Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Neurosci ; 24(8): 1089-1099, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34083786

RESUMEN

Methods to enhance adult neurogenesis by reprogramming glial cells into neurons enable production of new neurons in the adult nervous system. Development of therapeutically viable approaches to induce new neurons is now required to bring this concept to clinical application. Here, we successfully generate new neurons in the cortex and dentate gyrus of the aged adult mouse brain by transiently suppressing polypyrimidine tract binding protein 1 using an antisense oligonucleotide delivered by a single injection into cerebral spinal fluid. Radial glial-like cells and other GFAP-expressing cells convert into new neurons that, over a 2-month period, acquire mature neuronal character in a process mimicking normal neuronal maturation. The new neurons functionally integrate into endogenous circuits and modify mouse behavior. Thus, generation of new neurons in the dentate gyrus of the aging brain can be achieved with a therapeutically feasible approach, thereby opening prospects for production of neurons to replace those lost to neurodegenerative disease.


Asunto(s)
Giro Dentado , Células Ependimogliales , Neurogénesis/fisiología , Neuronas , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Animales , Reprogramación Celular/fisiología , Giro Dentado/citología , Giro Dentado/fisiología , Células Ependimogliales/citología , Células Ependimogliales/fisiología , Ratones , Neuronas/citología , Neuronas/fisiología , Oligonucleótidos Antisentido
2.
Cell Mol Neurobiol ; 41(6): 1271-1284, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32529543

RESUMEN

Temozolomide (TMZ) is widely used for glioma therapy in the clinic. Currently, the development of TMZ resistance has largely led to poor prognosis. However, very little is understood about the role of MIR155HG, as a long noncoding RNA, in TMZ resistance. In our study, MIR155HG level was markedly higher in glioma patients than in normal controls and that poor survival was positively correlated with MIR155HG expression. It was apparent that TMZ sensitivity was promoted by downregulation of MIR155HG, and this could be reversed by MIR155HG overexpression in vivo and in vitro. Furthermore, polypyrimidine tract binding protein 1 (PTBP1) was proven to bind with MIR155HG and to regulate MIR155HG-related TMZ resistance. Mechanistic investigation showed that the expression levels of both MIR155HG and PTBP1 influenced the expression of relevant proteins in the Wnt/ß-catenin pathway. Collectively, the study demonstrated that the knockdown of MIR155HG increased glioma sensitivity to TMZ by inhibiting Wnt/ß-catenin pathway activation via potently downregulating PTBP1.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , MicroARNs/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Temozolomida/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Glioma/tratamiento farmacológico , Glioma/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Unión Proteica/fisiología , Temozolomida/uso terapéutico , Vía de Señalización Wnt/fisiología
3.
Brain Res Bull ; 163: 1-13, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32562719

RESUMEN

BACKGROUND: Glioma is a primary intracranial malignancy with poor prognosis, of which the pathogenesis remains to be elucidated. Therein, the aim of this study is to discuss the impacts of lncRNA plasmacytoma variant translocation 1 (PVT1)/microRNA-128-1-5p (miR-128-1-5p)/polypyrimidine tract-binding protein 1 (PTBP1) axis on the biological characteristics of glioma cells. METHODS: Glioma tissue samples (72 cases) and normal brain tissue samples (35 cases) were harvested. The expression of PVT1, miR-128-1-5p and PTBP1 in glioma tissues and cells was detected. Glioma cells were transfected with sh-PVT1, miR-128-1-5p mimics or miR-128-1-5p inhibitors to verify the impacts of PVT1 and miR-128-1-5p on DNA damage, cell colony formation, invasion, proliferation, migration and apoptosis of glioma U87 and U251 cells. The growth of transplanted tumor was tested by tumor xenograft in nude mice. The combination of PVT1 and miR-128-1-5p and the targeting relationship between miR-128-1-5p and PTBP1 were verified. RESULTS: PVT1 and PTBP1 expression was enhanced and miR-128-1-5p expression was degraded in glioma tissues and cells. Overexpressed miR-128-1-5p and lowly-expressed PVT1 promoted DNA damage, suppressed colony formation, invasion, proliferation and migration as well as boosted apoptosis of U251 and U87 cells. Up-regulating miR-128-1-5p and down-regulating PVT1 reduced transplanted tumor volume and weight of glioma in mice. Low expression miR-128-1-5p reversed the effect of low expression PVT1 on the biological characteristics of glioma cells. PVT1 specifically bound to miR-128-1-5p and PTBP1 was the target gene of miR-128-1-5p. CONCLUSION: This study suggests that down-regulated PVT1 or up-regulated miR-128-1-5p boosts apoptosis and attenuates proliferation of glioma cells by inhibiting PTBP1 expression. This study is essential for finding new therapeutic targets for glioma.


Asunto(s)
Apoptosis/fisiología , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/biosíntesis , MicroARNs/biosíntesis , Proteína de Unión al Tracto de Polipirimidina/biosíntesis , ARN Largo no Codificante/biosíntesis , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Glioma/genética , Glioma/patología , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , ARN Largo no Codificante/genética , Regulación hacia Arriba/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Toxicol Appl Pharmacol ; 383: 114747, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31499192

RESUMEN

Abnormal proliferation of vascular smooth muscle cells (VSMCs) is a hallmark of vascular restenosis. We investigated whether polypyrimidine tract-binding protein 1 (PTBP1), a novel regulator of cell proliferation and differentiation, is implicated in VSMC proliferation and neointima hyperplasia responding to injury. C57BL/6 J mice of 10-12 weeks old were randomly divided into sham and carotid artery injury group. Primary VSMCs obtained from thoracic aortas of 10- to 12-week-old mice were treated with physiological saline and platelet derived growth factor-BB (PDGF-BB). Adenovirus expressing shCon, shPTBP1 or shYY2 were transfected into the injured common carotid artery or VSMCs. qRT-PCR and immunoblotting were used to determine the mRNA and protein expression levels, respectively. Immunohistochemical staining of H&E and Ki-67 were used to evaluate restenosis of vessels. Cell counting kit-8 assay and Ki-67 immunofluorescent staining were utilized to evaluate the rate of VSMC proliferation. The expression of PTBP1 were upregulated both in injured arteries and in PDGF-BB-treated VSMCs. PTBP1 inhibition significantly attenuated neointima hyperplasia and Ki-67 positive area induced by injury. Knockdown of PTBP1 in vitro also suppressed VSMC proliferation after PDGF-BB treatment. The effects of PTBP1 inhibition mentioned above were all abolished by knockdown of YY2. Finally, we identified four cell cycle regulators (p53, p21, Cdkn1c, Cdkn2b) that were regulated by PTBP1/YY2 axis both in vitro and in vivo. These findings demonstrated that PTBP1 is a critical regulator of VSMC proliferation and neointima hyperplasia via modulating the expression of YY2.


Asunto(s)
Proliferación Celular/fisiología , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Hiperplasia/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Proteína de Unión al Tracto de Polipirimidina/fisiología , Factores de Transcripción/biosíntesis , Animales , Becaplermina/farmacología , Proliferación Celular/efectos de los fármacos , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Hiperplasia/patología , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Neointima/patología , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
5.
Nat Commun ; 10(1): 1590, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30962446

RESUMEN

Alternative splicing, a fundamental step in gene expression, is deregulated in many diseases. Splicing factors (SFs), which regulate this process, are up- or down regulated or mutated in several diseases including cancer. To date, there are no inhibitors that directly inhibit the activity of SFs. We designed decoy oligonucleotides, composed of several repeats of a RNA motif, which is recognized by a single SF. Here we show that decoy oligonucleotides targeting splicing factors RBFOX1/2, SRSF1 and PTBP1, can specifically bind to their respective SFs and inhibit their splicing and biological activities both in vitro and in vivo. These decoy oligonucleotides present an approach to specifically downregulate SF activity in conditions where SFs are either up-regulated or hyperactive.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas/genética , Oligonucleótidos/farmacología , Proteína de Unión al Tracto de Polipirimidina/genética , Factores de Empalme de ARN/genética , Factores de Empalme Serina-Arginina/genética , Empalme Alternativo , Animales , Animales Modificados Genéticamente , Sitios de Unión , Glioblastoma/genética , Glioblastoma/patología , Células HEK293 , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Músculo Esquelético/crecimiento & desarrollo , Degradación de ARNm Mediada por Codón sin Sentido , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/metabolismo , Factores de Empalme Serina-Arginina/antagonistas & inhibidores , Factores de Empalme Serina-Arginina/metabolismo , Secuencias Repetidas en Tándem , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/embriología , Pez Cebra/genética
6.
Cell Res ; 28(10): 981-995, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30143796

RESUMEN

Activation-induced cytidine deaminase (AID) mediates class switching by binding to a small fraction of single-stranded DNA (ssDNA) to diversify the antibody repertoire. The precise mechanism for highly selective AID targeting in the genome has remained elusive. Here, we report an RNA-binding protein, ROD1 (also known as PTBP3), that is both required and sufficient to define AID-binding sites genome-wide in activated B cells. ROD1 interacts with AID via an ultraconserved loop, which proves to be critical for the recruitment of AID to ssDNA using bi-directionally transcribed nascent RNAs as stepping stones. Strikingly, AID-specific mutations identified in human patients with hyper-IgM syndrome type 2 (HIGM2) completely disrupt the AID interacting surface with ROD1, thereby abolishing the recruitment of AID to immunoglobulin (Ig) loci. Together, our results suggest that bi-directionally transcribed RNA traps the RNA-binding protein ROD1, which serves as a guiding system for AID to load onto specific genomic loci to induce DNA rearrangement during immune responses.


Asunto(s)
Citidina Desaminasa/metabolismo , Genoma , Isotipos de Inmunoglobulinas/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Secuencia de Aminoácidos , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Citidina Desaminasa/química , Citidina Desaminasa/genética , Células HEK293 , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM/genética , Síndrome de Inmunodeficiencia con Hiper-IgM/patología , Isotipos de Inmunoglobulinas/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , Unión Proteica , ARN/química , ARN/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
7.
Circulation ; 136(25): 2451-2467, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-28971999

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is characterized by abnormal growth and enhanced glycolysis of pulmonary artery endothelial cells. However, the mechanisms underlying alterations in energy production have not been identified. METHODS: Here, we examined the miRNA and proteomic profiles of blood outgrowth endothelial cells (BOECs) from patients with heritable PAH caused by mutations in the bone morphogenetic protein receptor type 2 (BMPR2) gene and patients with idiopathic PAH to determine mechanisms underlying abnormal endothelial glycolysis. We hypothesized that in BOECs from patients with PAH, the downregulation of microRNA-124 (miR-124), determined with a tiered systems biology approach, is responsible for increased expression of the splicing factor PTBP1 (polypyrimidine tract binding protein), resulting in alternative splicing of pyruvate kinase muscle isoforms 1 and 2 (PKM1 and 2) and consequently increased PKM2 expression. We questioned whether this alternative regulation plays a critical role in the hyperglycolytic phenotype of PAH endothelial cells. RESULTS: Heritable PAH and idiopathic PAH BOECs recapitulated the metabolic abnormalities observed in pulmonary artery endothelial cells from patients with idiopathic PAH, confirming a switch from oxidative phosphorylation to aerobic glycolysis. Overexpression of miR-124 or siRNA silencing of PTPB1 restored normal proliferation and glycolysis in heritable PAH BOECs, corrected the dysregulation of glycolytic genes and lactate production, and partially restored mitochondrial respiration. BMPR2 knockdown in control BOECs reduced the expression of miR-124, increased PTPB1, and enhanced glycolysis. Moreover, we observed reduced miR-124, increased PTPB1 and PKM2 expression, and significant dysregulation of glycolytic genes in the rat SUGEN-hypoxia model of severe PAH, characterized by reduced BMPR2 expression and endothelial hyperproliferation, supporting the relevance of this mechanism in vivo. CONCLUSIONS: Pulmonary vascular and circulating progenitor endothelial cells isolated from patients with PAH demonstrate downregulation of miR-124, leading to the metabolic and proliferative abnormalities in PAH ECs via PTPB1 and PKM1/PKM2. Therefore, the manipulation of this miRNA or its targets could represent a novel therapeutic approach for the treatment of PAH.


Asunto(s)
Hipertensión Pulmonar Primaria Familiar/patología , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , MicroARNs/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Piruvato Quinasa/metabolismo , Animales , Antagomirs/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/antagonistas & inhibidores , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Hipertensión Pulmonar Primaria Familiar/genética , Hipertensión Pulmonar Primaria Familiar/metabolismo , Glucólisis , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Quinasas Lim/metabolismo , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , Piruvato Quinasa/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Simportadores/metabolismo
8.
Circulation ; 136(25): 2468-2485, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-28972001

RESUMEN

BACKGROUND: An emerging metabolic theory of pulmonary hypertension (PH) suggests that cellular and mitochondrial metabolic dysfunction underlies the pathology of this disease. We and others have previously demonstrated the existence of hyperproliferative, apoptosis-resistant, proinflammatory adventitial fibroblasts from human and bovine hypertensive pulmonary arterial walls (PH-Fibs) that exhibit constitutive reprogramming of glycolytic and mitochondrial metabolism, accompanied by an increased ratio of glucose catabolism through glycolysis versus the tricarboxylic acid cycle. However, the mechanisms responsible for these metabolic alterations in PH-Fibs remain unknown. We hypothesized that in PH-Fibs microRNA-124 (miR-124) regulates PTBP1 (polypyrimidine tract binding protein 1) expression to control alternative splicing of pyruvate kinase muscle (PKM) isoforms 1 and 2, resulting in an increased PKM2/PKM1 ratio, which promotes glycolysis and proliferation even in aerobic environments. METHODS: Pulmonary adventitial fibroblasts were isolated from calves and humans with severe PH (PH-Fibs) and from normal subjects. PTBP1 gene knockdown was achieved via PTBP1-siRNA; restoration of miR-124 was performed with miR-124 mimic. TEPP-46 and shikonin were used to manipulate PKM2 glycolytic function. Histone deacetylase inhibitors were used to treat cells. Metabolic products were determined by mass spectrometry-based metabolomics analyses, and mitochondrial function was analyzed by confocal microscopy and spectrofluorometry. RESULTS: We detected an increased PKM2/PKM1 ratio in PH-Fibs compared with normal subjects. PKM2 inhibition reversed the glycolytic status of PH-Fibs, decreased their cell proliferation, and attenuated macrophage interleukin-1ß expression. Furthermore, normalizing the PKM2/PKM1 ratio in PH-Fibs by miR-124 overexpression or PTBP1 knockdown reversed the glycolytic phenotype (decreased the production of glycolytic intermediates and byproducts, ie, lactate), rescued mitochondrial reprogramming, and decreased cell proliferation. Pharmacological manipulation of PKM2 activity with TEPP-46 and shikonin or treatment with histone deacetylase inhibitors produced similar results. CONCLUSIONS: In PH, miR-124, through the alternative splicing factor PTBP1, regulates the PKM2/PKM1 ratio, the overall metabolic, proliferative, and inflammatory state of cells. This PH phenotype can be rescued with interventions at various levels of the metabolic cascade. These findings suggest a more integrated view of vascular cell metabolism, which may open unique therapeutic prospects in targeting the dynamic glycolytic and mitochondrial interactions and between mesenchymal inflammatory cells in PH.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Hipertensión Pulmonar/patología , MicroARNs/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Piruvato Quinasa/metabolismo , Empalme Alternativo , Animales , Antagomirs/metabolismo , Bovinos , Proliferación Celular , Endotelio Vascular/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Glucólisis , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Hipertensión Pulmonar/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Naftoquinonas/farmacología , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Interferencia de ARN
9.
Mol Med Rep ; 16(5): 7221-7228, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28944929

RESUMEN

Glioblastoma multiforme (GBM) is the leading type of brain tumor, exhibiting unlimited proliferation and invasion potential. The present study indicated that a high expression level of miR­132 was detected in the neural subtype of GBM and predicted an unfavorable prognosis for patients from The Cancer Genome Atlas cohort (n=526). Cox hazard regression analysis demonstrated miR­132 as an independent prognostic indicator for GBM patients. Further in vitro experiments indicated that miR­132 promoted the proliferation and sphere formation of U87 cells. Unsupervised hierarchical clustering analysis was performed to compare differently expressed genes between two Gene Expression Omnibus (GEO) datasets and Gene Ontology analysis was applied to evaluate the significant signaling pathways modulated by miR­132 in GBM cells within a genetic bioinformatic lab, the Gene­Cloud of Biotechnology Information. By combining the results based on GEO datasets and the miRNA bioinformatic prediction, polypyrimidine tract­binding protein 2 (PTBP2), a brain tissue­specific post­transcriptional protein, was identified as a potential downstream target of miR­132 in GBM. Thus, miR­132 overexpression in GBM cells predicted an unfavorable outcome for patients, and sustained the proliferation and self­renewal abilities of GBM cells in an miR­132/PTBP2 signaling pathway.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Antagomirs/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Proliferación Celular , Autorrenovación de las Células , Estudios de Cohortes , Biología Computacional , Bases de Datos Genéticas , Femenino , Redes Reguladoras de Genes , Glioblastoma/genética , Glioblastoma/mortalidad , Humanos , Estimación de Kaplan-Meier , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Oncogenes/genética , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Pronóstico , Modelos de Riesgos Proporcionales , Transducción de Señal
10.
PLoS One ; 12(3): e0170991, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28282372

RESUMEN

Multiplexed immunofluorescent testing has not entered into diagnostic neuropathology due to the presence of several technical barriers, amongst which includes autofluorescence. This study presents the implementation of a methodology capable of overcoming the visual challenges of fluorescent microscopy for diagnostic neuropathology by using automated digital image analysis, with long term goal of providing unbiased quantitative analyses of multiplexed biomarkers for solid tissue neuropathology. In this study, we validated PTBP1, a putative biomarker for glioma, and tested the extent to which immunofluorescent microscopy combined with automated and unbiased image analysis would permit the utility of PTBP1 as a biomarker to distinguish diagnostically challenging surgical biopsies. As a paradigm, we utilized second resections from patients diagnosed either with reactive brain changes (pseudoprogression) and recurrent glioblastoma (true progression). Our image analysis workflow was capable of removing background autofluorescence and permitted quantification of DAPI-PTBP1 positive cells. PTBP1-positive nuclei, and the mean intensity value of PTBP1 signal in cells. Traditional pathological interpretation was unable to distinguish between groups due to unacceptably high discordance rates amongst expert neuropathologists. Our data demonstrated that recurrent glioblastoma showed more DAPI-PTBP1 positive cells and a higher mean intensity value of PTBP1 signal compared to resections from second surgeries that showed only reactive gliosis. Our work demonstrates the potential of utilizing automated image analysis to overcome the challenges of implementing fluorescent microscopy in diagnostic neuropathology.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Microscopía Fluorescente , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Adolescente , Adulto , Anciano , Animales , Anticuerpos Monoclonales/inmunología , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Glioma/diagnóstico , Glioma/genética , Glioma/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/inmunología , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Ratones , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/inmunología , Interferencia de ARN , Estudios Retrospectivos , Adulto Joven
11.
Br J Cancer ; 116(7): 903-911, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28222070

RESUMEN

BACKGROUND: Human polypyrimidine tract binding protein 3 (PTBP3) was first discovered in 1999 and has been well characterised as a differentiation regulator. However, its role in human cancer has rarely been reported. Our previous study revealed increased PTBP3 protein level in gastric cancer tissues. Downregulation of PTBP3 suppressed the proliferation and differentiation of gastric cancer cells in vivo. METHODS: PTBP3 mRNA levels in human gastric cancer and adjuvant non-tumour tissues were detected. Apoptosis and 5-FU effect were determined in PTBP3-silenced gastric cancer cells. Underlying molecular mechanisms were investigated. RESULTS: MRNA expression of PTBP3 was upregulated in gastric cancer tissues, especially in those at an advanced stage. PTBP3 silencing led to apoptosis, under which modulation of PTB and thereby switch of Bcl-x pre-mRNA splicing pattern might be an important mechanism. Further research found that inhibition of PTBP3 expression enhanced the chemosensitivity of gastric cancer cells towards 5-FU treatment. This was mediated by reduced expression of histone deacetylase 6 (HDAC6), which further inhibited the phosphorylation of Akt and the expression of thymidylate synthase (TYMS), the critical determinant of 5-FU cytotoxicity. CONCLUSIONS: PTBP3 might serve as a biomarker of gastric cancer or potential target for anti-cancer therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Fluorouracilo/farmacología , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Neoplasias Gástricas/patología , Anciano , Antimetabolitos Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Proliferación Celular/efectos de los fármacos , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metástasis Linfática , Masculino , Invasividad Neoplásica , Estadificación de Neoplasias , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Pronóstico , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Células Tumorales Cultivadas
12.
Cell Rep ; 17(1): 104-113, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681424

RESUMEN

The fidelity of RNA splicing is maintained by a network of factors, but the molecular mechanisms that govern this process have yet to be fully elucidated. We previously found that TDP-43, an RNA-binding protein implicated in neurodegenerative disease, utilizes UG microsatellites to repress nonconserved cryptic exons and prevent their incorporation into mRNA. Here, we report that two well-characterized splicing factors, polypyrimidine tract-binding protein 1 (PTBP1) and polypyrimidine tract-binding protein 2 (PTBP2), are also nonconserved cryptic exon repressors. In contrast to TDP-43, PTBP1 and PTBP2 utilize CU microsatellites to repress both conserved tissue-specific exons and nonconserved cryptic exons. Analysis of these conserved splicing events suggests that PTBP1 and PTBP2 repression is titrated to generate the transcriptome diversity required for neuronal differentiation. We establish that PTBP1 and PTBP2 are members of a family of cryptic exon repressors.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Empalme del ARN , ARN Mensajero/genética , Transcriptoma , Animales , Secuencia de Bases , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular , Exones , Células HeLa , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Ratones , Repeticiones de Microsatélite , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
13.
Mol Cell Biol ; 35(4): 758-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25512611

RESUMEN

The output of alternative splicing depends on the cooperative or antagonistic activities of several RNA-binding proteins (RBPs), like Ptbp1 and Esrp1 in Xenopus. Fine-tuning of the RBP abundance is therefore of prime importance to achieve tissue- or cell-specific splicing patterns. Here, we addressed the mechanisms leading to the high expression of the ptbp1 gene, which encodes Ptbp1, in Xenopus epidermis. Two splice isoforms of ptbp1 mRNA differ by the presence of an alternative exon 11, and only the isoform including exon 11 can be translated to a full-length protein. In vivo minigene assays revealed that the nonproductive isoform was predominantly produced. Knockdown experiments demonstrated that Esrp1, which is specific to the epidermis, strongly stimulated the expression of ptbp1 by favoring the productive isoform. Consequently, knocking down esrp1 phenocopied ptbp1 inactivation. Conversely, Ptbp1 repressed the expression of its own gene by favoring the nonproductive isoform. Hence, a complex posttranscriptional mechanism controls Ptbp1 abundance in Xenopus epidermis: skipping of exon 11 is the default splicing pattern, but Esrp1 stimulates ptbp1 expression by favoring the inclusion of exon 11 up to a level that is limited by Ptbp1 itself. These results decipher a posttranscriptional mechanism that achieves various abundances of the ubiquitous RBP Ptbp1 in different tissues.


Asunto(s)
Proteínas Anfibias/genética , Epidermis/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Xenopus laevis/genética , Empalme Alternativo , Proteínas Anfibias/antagonistas & inhibidores , Proteínas Anfibias/metabolismo , Animales , Embrión no Mamífero , Epidermis/crecimiento & desarrollo , Exones , Genotipo , Intrones , Fenotipo , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/metabolismo
14.
J Clin Invest ; 124(7): 2861-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24865424

RESUMEN

Tissue-specific alternative splicing is critical for the emergence of tissue identity during development, yet the role of this process in malignant transformation is undefined. Tissue-specific splicing involves evolutionarily conserved, alternative exons that represent only a minority of the total alternative exons identified. Many of these conserved exons have functional features that influence signaling pathways to profound biological effect. Here, we determined that lineage-specific splicing of a brain-enriched cassette exon in the membrane-binding tumor suppressor annexin A7 (ANXA7) diminishes endosomal targeting of the EGFR oncoprotein, consequently enhancing EGFR signaling during brain tumor progression. ANXA7 exon splicing was mediated by the ribonucleoprotein PTBP1, which is normally repressed during neuronal development. PTBP1 was highly expressed in glioblastomas due to loss of a brain-enriched microRNA (miR-124) and to PTBP1 amplification. The alternative ANXA7 splicing trait was present in precursor cells, suggesting that glioblastoma cells inherit the trait from a potential tumor-initiating ancestor and that these cells exploit this trait through accumulation of mutations that enhance EGFR signaling. Our data illustrate that lineage-specific splicing of a tissue-regulated alternative exon in a constituent of an oncogenic pathway eliminates tumor suppressor functions and promotes glioblastoma progression. This paradigm may offer a general model as to how tissue-specific regulatory mechanisms can reprogram normal developmental processes into oncogenic ones.


Asunto(s)
Empalme Alternativo , Anexina A7/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Linaje de la Célula/genética , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Receptores ErbB/genética , Receptores ErbB/metabolismo , Exones , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Glioblastoma/patología , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Patológica/genética , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Transducción de Señal/genética , Células Tumorales Cultivadas
15.
BMB Rep ; 47(4): 233-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24286314

RESUMEN

Polypyrimidine tract-binding protein 1 (PTBP1) and its brainspecific homologue, PTBP2, are associated with pre-mRNAs and influence pre-mRNA processing, as well as mRNA metabolism and transport. They play important roles in neural differentiation and glioma development. In our study, we detected the expression of the two proteins in glioma cells and predicted that they may be sumoylated using SUMOplot analyses. We confirmed that PTBP1 and PTBP2 can be modified by SUMO1 with co-immunoprecipitation experiments using 293ET cells transiently co-expressing SUMO1 and either PTBP1 or PTBP2. We also found that SUMO1 modification of PTBP2 was enhanced by Ubc9 (E2). The mutation of the sumoylation site (Lys137) of PTBP2 markedly inhibited its modification by SUMO1. Interestingly, in T98G glioma cells, the level of sumoylated PTBP2 was reduced compared to that of normal brain cells. Overall, this study shows that PTBP2 is posttranslationally modified by SUMO1.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Procesamiento Proteico-Postraduccional , Proteína SUMO-1/metabolismo , Células HEK293 , Humanos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , Células Tumorales Cultivadas
16.
Cell Death Differ ; 21(1): 161-71, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24141718

RESUMEN

Post-transcriptional control of gene expression is mediated by the interaction of RNA-binding proteins with their cognate mRNAs that specifically regulate their stability, localization and translation. mRNA-binding proteins are multifunctional and it has been proposed therefore that a combinatorial RNA-binding protein code exists that allows specific protein sub-complexes to control cytoplasmic gene expression under a range of pathophysiological conditions. We show that polypyrimidine tract-binding protein (PTB) is central to one such complex that forms in apoptotic cells. Thus, during apoptosis initiated by TNF-related apoptosis inducing ligand there is a change in the repertoire of RNA-binding proteins with which PTB interacts. We show that altering the cellular levels of PTB and its binding partners, either singly or in combination, is sufficient to directly change the rates of apoptosis with increased expression of PTB, YBX1, PSF and NONO/p54(nrb) accelerating this process. Mechanistically, we show that these proteins post-transcriptionally regulate gene expression, and therefore apoptotic rates, by interacting with and stimulating the activity of RNA elements (internal ribosome entry segments) found in mRNAs that are translated during apoptosis. Taken together, our data show that PTB function is controlled by a set of co-recruited proteins and importantly provide further evidence that it is possible to dictate cell fate by modulating cytoplasmic gene expression pathways alone.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Núcleo Celular/metabolismo , Ciclina T/genética , Ciclina T/metabolismo , Proteínas de Unión al ADN , Células HeLa , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Células MCF-7 , Proteínas Asociadas a Matriz Nuclear/antagonistas & inhibidores , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/antagonistas & inhibidores , Factores de Transcripción de Octámeros/genética , Factores de Transcripción de Octámeros/metabolismo , Factor de Empalme Asociado a PTB , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo
17.
J Cell Biol ; 193(3): 509-20, 2011 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-21518792

RESUMEN

Alternative splicing contributes largely to cell differentiation and functional specification. We previously reported that the RNA-binding protein RBM4 antagonizes the activity of splicing factor PTB to modulate muscle cell-specific exon selection of α-tropomyosin. Here we show that down-regulation of PTB and its neuronal analogue nPTB during muscle cell differentiation may involve alternative splicing-coupled nonsense-mediated mRNA decay. RBM4 regulates PTB/nPTB expression by activating exon skipping of their transcripts during myogenesis. Moreover, RBM4 and PTB target a common set of transcripts that undergo muscle cell-specific alternative splicing. Overexpression of RBM4 invariably promoted expression of muscle cell-specific isoforms, which recapitulated in vivo alternative splicing changes during muscle differentiation, whereas PTB acted oppositely to RBM4 in expression of mRNA isoforms specific for late-stage differentiation. Therefore, RBM4 may synergize its effect on muscle cell-specific alternative splicing by down-regulating PTB expression and antagonizing the activity of PTB in exon selection, which highlights a hierarchical role for RBM4 in a splicing cascade that regulates myogenesis.


Asunto(s)
Regulación hacia Abajo , Regulación de la Expresión Génica , Músculos/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteínas de Unión al ARN/biosíntesis , Empalme Alternativo , Animales , Línea Celular , Exones , Humanos , Ratones , Desarrollo de Músculos , Neuronas/metabolismo , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Isoformas de Proteínas , ARN Mensajero/metabolismo
18.
Exp Cell Res ; 317(1): 94-106, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20875406

RESUMEN

Exon 6B from the chicken ß-tropomyosin pre-mRNA is alternatively spliced during myogenic differentiation. Exon 6B is excluded in mRNA from myoblasts and included in mRNA from myotubes. We investigated the regulation of exon 6B inclusion ex vivo in a quail myogenic cell line, which behaves as myoblasts in undifferentiated state and as myotubes after differentiation. We show that the ß-tropomyosin exon 6B is a novel target of CUG-BP and ETR-3-like factor (CELF). Overexpression of CELF proteins in myoblasts activates splicing of exon 6B. Using a dominant-negative form of CELF4, we demonstrate that CELF proteins are involved in switching splicing from exon 6A towards exon 6B inclusion during myogenic differentiation. We also found that polypyrimidine tract binding protein (PTB) is required for splicing repression of exon 6B in myoblasts. CELF and PTB proteins exhibit antagonistic properties toward inclusion of exon 6B during myogenic differentiation. Our results suggest that a change in the protein level of CUGBP1 and PTB proteins, associated with a distinct pattern of PTB during the transition from myoblasts to myotubes is one of the parameters involved in regulating splicing of exon 6B during myogenesis.


Asunto(s)
Empalme Alternativo/genética , Desarrollo de Músculos/genética , Proteína de Unión al Tracto de Polipirimidina/fisiología , Proteínas de Unión al ARN/fisiología , Tropomiosina/genética , Empalme Alternativo/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Embrión de Pollo , Pollos , Exones/genética , Células HeLa , Humanos , Desarrollo de Músculos/efectos de los fármacos , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Codorniz/embriología , Precursores del ARN/metabolismo , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Tropomiosina/metabolismo
19.
Mol Cell Biol ; 31(5): 935-54, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21189287

RESUMEN

Prevention of skipping of exon 7 during pre-mRNA splicing of Survival Motor Neuron 2 (SMN2) holds the promise for cure of spinal muscular atrophy (SMA), a leading genetic cause of infant mortality. Here, we report T-cell-restricted intracellular antigen 1 (TIA1) and TIA1-related (TIAR) proteins as intron-associated positive regulators of SMN2 exon 7 splicing. We show that TIA1/TIAR stimulate exon recognition in an entirely novel context in which intronic U-rich motifs are separated from the 5' splice site by overlapping inhibitory elements. TIA1 and TIAR are modular proteins with three N-terminal RNA recognition motifs (RRMs) and a C-terminal glutamine-rich (Q-rich) domain. Our results reveal that any one RRM in combination with a Q domain is necessary and sufficient for TIA1-associated regulation of SMN2 exon 7 splicing in vivo. We also show that increased expression of TIA1 counteracts the inhibitory effect of polypyrimidine tract binding protein, a ubiquitously expressed factor recently implicated in regulation of SMN exon 7 splicing. Our findings expand the scope of TIA1/TIAR in genome-wide regulation of alternative splicing under normal and pathological conditions.


Asunto(s)
Empalme Alternativo , Exones , Atrofia Muscular Espinal/genética , Proteínas de Unión a Poli(A)/metabolismo , Proteínas de Unión al ARN/metabolismo , Línea Celular , Humanos , Intrones , Proteínas de Unión a Poli(A)/genética , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Antígeno Intracelular 1 de las Células T
20.
Nucleic Acids Res ; 38(20): 7068-78, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20576698

RESUMEN

The physiological and behavioral circadian rhythms of most creatures are controlled by a harmony of functional relationships between clock genes. In mammals, several core clock genes show rhythmic profiles of their mRNA and protein expression. Among them, Rev-erb α functions as a transcriptional repressor, affecting expression patterns of other clock genes. For the continuous and robust oscillation of the molecular clock system, the levels of Rev-erb α protein are expected to be tightly regulated with the correct timing. Here, we demonstrate that Rev-erb α has an internal ribosomal entry site (IRES) in its 5' untranslated region. Furthermore, we demonstrate that heterogeneous nuclear ribonucleoprotein Q and polypyrimidine tract-binding protein (PTB) modulate the IRES-mediated translation of Rev-erb α. We suggest that the rhythmic binding affinity of hnRNP Q to the Rev-erb α IRES and the change in PTB cytosolic levels lead to maintenance of the oscillation profile of the Rev-erb α protein.


Asunto(s)
Regiones no Traducidas 5' , Ritmo Circadiano/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Biosíntesis de Proteínas , Animales , Regulación de la Expresión Génica , Cinética , Ratones , Células 3T3 NIH , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Interferencia de ARN , Sirolimus/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...