed short duplex RNAs targeted at the promoter of a particular gene to upregulate its expression via a mechanism known as RNA activation (RNAa) and hold great promise for treating a wide variety of diseases including those undruggable by conventional therapies. The therapeutic benefits of saRNAs have been demonstrated in a number of preclinical studies carried out in different disease models including cancer. With many tumor suppressor genes (TSGs) downregulated due to either epigenetic mechanisms or haploinsufficiency resulting from deletion/mutation, cancer is an ideal disease space for saRNA therapeutics which can restore the expression of TSGs via epigenetic reprogramming. The p21WAF1/CIP gene is a TSG frequently downregulated in cancer and an saRNA for p21WAF1/CIP known as dsP21-322 has been identified to be a sequence-specific p21WAF1/CIP activator in a number of cancer types. In this chapter, we review preclinical development of medicinal dsP21-322 for cancer, especially prostate cancer and bladder cancer, and highlight its potential for further clinical development.


Asunto(s)
Neoplasias de la Próstata/terapia , ARN Bicatenario/uso terapéutico , ARN Pequeño no Traducido/uso terapéutico , Neoplasias de la Vejiga Urinaria/terapia , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Humanos , Masculino , Regiones Promotoras Genéticas
3.
PLoS Genet ; 9(9): e1003821, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086155

RESUMEN

Argonaute proteins are often credited for their cytoplasmic activities in which they function as central mediators of the RNAi platform and microRNA (miRNA)-mediated processes. They also facilitate heterochromatin formation and establishment of repressive epigenetic marks in the nucleus of fission yeast and plants. However, the nuclear functions of Ago proteins in mammalian cells remain elusive. In the present study, we combine ChIP-seq (chromatin immunoprecipitation coupled with massively parallel sequencing) with biochemical assays to show that nuclear Ago1 directly interacts with RNA Polymerase II and is widely associated with chromosomal loci throughout the genome with preferential enrichment in promoters of transcriptionally active genes. Additional analyses show that nuclear Ago1 regulates the expression of Ago1-bound genes that are implicated in oncogenic pathways including cell cycle progression, growth, and survival. Our findings reveal the first landscape of human Ago1-chromosomal interactions, which may play a role in the oncogenic transcriptional program of cancer cells.


Asunto(s)
Proteínas Argonautas/genética , Cromosomas/genética , ARN Polimerasas Dirigidas por ADN/genética , Factores Eucarióticos de Iniciación/genética , Neoplasias/genética , Proteínas Argonautas/metabolismo , Secuencia de Bases , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Genoma Humano , Heterocromatina/genética , Humanos , MicroARNs/genética , Neoplasias/patología , Regiones Promotoras Genéticas , Unión Proteica/genética
4.
J Cell Physiol ; 229(7): 834-44, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24243035

RESUMEN

Castration-resistant prostate cancer (CRPC) and its treatment are challenging issues in prostate cancer management. Here, we report that miR-663 is upregulated in CRPC tissues. Overexpression of miR-663 in prostate LNCaP cells promotes cell proliferation and invasion, neuroendocrine differentiation, and reduction in dihydrotestosterone-induced upregulation of prostate-specific antigen expression. Furthermore, results of in situ hybridization show that miR-663 expression is correlated with Gleason score and TNM stage and is an independent prognostic predictor of clinical recurrence. Together, these findings suggest that miR-663 is a potential oncomiR for CRPC and may serve as a tumor biomarker for the early diagnosis of CRPC.


Asunto(s)
MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Neoplasias de la Próstata Resistentes a la Castración/diagnóstico , Neoplasias de la Próstata Resistentes a la Castración/genética , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Detección Precoz del Cáncer , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , MicroARNs/biosíntesis , Invasividad Neoplásica/genética , Recurrencia Local de Neoplasia/patología , Neoplasias de la Próstata Resistentes a la Castración/patología
5.
RNA Biol ; 11(1): 18-24, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24384674

RESUMEN

The Argonaute family of proteins is highly evolutionarily conserved and plays essential roles in small RNA-mediated gene regulatory pathways and in a wide variety of cellular processes. They were initially discovered by genetics studies in plants and have been well characterized as key components of gene silencing pathways guided by small RNAs, a phenomenon known as RNA interference. Conventionally, guided by different classes of small RNAs, Argonautes bind to and silence homologous target sequences at the post-transcriptional level. Increasing lines of evidence support their multi-functional roles in the nucleus. Advances in high-throughput genome-wide methodologies have greatly facilitated our understanding of their functions in post-transcriptional gene silencing as well as in other nuclear events. In this point-of-view, we will summarize key findings from genome-wide analyses of the Ago subfamily of proteins in mammals and Drosophila, discuss their nuclear functions in the regulation of transcription and alternative splicing identified in recent years, and briefly touch upon their potential implications in cancer.


Asunto(s)
Proteínas Argonautas/metabolismo , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Interferencia de ARN/fisiología , Empalme Alternativo , Animales , Epigénesis Genética , Silenciador del Gen , Genoma , Humanos , Familia de Multigenes , Neoplasias/metabolismo
6.
RNA Biol ; 11(10): 1221-5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25602906

RESUMEN

Small RNA programmed Argonautes are sophisticated cellular effector platforms known to be involved in a diverse array of functions ranging from mRNA cleavage, translational inhibition, DNA elimination, epigenetic silencing, alternative splicing and even gene activation. First observed in human cells, small RNA-induced gene activation, also known as RNAa, involves the targeted recruitment of Argonaute proteins to specific promoter sequences followed by induction of stable epigenetic changes which promote transcription. The existence of RNAa remains contentious due to its elusive mechanism. A string of recent studies in C. elegans provides unequivocal evidence for RNAa's fundamental role in sculpting the epigenetic landscape and maintaining active transcription of endogenous genes and supports the presence of a functionally sophisticated network of small RNA-Argonaute pathways consisting of opposite yet complementary "yin and yang" regulatory elements. In this review, we summarize key findings from recent studies of endogenous RNAa in C. elegans, with an emphasis on the Argonaute protein CSR-1.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Epigénesis Genética , Regulación de la Expresión Génica , MicroARNs/genética , ARN de Helminto/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Humanos
7.
Nucleic Acids Res ; 40(4): 1695-707, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22053081

RESUMEN

It is largely recognized that microRNAs (miRNAs) function to silence gene expression by targeting 3'UTR regions. However, miRNAs have also been implicated to positively-regulate gene expression by targeting promoter elements, a phenomenon known as RNA activation (RNAa). In the present study, we show that expression of mouse Cyclin B1 (Ccnb1) is dependent on key factors involved in miRNA biogenesis and function (i.e. Dicer, Drosha, Ago1 and Ago2). In silico analysis identifies highly-complementary sites for 21 miRNAs in the Ccnb1 promoter. Experimental validation identified three miRNAs (miR-744, miR-1186 and miR-466d-3p) that induce Ccnb1 expression in mouse cell lines. Conversely, knockdown of endogenous miR-744 led to decreased Ccnb1 levels. Chromatin immunoprecipitation (ChIP) analysis revealed that Ago1 was selectively associated with the Ccnb1 promoter and miR-744 increased enrichment of RNA polymerase II (RNAP II) and trimethylation of histone 3 at lysine 4 (H3K4me3) at the Ccnb1 transcription start site. Functionally, short-term overexpression of miR-744 and miR-1186 resulted in enhanced cell proliferation, while prolonged expression caused chromosomal instability and in vivo tumor suppression. Such phenotypes were recapitulated by overexpression of Ccnb1. Our findings reveal an endogenous system by which miRNA functions to activate Ccnb1 expression in mouse cells and manipulate in vivo tumor development/growth.


Asunto(s)
Transformación Celular Neoplásica/genética , Ciclina B1/genética , MicroARNs/metabolismo , Regulación hacia Arriba/genética , Animales , Proteínas Argonautas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Inestabilidad Cromosómica , Ciclina B1/biosíntesis , Factores Eucarióticos de Iniciación/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo
8.
PLoS One ; 19(5): e0299522, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38696452

RESUMEN

Hepatocellular carcinoma (HCC) is the most common primary liver cancer worldwide and no pharmacological treatment is available that can achieve complete remission of HCC. Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a recently identified HCC tumor suppressor gene which plays an important role in the development of HCC and its inactivation and reactivation has been shown to result in respectively HCC tumorigenesis and suppression. Small activating RNAs (saRNAs) have been used to achieve targeted activation of therapeutic genes for the restoration of their encoded protein through the RNAa mechanism. Here we designed and validated saRNAs that could activate LHPP expression at both the mRNA and protein levels in HCC cells. Activation of LHPP by its saRNAs led to the suppression of HCC proliferation, migration and the inhibition of Akt phosphorylation. When combined with targeted anticancer drugs (e.g., regorafenib), LHPP saRNA exhibited synergistic effect in inhibiting in vitro HCC proliferation and in vivo antitumor growth in a xenograft HCC model. Findings from this study provides further evidence for a tumor suppressor role of LHPP and potential therapeutic value of restoring the expression of LHPP by saRNA for the treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular , Proliferación Celular , Pirofosfatasa Inorgánica , Neoplasias Hepáticas , Humanos , Pirofosfatasa Inorgánica/metabolismo , Pirofosfatasa Inorgánica/genética , Proliferación Celular/efectos de los fármacos , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Ratones , Línea Celular Tumoral , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones Desnudos
9.
Mol Ther Nucleic Acids ; 35(1): 102147, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38435120

RESUMEN

Antisense oligonucleotides (ASOs) were the first modality to pioneer targeted gene knockdown in the treatment of amyotrophic lateral sclerosis (ALS) caused by mutant superoxide dismutase 1 (SOD1). RNA interference (RNAi) is another mechanism of gene silencing in which short interfering RNAs (siRNAs) effectively degrade complementary transcripts. However, delivery to extrahepatic tissues like the CNS has been a bottleneck in the clinical development of RNAi. Herein, we identify potent siRNA duplexes for the knockdown of human SOD1 in which medicinal chemistry and conjugation to an accessory oligonucleotide (ACO) enable activity in CNS tissues. Local delivery via intracerebroventricular or intrathecal injection into SOD1G93A mice delayed disease progression and extended animal survival with superior efficacy compared with an ASO resembling tofersen in sequence and chemistry. Treatment also prevented disease-related declines in motor function, including improvements in animal mobility, muscle strength, and coordination. The ACO itself does not target any specific complementary nucleic acid sequence; rather, it imparts benefits conducive to bioavailability and delivery through its chemistry. The complete conjugate (i.e., siRNA-ACO) represents a novel modality for delivery of duplex RNA (e.g., siRNA) to the CNS that is currently being tested in the clinic for treatment of ALS.

10.
Prostate ; 73(14): 1591-601, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836514

RESUMEN

BACKGROUND: RNA activation (RNAa) is a small RNA-mediated gene regulation mechanism by which expression of a particular gene can be induced by targeting its promoter using small double-stranded RNA also known as small activating RNA (saRNA). We used saRNA as a molecular tool to examine NKX3-1's role as a tumor suppressor and tested in vitro and in vivo antitumor effects of NKX3-1 induction by saRNA. MATERIALS AND METHODS: NKX3-1 saRNA was transfected into human prostate cancer cells including LNCaP, CWR22R, PC-3, CWR22RV1, DuPro, LAPC4, and DU145. The transfected cells were used for analysis of gene expression by RT-PCR and immunoblotting, proliferation, apoptosis and cell cycle distribution. PC-3 xenograft models were established in immunocompromised mice and treated with NKX3-1 saRNA. RESULTS: NKX3-1 saRNA induced NKX3-1 expression in different prostate cancer cell lines, resulting in inhibited cell proliferation and survival, cell cycle arrest and apoptotic cell death. These effects were partly mediated by NKX3-1's regulation of several downstream genes including the upregulation of p21 and p27, and the inhibition of VEGFC expression. Treatment of mouse xenograft prostate tumors with intratumoral delivery of NKX3-1 saRNA formulated in lipid nanoparticles significantly inhibited tumor growth and prolonged animal survival. CONCLUSIONS: By revealing several important target genes of NKX3-1, our findings corroborated NKX3-1's role as a tumor suppressor gene through direct regulation of the cell cycle and growth/survival pathways. This study also validated the therapeutic potential of saRNA for the treatment of prostate cancer via targeted activation of tumor suppressor genes.


Asunto(s)
Fenómenos Fisiológicos Celulares/efectos de los fármacos , Proteínas de Homeodominio , Neoplasias de la Próstata , ARN Bicatenario , Factores de Transcripción , Animales , Línea Celular Tumoral , Fenómenos Fisiológicos Celulares/genética , Genes Supresores de Tumor , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/farmacología , Humanos , Masculino , Ratones , Modelos Animales , Plásmidos , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Bicatenario/genética , ARN Bicatenario/farmacología , Factores de Transcripción/genética , Factores de Transcripción/farmacología , Activación Transcripcional/efectos de los fármacos , Transfección , Trasplante Heterólogo , Resultado del Tratamiento
11.
J Urol ; 190(2): 790-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23523927

RESUMEN

PURPOSE: Promoter targeted saRNAs mediate sequence specific up-regulation of gene expression. We explored the therapeutic effect of RNA activation mediated iNOS gene activation on improving erectile function in a rat model of diabetes mellitus. MATERIALS AND METHODS: An optimal saRNA sequence specific for iNOS promoter was cloned into an adenoviral vector, resulting in AdU6/shiNOS and AdU6/shControl. The corresponding viruses were used to transduce cultured rat cavernous smooth muscle cells. Streptozotocin induced diabetes models were established in rats and used to test the effects of intracavernous delivery of iNOS saRNA viruses on erectile function. iNOS expression in the cavernous smooth muscle cells or penile tissue of treated rats was assessed by reverse transcriptase-polymerase chain reaction and Western blot. Cyclic guanosine monophosphate was analyzed by enzyme-linked immunosorbent assay. Intracavernous pressure in response to cavernous nerve stimulation was measured using a data acquisition system on post-injection days 1, 3, 5, 7, 10 and 14. RESULTS: Adenovirus mediated expression of iNOS saRNA caused sustained up-regulation of iNOS in cavernous smooth muscle cells. Intracavernous injection of AdU6/shiNOS activated iNOS expression in vivo and significantly increased peak intracavernous pressure in streptozotocin induced diabetic rats via nitric oxide/intracellular cyclic guanosine monophosphate activation. CONCLUSIONS: Results show that saRNA mediated iNOS over expression in the penis can restore erectile function in streptozocin diabetic rats via the nitric oxide-cyclic guanosine monophosphate pathway.


Asunto(s)
Diabetes Mellitus Experimental , Óxido Nítrico Sintasa de Tipo II/metabolismo , Erección Peniana/fisiología , ARN/farmacología , Adenoviridae , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , GMP Cíclico/fisiología , Ensayo de Inmunoadsorción Enzimática , Masculino , Músculo Liso/fisiología , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN , Transducción de Señal , Regulación hacia Arriba
12.
Biochem J ; 443(3): 821-8, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22339500

RESUMEN

RNAa (RNA activation) is a mechanism by which small dsRNA (double-stranded RNA), termed saRNA (small activating RNA), target promoter sequences to induce gene expression. This technique represents a novel approach to gene overexpression without the use of exogenous DNA. In the present study, we investigated whether RNAa can modulate expression of the development-related gene NANOG and manipulate cell fate. Using a lentivirus-based reporter system as a screening tool, we identified synthetic saRNAs that stimulate NANOG expression in human NCCIT embryonic carcinoma cells. Mismatch mutations to saRNA duplexes define sequence requirement for gene activation. Functional analysis of NANOG induction reveals saRNA treatment predictably modulates the expression of several known downstream target genes, including FOXH1 (forkhead box H1), REST (RE1-silencing transcription factor), OCT4 (octamer-binding protein 4) and REX1 (reduced expression protein 1). Treatment with RA (retinoic acid) triggers NCCIT cell differentiation, reducing NANOG and OCT4 expression and up-regulating several neural markers [i.e. ASCL1 (achaete-scute complex homologue 1), NEUROD1 (neuronal differentiation 1) and PAX6 (paired box 6)]. However, co-treatment with saRNA antagonizes NANOG down-regulation and RA-induced differentiation. Ectopic overexpression of NANOG via lentiviral transduction further recapitulates saRNA results, providing proof-of-concept that RNAa may be utilized to activate development-related genes and manipulate cell fate.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proteínas de Homeodominio/genética , Regiones Promotoras Genéticas , ARN/genética , Tretinoina/farmacología , Humanos , Proteína Homeótica Nanog
13.
Int J Urol ; 20(4): 362-71, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23163774

RESUMEN

Prostate cancer is a leading cause of cancer deaths in men worldwide. Management of the disease has remained a great challenge and even more so is the aggressive advanced stage with castration-resistant behavior. The mechanisms and timing of development of castration-resistant prostate cancer are unclear and remain debatable. Progression to castration-resistant prostate cancer is undoubtedly multifactorial, with a number of molecular-genetic aberrations implicated. However, a key question that remains unanswered is: when in the evolution of prostate cancer do the changes that confer castration resistance occur? Earlier attempts to address this question led to two proposed models: the "adaptation" and the "clonal selection" models. Although the prevailing hypothesis is the adaptation model, there is recent evidence in favor of the clonal selection model. Clarification of the model development of castration-resistant prostate cancer might significantly alter our diagnostic and therapeutic strategies, and potentially lead to improved outcome of management of this daunting condition. Here we review existing knowledge and current research findings addressing the timing of events in the course of prostate cancer progression to castration-resistant prostate cancer.


Asunto(s)
Selección Clonal Mediada por Antígenos , Orquiectomía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Animales , Progresión de la Enfermedad , Epigénesis Genética/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/genética
14.
PLoS One ; 18(2): e0282063, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36821623

RESUMEN

PURPOSE: Proliferative vitreoretinopathy (PVR) is a disease process resulting from proliferation of retinal pigment epithelial (RPE) cells in the vitreous and periretinal area, leading to periretinal membrane formation and traction and eventually to postoperative failure after vitreo-retinal surgery for primary rhegmatogenous retinal detachment (RRD). The present study was designed to test the therapeutic potential of a p21CIP/WAF1 (p21) inducing saRNA for PVR. METHODS: A chemically modified p21 saRNA (RAG1-40-53) was tested in cultured human RPE cells for p21 induction and for the inhibition of cell proliferation, migration and cell cycle progression. RAG1-40-53 was further conjugated to a cholesterol moiety and tested for pharmacokinetics and pharmacodynamics in rabbit eyes and for therapeutic effects after intravitreal administration in a rabbit PVR model established by injecting human RPE cells. RESULTS: RAG1-40-53 (0.3 mg, 1 mg) significantly induced p21 expression in RPE cells and inhibited cell proliferation, the progression of cell cycle at the G0/G1 phase and TGF-ß1 induced migration. After a single intravitreal injection into rabbit eyes, cholesterol-conjugated RAG1-40-53 exhibited sustained concentration in the vitreal humor beyond at least 8 days and prevented the progression of established PVR. CONCLUSION: p21 saRNA could represent a novel therapeutics for PVR by exerting a antiproliferation and antimigration effect on RPE cells.


Asunto(s)
Vitreorretinopatía Proliferativa , Animales , Conejos , Humanos , Vitreorretinopatía Proliferativa/tratamiento farmacológico , Vitreorretinopatía Proliferativa/metabolismo , Células Cultivadas , Ojo/metabolismo , División Celular , Proteínas de Homeodominio/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
15.
RNA Biol ; 9(3): 269-73, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22336708

RESUMEN

microRNAs (miRNAs), defined as 21-24 nucleotide non-coding RNAs, are important regulators of gene expression. Initially, the functions of miRNAs were recognized as post-transcriptional regulators on mRNAs that result in mRNA degradation and/or translational repression. It is becoming evident that miRNAs are not only restricted to function in the cytoplasm, they can also regulate gene expression in other cellular compartments by a spectrum of targeting mechanisms via coding regions, 5' and 3'untransalated regions (UTRs), promoters, and gene termini. In this point-of-view, we will specifically focus on the nuclear functions of miRNAs and discuss examples of miRNA-directed transcriptional gene regulation identified in recent years.


Asunto(s)
Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , MicroARNs/metabolismo , Animales , Proteínas Argonautas/metabolismo , Ciclina B1/genética , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas
16.
Bioengineered ; 13(3): 6729-6739, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35246011

RESUMEN

The loss of inner ear hair cells leads to irreversible acoustic injury in mammals, and regeneration of inner ear hair cells to restore hearing loss is challenging. ATOH1 is a key gene in the development and regeneration of hair cells. Small activating RNAs (saRNAs) can target a gene to specifically upregulate its expression. This study aimed to explore whether small activating RNAs could induce the differentiation of human adipose-derived mesenchymal stem cells into hair cell-like cells with a combination of growth factors in vitro and thus provide a new strategy for hair cell regeneration and the treatment of sensorineural hearing loss. Fifteen small activating RNAs targeting the human ATOH1 gene were designed and screened in 293 T and human adipose-derived mesenchymal stem cells, and 3 of these candidates were found to be capable of effectively and stably activating ATOH1 gene expression. The selected small activating RNAs were then transfected into hair cell progenitor cells, and hair cell markers were examined 10 days after transfection. After transfection of the selected small activating RNAs, the expression of the characteristic markers of inner ear hair cells, POU class 4 homeobox 3 (POU4F3) and myosin VIIA (MYO7A), was detected. Human adipose-derived mesenchymal stem cells have the potential to differentiate into human hair cell progenitor cells. In vitro, small activating RNAs were able to induce the differentiation of hair cell progenitor cells into hair cell-like cells. Therefore, RNA activation technology has the potential to provide a new strategy for the regeneration of hair cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , ARN , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Cabello/metabolismo , Células Ciliadas Auditivas/metabolismo , Humanos , Mamíferos/genética , ARN/metabolismo , Regeneración/genética
17.
Proc Natl Acad Sci U S A ; 105(5): 1608-13, 2008 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-18227514

RESUMEN

Recent studies have shown that microRNA (miRNA) regulates gene expression by repressing translation or directing sequence-specific degradation of complementary mRNA. Here, we report new evidence in which miRNA may also function to induce gene expression. By scanning gene promoters in silico for sequences complementary to known miRNAs, we identified a putative miR-373 target site in the promoter of E-cadherin. Transfection of miR-373 and its precursor hairpin RNA (pre-miR-373) into PC-3 cells readily induced E-cadherin expression. Knockdown experiments confirmed that induction of E-cadherin by pre-miR-373 required the miRNA maturation protein Dicer. Further analysis revealed that cold-shock domain-containing protein C2 (CSDC2), which possesses a putative miR-373 target site within its promoter, was also readily induced in response to miR-373 and pre-miR-373. Furthermore, enrichment of RNA polymerase II was detected at both E-cadherin and CSDC2 promoters after miR-373 transfection. Mismatch mutations to miR-373 indicated that gene induction was specific to the miR-373 sequence. Transfection of promoter-specific dsRNAs revealed that the concurrent induction of E-cadherin and CSDC2 by miR-373 required the miRNA target sites in both promoters. In conclusion, we have identified a miRNA that targets promoter sequences and induces gene expression. These findings reveal a new mode by which miRNAs may regulate gene expression.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/fisiología , Activación Transcripcional , Disparidad de Par Base , Secuencia de Bases , Cadherinas/genética , Línea Celular Tumoral , Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , MicroARNs/farmacología , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/genética , Ribonucleasa III/antagonistas & inhibidores , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
18.
J Biol Methods ; 8(1): e142, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33604394

RESUMEN

We measured anomalous diffusion in human prostate cancer cells which were transfected with the Alexa633 fluorescent RNA probe and co-transfected with enhanced green fluorescent protein-labeled argonaute2 protein by laser scanning microscopy. The image analysis arose from diffusion based on a "two-level system". A trap was an interaction site where the diffusive motion was slowed down. Anomalous subdiffusive spreading occurred at cellular traps. The cellular traps were not immobile. We showed how the novel analysis method of imaging data resulted in new information about the number of traps in the crowded and heterogeneous environment of a single human prostate cancer cell. The imaging data were consistent with and explained by our modern ideas of anomalous diffusion of mixed origins in live cells. Our original research presented in this study is significant as we obtained a complex diffusion mechanism in live single cells.

19.
Hum Exp Toxicol ; 40(12_suppl): S519-S529, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34670429

RESUMEN

OBJECTIVE: Ketamine is an anesthetic that induces neurotoxicity when administered at high doses. In this work, we explored the protective effects of lipoxin A4 methyl ester (LXA4 ME) against ketamine-induced neurotoxicity and the underlying protective mechanism in pheochromocytoma (PC12) cells. METHODS: PC12 cells were treated with 50 µM of ketamine and different LXA4 ME concentrations of LXA4 ME (5-50 nM) for 24 h, and their viability, apoptosis, and oxidative status were assessed. RESULTS: Quantitative real-time polymerase chain reaction experiments showed that ketamine downregulated miR-22 expression and upregulated Bcl-2-associated athanogene 5 (BAG5) in PC12 cells in a concentration-dependent manner. LXA4 ME induced the opposite effects, thus attenuating ketamine-induced neurotoxicity. Further in vitro assays showed that miR-22 directly targeted BAG5, thus promoting cell viability by suppressing cell apoptosis and oxidative stress. Under expression miR-22 or upregulation of BAG5 antagonized the effects of LXA4 ME. CONCLUSION: LXA4 ME can protect PC12 cells from ketamine-induced neurotoxicity by activating the miR-22/BAG5 signaling pathway. Thus, LXA4 ME can be used as a protective drug against ketamine-induced neural damage.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ketamina/toxicidad , Lipoxinas/farmacología , MicroARNs/metabolismo , Neuronas/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Supervivencia Celular/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Células PC12 , Ratas
20.
Prostate ; 70(10): 1066-73, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20232361

RESUMEN

BACKGROUND: Obesity is a risk factor for prostate cancer development, but the underlying mechanism is unknown. The present study tested the hypothesis that stromal cells of the adipose tissue might be recruited by cancer cells to help tumor growth. METHODS: PC3 prostate cancer cells were transplanted into the subcutaneous space of the right flank of athymic mice. One week later, adipose tissue-derived stromal or stem cells (ADSC) or phosphate-buffered saline (PBS, as control) was transplanted similarly to the left flank. Tumor size was monitored for the next 34 days; afterwards, the mice were sacrificed and their tumors harvested for histological examination. The ability of PC3 cells to attract ADSC was tested by migration assay. The involvement of the CXCL12/CXCR4 axis was tested by migration assay in the presence of a specific inhibitor AMD3100. RESULTS: Throughout the entire course, the average size of PC3 tumors in ADSC-treated mice was larger than in PBS-treated mice. ADSC were identified inside the tumors of ADSC-treated mice; CXCR4 expression was also detected. Migration assay indicated the involvement of the CXCL12/CXCR4 axis in the migration of ADSC toward PC3 cells. Capillary density was twice as high in the tumors of ADSC-treated mice than in the tumors of PBS-treated mice. VEGF expression was similar but FGF2 expression was significantly higher in tumors of ADSC-treated mice than in the tumors of PBS-tread mice. CONCLUSION: Prostate cancer cells recruited ADSC by the CXCL12/CXCR4 axis. ADSC helps tumor growth by increasing tumor vascularity, and which was mediated by FGF2.


Asunto(s)
Tejido Adiposo/patología , Neoplasias de la Próstata/patología , Trasplante de Células Madre , Animales , Bencilaminas , Movimiento Celular/fisiología , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/metabolismo , Ciclamas , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Compuestos Heterocíclicos/farmacología , Masculino , Ratones , Ratones Desnudos , Microscopía Fluorescente , Neovascularización Patológica/patología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/metabolismo , Distribución Aleatoria , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA