Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Life Sci ; 330: 121977, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37499934

RESUMEN

BACKGROUND: Bleomycin (BLM) is a chemotherapeutic agent with potent antitumor activity against the tumor. However, lung fibrosis is the main drawback that limits BLM use. Tumor targeted, safe, efficient and natural delivery of BLM is important to increase the effectiveness and reduce the toxic side effects. Although tumor derived Exosomes (Exo), provide a potential vehicle for in vivo drug delivery due to their cell tropism. This study primarily focuses on generating a natural delivery platform for Exo loaded with BLM and testing its therapeutic efficacy against cancer. METHODS: Exosomes were isolated from cancer cells and incubated with BLM. Exo were characterized by transmission electron microscopy, western blot analysis and nanoparticle tracking analysis. We performed in vitro and in vivo analyses to evaluate the effect of Exo-BLM. RESULTS: Exosomes loaded with BLM are highly cancer targeting and cause the cytotoxicity of tumor cells by ROS. The fluorescence images showed that Exo-BLM accumulated in cancer cells. The results revealed that Exo-BLM induces tumor cell apoptosis by the caspase pathway. In vivo, the treatment of Exo-BLM showed targeted ability and enhanced the antitumor activity. CONCLUSION: This study provides an avenue for specific BLM therapeutics with minimal side effects.


Asunto(s)
Exosomas , Fibrosis Pulmonar , Humanos , Bleomicina/farmacología , Exosomas/metabolismo , Línea Celular Tumoral , Apoptosis , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo
2.
Biomed Res Int ; 2022: 1557010, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677097

RESUMEN

Ischemic stroke is a devastating CNS insult with few clinical cures. Poor understanding of underlying mechanistic network is the primary limitation to develop novel curative therapies. Extracellular accumulation of granzyme B subsequent ischemia promotes neurodegeneration. Inhibition of granzyme B can be one of the potent strategies to mitigate neuronal damage. In present study, we investigated the effect of murine Serpina3n and human (homolog) SERPINA3 against cerebral ischemia through granzyme B inactivation. Recombinant Serpina3n/SERPINA3 were expressed by transfected 293 T cells, and eluted proteins were examined for postischemic influence both in vitro and in vivo. During in vitro test, Serpina3n was found effective enough to inhibit granzyme B, while SERPINA3 was ineffectual to counter cytotoxic protease. Treatment of hypoxic culture with recombinant Serpina3n/SERPINA3 significantly increased cell viability in dosage-dependent manner, recorded maximum at the highest concentration (4 mM). Infarct volume analysis confirmed that 50 mg/kg dosage of exogenous Serpina3n was adequate to reduce disease severity, while SERPINA3 lacked behind in analeptic effect. Immunohistochemical test, western blot analysis, and protease activity assay's results illustrated successful diffusion of applied protein to the ischemic lesion and reactivity with the target protease. Taken together, our findings demonstrate therapeutic potential of Serpina3n by interfering granzyme B-mediated neuronal death subsequent cerebral ischemia.


Asunto(s)
Isquemia Encefálica , Serpinas , Proteínas de Fase Aguda/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Infarto Cerebral , Granzimas , Humanos , Ratones , Neuronas/metabolismo , Serpinas/metabolismo , Linfocitos T Citotóxicos
3.
Cancer Gene Ther ; 29(3-4): 292-303, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33707625

RESUMEN

Long non-coding RNAs (lncRNAs) have recently been reported to act as crucial regulators and prognostic biomarkers of human tumorigenesis. Based on microarray data, RP11-301G19.1 was previously identified as an upregulated lncRNA during B cell development. However, the effect of RP11-301G19.1 on multiple myeloma (MM) cells remains unclear. In the present study, the effects of RP11-301G19.1 on tumour progression were ascertained both in vitro and in vivo. Our results demonstrated that RP11-301G19.1 was upregulated in MM cell lines and that its downregulation inhibited the proliferation and cell cycle progression and promoted the apoptosis of MM cells. Bioinformatic analysis and luciferase reporter assay results revealed that RP11-301G19.1 can upregulate the miR-582-5p-targeted gene HMGB2 as a competing endogenous RNA (ceRNA). Furthermore, Western blot results indicated that RP11-301G19.1 knockdown decreased the levels of PI3K and AKT phosphorylation without affecting their total protein levels. Additionally, in a xenograft model of human MM, RP11-301G19.1 knockdown significantly inhibited tumour growth by downregulating HMGB2. Overall, our data demonstrated that RP11-301G19.1 is involved in MM cell proliferation by sponging miR-582-5p and may serve as a therapeutic target for MM.


Asunto(s)
MicroARNs , Mieloma Múltiple , ARN Largo no Codificante , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Mieloma Múltiple/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factores de Transcripción/genética
4.
Immunology ; 165(1): 88-98, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34435359

RESUMEN

B-cell development undergoes a series of steps from the bone marrow to the secondary lymphoid organs. A defect in B-cell development can lead to immunodeficiency or malignant disorders, such as leukaemia or lymphoma. Long non-coding RNAs have been reported to act as important regulators of many pathological processes. However, very little is known regarding the role of lncRNAs during B-cell development and the regulation of their expression. In this study, we explored the expression and role of lncRNA Gme00492 in B-cell development. We observed that lnc00492 was highly expressed in B-cell development and primarily expressed in the nucleus. Lnc00492-deficient mice had fewer marginal zone B cells in the spleen, likely due to a developmental block. Importantly, lnc00492 interacts with CTBP1 and targets it for ubiquitination and degradation during B-cell development, whereas the transcriptional corepressor factor CTBP1 plays a critical role in Notch2 signalling. Thus, we identified a novel regulatory axis between lnc00492 and CTBP1 in B cells, suggesting that lnc00492 is essential for marginal zone B-cell development.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Diferenciación Celular/genética , Linfopoyesis/genética , ARN Largo no Codificante/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Linfocitos B/inmunología , Biomarcadores , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Diferenciación Celular/inmunología , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Inmunofenotipificación , Ratones , Ratones Noqueados , Modelos Biológicos , Unión Proteica , Receptor Notch2/metabolismo , Transducción de Señal , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Ubiquitinación
5.
Apoptosis ; 26(11-12): 628-638, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34748127

RESUMEN

ANNEXIN V belongs to a family of phospholipid binding proteins which is able to bind to negatively charged phospholipids such as phosphatidylserine (PS) in the presence of a high affinity Ca2+ ion. When apoptosis occurs, even at early stage, PS will be exposed to the outside of the cell surface from the cytoplasm side of membrane leaflets., Therefore ANNEXIN V has been suggested as a bio-marker for imaging early apoptotic events of various cell death including those in disease conditions. However, most ANNEXIN V-based apoptotic detecting techniques were in vitro approaches. Here, we presented a new BRET (Bioluminescence Resonance Energy Transfer) based genetic coded biosensor by fusing ANNEXIN V and a BRET version of NanoLuc (teLuc) for both in vitro and in vivo apoptosis detection. The BRET feature of this new sensor makes it convenient to be applied to both conventional fluorescent-based in vitro apoptosis detection and bioluminescence-based animal live imaging for in vivo study. Because of its robust bioluminescence signal, it is possible to perform the evaluation of the disease-induced apoptotic damage and recovery process directly at deep tissue level in live animal.


Asunto(s)
Apoptosis , Técnicas Biosensibles , Animales , Anexina A5/genética , Luciferasas
6.
Cell Immunol ; 363: 104321, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33773377

RESUMEN

B cells play a crucial role in immune responses. The main functions include B cell protective antibody production, inflammation reduction, activation and proliferation. Long non-coding RNAs (lncRNAs) have been reported to act as important regulators of many pathological processes. However, few lncRNAs have been reported to affect B cell function. In this study, we explored the expression and role of lncRNA 2900052N01Rik (lnc-290) in lipopolysaccharide (LPS)-induced B cells purified from mouse spleens in vitro. Here, we confirmed that lnc-290 was highly expressed in B cells stimulated by LPS. Knockdown of lnc-290 inhibited the expression of CD69/CD86 and the growth of B cells. Moreover, down-regulated lnc-290 reduced B cell differentiation and immunoglobulin production in vitro. In addition, we found that lnc-290 regulated LPS-induced B cell activation via the NF-κB/ERK pathways. Interestingly, abnormal lnc-290 expression did not alter the B cell activation or proliferation induced by IL-4 or CD40/CD40L. Accordingly, these results indicated, for the first time, that lnc-290 down-regulation inhibits LPS-induced B cell proliferation, activation and differentiation by blocking the LPS/TLR4 signaling pathway. Together, the in vitro data demonstrate that lnc-290 participated in the inflammation and tissue damage mediated by LPS-activated B cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/fisiología , ARN Largo no Codificante/genética , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Linfocitos B/metabolismo , Antígeno B7-2/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Lectinas Tipo C/genética , Lipopolisacáridos/farmacología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Bazo/inmunología , Receptor Toll-Like 4/inmunología
7.
Int Immunopharmacol ; 95: 107532, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33756230

RESUMEN

Human multiple myeloma (MM) is a currently incurable haematopoietic malignancies. Our research investigate the anti-tumour effect of iguratimod (IGU) encapsulated in poly(lactic-co-glycolic acid) PLGA nanoparticles (IGU-PLGA-NPs) on MM cells in vitro and in vivo. A significant inhibitory effect of IGU-PLGA-NPs on MM cancer cells and MM CSCs was demonstrated by the Cell Counting Kit-8 (CCK-8) assay. Treatment with IGU-PLGA-NPs induced significant cell cycle arrest at G1 in MM cells and reduced tumour colony formation in MM CSCs. Mechanistically, IGU-PLGA-NPs increase apoptosis in MM cells by activating Caspase-dependent signalling pathway to increase the levels of bax, cytochrome c (cyt-c), caspase-9 and caspase-3 proteins. Moreover, IGU-PLGA-NPs effectively increase ROS production assayed using a DCFH-DA fluorescent probe in MM cells. The data indicate that IGU-PLGA-NPs induce a significant reduction in the tumour volume and a marked increase in the survival rate in a mouse model of multiple myeloma. Overall, our findings indicate that IGU-PLGA-NPs are a potential therapeutic strategy that may contribute to the therapy of MM and elimination of MM CSCs in future clinical trials.


Asunto(s)
Antineoplásicos/administración & dosificación , Cromonas/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Nanopartículas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Sulfonamidas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Mieloma Múltiple/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Mol Neurobiol ; 58(7): 3362-3373, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33694060

RESUMEN

Glioma is the most common type of solid tumour affecting the central nervous system, and the survival rate of patients with glioma is low. However, the mechanism associated with glioma progression remains unclear. Growing evidence suggests that lncRNAs play essential roles in the initiation and progression of tumours, including gliomas. In the present study, we identified and verified the expression of the novel lncRNA RP11-279C4.1 by analyzing the TANRIC database and performing qRT-PCR assays, the results of which revealed its upregulation in glioma tissues and cell lines. The results of multiple functional experiments demonstrated that RP11-279C4.1 knockdown inhibited glioma malignant phenotypes, including cell proliferation, migration, invasion and cell self-renew ability in vitro. In addition, RP11-279C4.1 downregulation suppressed tumour growth in vivo. Mechanistically, RP11-279C4.1 induced CBX3 activation via competitively sponging miR-1273g-3p, and rescue assay results confirmed the importance of the RP11-279C4.1/miR-1273g-3p/CBX3 axis. Overall, the results of our present study demonstrated that RP11-279C4.1 functions as an oncogene that promotes tumour progression by modulating the miR-1273g-3p/CBX3 axis in glioma, suggesting that RP11-279C4.1 may be a novel therapeutic target for glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Glioma/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Femenino , Glioma/patología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Biochem Biophys Res Commun ; 525(2): 469-476, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32107003

RESUMEN

AIM: The fragile X mental retardation protein (FMRP), the product of the FMR1 gene, is responsible for the fragile X syndrome (FXS). FMRP regulates miRNA expression and is involved in miRNA-mediated gene silencing. However, the question of whether FMRP is, in turn, regulated by miRNAs remains unanswered. MAIN METHODS: We detected the FMRP expression pattern by in situ hybridization. MiR-315 overexpression and knockout models were generated by germ-line transformation and ends-out homologous recombination, respectively. Western blotting and immunohistochemistry were used to detect Drosophila FMRP (dFMRP) and a Luciferase reporter assay was used to confirm the regulation of dfmr1 mRNA by mir-315. Synaptic structural quantification and electrophysiological methods were used to compare synaptic functions among groups. KEY FINDINGS: Here, we determined that the transcription product of dFMR1, the Drosophila homologue of FMR1, is a direct target of miR-315. MiR-315 is mainly expressed in the nervous system of Drosophila. Flies overexpressing miR-315 showed pupation defects and reduced hatching rates. A homozygous miR-315 knockout status is embryonic lethal in flies. These observations indicate that miR-315 is a key regulator of the Drosophila nervous system. Furthermore, computational prediction and cell-based luciferase and in vivo assays demonstrated that dfmr1 is directly targeted by miR-315. Lastly, using the neuromuscular junction as a model, we found that miR-315 regulates synaptic structure and transmission by targeting dfmr1. SIGNIFICANCE: These findings provide compelling evidence that miR-315 targets dfmr1 in the Drosophila nervous system, acting as a regulatory factor for the fine-tuned modulation of FMRP expression.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Regulación del Desarrollo de la Expresión Génica , Animales , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Neurogénesis , Unión Neuromuscular/genética , Sinapsis/genética
10.
J Drug Target ; 28(4): 368-378, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31790278

RESUMEN

Serpina3n is a secretory serine protease inhibitor belonging to clade "a" exhibiting unique structural and physiological characteristics, playing significant roles ranging from complement cascade, apoptosis, wound healing to Alzheimer by inhibiting a wide range of proteases. Recently studies have reported its significant roles during various pathologies. Although its full range of potential applications are yet to reveal, its reported implications particularly in CNS insults are making it potential therapeutical approach. Here we aim to draw together the literature shedding light on the potential therapeutical applications of Serpina3n/SERPINA3 in various diseases and a brief comparison between murine Serpina3n and its human ortholog SERPINA3 (α1-antichymotrypsin) accounting their biological roles and significance.


Asunto(s)
Preparaciones Farmacéuticas/administración & dosificación , Serpinas/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Humanos , Inhibidores de Serina Proteinasa/farmacología , Cicatrización de Heridas/efectos de los fármacos
11.
J Cell Physiol ; 234(12): 21937-21946, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31054187

RESUMEN

Accumulating evidence suggests that noncoding RNAs play a vital role in cancer biology. Circular RNAs (circRNAs), a newly defined class of endogenously widespread noncoding RNAs, have been intensively reported to influence cell function and development, and even cancer prognosis by sponging microRNAs in various types of cancer. Nevertheless, the circRNAs research in hepatocellular carcinoma (HCC) still remains far insufficient. Herein, we investigated the role of a newly defined circRNAs, circ_0005075, in HCC development. We found circ_0005075 was upregulated in HCC tissues. HCC progression was suppressed by downregulation of circ_0005075 in vitro and in vivo, and the suppression was partially reversed by inhibition of microRNA-335 (miR-335) expression. Further, we found the expression of mitogen-activated protein kinase 1 (MAPK1) was substantially regulated by circ_0005075 and miR-335. Mechanically, it was demonstrated that circ_0005075 could directly bind to miR-335 and miR-335 could bind to MAPK1. Our data provide evidence that circ_0005705 promotes the HCC progression by sponging miR-335 and further regulating MAPK1 expression.


Asunto(s)
Carcinoma Hepatocelular/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , ARN Circular/genética , Animales , Progresión de la Enfermedad , Xenoinjertos , Humanos , Neoplasias Hepáticas/patología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética
12.
Life Sci ; 218: 31-37, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30528775

RESUMEN

AIMS: Fragile X mental retardation protein (FMRP) plays a vital role in mRNA trafficking and translation inhibition to regulate the synthesis of local proteins in neuronal axons and dendritic terminals. However, there are no reports on microRNA (miRNA)-mediated regulation of FMRP levels in Drosophila. Here, we aimed to identify miRNAs regulating FMRP levels in Drosophila. MAIN METHODS: Using online software, we predicted and selected 11 miRNAs potentially acting on the Drosophila fragile X mental retardation 1 (dFMR1) transcript. These candidates were screened for modulation of dFMR1 transcript levels at the cellular level using a dual luciferase reporter system. In addition, we constructed a transgenic Drosophila model overexpressing miR-219 in the nervous system and quantified dFMRP by western blotting. The neuromuscular junction phenotype in the model was studied by immunofluorescence staining. KEY FINDINGS: Among the 11 miRNAs screened, miR-219 and miR-960 reduced luciferase gene activity by binding to the 3'-UTR of the dFMR1 transcript. Mutation of the miR-219 or miR-960 binding sites on the transcript resulted in complete or partial elimination of the miRNA-induced repression. Western blots revealed that dFMRP expression was decreased in the miR-219 overexpression model (Elav>miR-219). Drosophila larvae overexpressing miR-219 showed morphological abnormalities at the neuromuscular junction (increased synaptic boutons and synaptic branches). This finding is consistent with some phenotypes observed in dfmr1 mutants. SIGNIFICANCE: Our results suggest that miR-219 regulates dFMR1 expression in Drosophila and is involved in fragile X syndrome pathogenesis. Collectively, these findings expand the current understanding of miRNA-mediated regulation of target molecule-related functions.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Unión Neuromuscular/patología , Animales , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Unión Neuromuscular/metabolismo
13.
Exp Eye Res ; 160: 21-30, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28434813

RESUMEN

Teleost fish continues to grow their eyes throughout life with the body size. In Astatotilapia burtoni, the fish retina increases by adding new retinal cells at the ciliary marginal zone (CMZ) and in the outer nuclear layer (ONL). Cell proliferation at both sites exhibits a daily rhythm in number of dividing cells. To understand how this diurnal rhythm of new cell production is controlled in retinal progenitor cells, we studied the transcription pattern of clock genes in retina, including clock1a, clock1b, bmal1a (brain and muscle ARNT-Like), and per1b (period1b). We found that these genes have a strong diurnal rhythmic transcription during light-dark cycles but not in constant darkness. An oscillation in pcna transcription was also observed during light-dark cycles, but again not in constant darkness. Our results also indicate an association between Clock proteins and the upstream region of pcna (proliferating cellular nuclear antigen) gene. A luciferase reporter assay conducted in an inducible clock knockdown cell line further demonstrated that the mutation on predicted E-Boxes in pcna promoter region significantly attenuated the transcriptional activation induced by Clock protein. These results suggested that the diurnal rhythmic expression of clock genes in A. burtoni retina could be light dependent and might contribute to the daily regulation of the proliferation of the retina progenitors through key components of cell cycle machinery, for instance, pcna.


Asunto(s)
Proteínas CLOCK/genética , Regulación de la Expresión Génica , Antígeno Nuclear de Célula en Proliferación/genética , ARN/genética , Retina/metabolismo , Animales , Western Blotting , Proteínas CLOCK/biosíntesis , División Celular , Línea Celular , Proliferación Celular , Cíclidos , Ritmo Circadiano/fisiología , Inmunohistoquímica , Hibridación in Situ , Luz , Ratones , Modelos Animales , Fotoperiodo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/citología , Células Madre/citología , Células Madre/metabolismo , Transcripción Genética
14.
Int J Mol Sci ; 17(11)2016 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-27801803

RESUMEN

MicroRNAs (miRNAs) are implicated in the regulation of tumor progression and stemness of cancer stem-like cells. Recently, miR-92a-3p was reported to be up-regulated in human glioma samples. Nevertheless, the precise role of miR-92a-3p in glioma cells and glioma stem-like cells (GSCs) has not been fully elucidated. It is necessary to clarify the function of miR-92a-3p in glioma and GSCs to develop novel therapeutic approaches for glioma patients. In the present study, we applied methyl-thiazolyl-tetrazolium (MTT) assay and Transwell assay to measure the proliferation rate and metastatic potential of glioma cells. Meanwhile, the self-renewal ability of GSCs was detected by tumor sphere formation assay. The results revealed that down-regulation of miR-92a-3p suppressed the glioma cell malignancy in vitro. Moreover, knockdown of miR-92a-3p led to a reduction of tumorgenesis in vivo. Interestingly, we also observed that up-regulation of miR-92a-3p could inhibit the stemness of GSCs. Subsequent mechanistic investigation indicated that cadherin 1 (CDH1)/ß-catenin signaling and Notch-1/Akt signaling were the downstream pathways of miR-92a-3p in glioma cells and GSCs, respectively. These results suggest that miR-92a-3p plays different roles in glioma cells and GSCs through regulating different signaling pathways.


Asunto(s)
Cadherinas/genética , Glioma/genética , MicroARNs/genética , Receptor Notch1/genética , Animales , Antígenos CD , Cadherinas/biosíntesis , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioma/patología , Humanos , Ratones , MicroARNs/biosíntesis , Células Madre Neoplásicas/patología , Proteína Oncogénica v-akt/genética , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
15.
Oncotarget ; 7(42): 67716-67731, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27626494

RESUMEN

Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by ß amyloid (Aß) deposition and neurofibril tangles. It has been reported that a bioflavonoid, quercetin, could ameliorate AD phenotypes in C. elegans and mice. However, the mechanism underlying the ameliorative effect of quercetin is not fully understood yet. Drosophila models could recapitulate AD-like phenotypes, such as shortened lifespan, impaired locomotive ability as well as defects in learning and memory. So in this study, we investigated the effects of quercetin on AD in Drosophila model and explored the underlying mechanisms. We found quercetin could effectively intervene in AD pathogenesis in vivo. Mechanism study showed quercetin could restore the expression of genes perturbed by Aß accumulation, such as those involved in cell cycle and DNA replication. Cyclin B, an important cell cycle protein, was chosen to test whether it participated in the AD ameliorative effects of quercetin. We found that cyclin B RNAi in the brain could alleviate AD phenotypes. Taken together, the current study suggested that the neuroprotective effects of quercetin were mediated at least partially by targeting cell cycle-related proteins.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Proteínas de Ciclo Celular/metabolismo , Modelos Animales de Enfermedad , Drosophila melanogaster/metabolismo , Quercetina/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Animales , Animales Modificados Genéticamente , Antioxidantes/farmacología , Proteínas de Ciclo Celular/genética , Ciclina B/genética , Ciclina B/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Humanos , Fármacos Neuroprotectores/farmacología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
16.
J Physiol Biochem ; 72(4): 733-744, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27520092

RESUMEN

Glioblastoma multiforme (GBM), the most common and lethal primary brain tumor in adults characterized by high proliferative ability and mortality rate, contains a small subpopulation of cancer stem-like cells (CSCs), which is responsible for GBM progression and therapeutic resistance. Numerous microRNAs are strongly implicated in the malignancy of glioma. However, their specific functions and roles have yet to be fully demonstrated. In the present study, we revealed that the upregulation of Let-7b, a member of the Let-7 microRNA family, inhibited proliferation, migration, and invasion in glioma cell lines. Using bioinformatics, expression analysis, and luciferase assay, E2F2 was confirmed as a candidate target of Let-7b. Moreover, we also observed that elevated levels of Let-7b resulted in a reduction of tumor sphere growth and stemness of glioma stem-like cells. Furthermore, we found that knockdown of E2F2 expression could reduce the proliferation of glioma and GSCs, while overexpression of E2F2 partially abrogated the inhibitory effect of Let-7b on the proliferation of glioma and GSCs. In conclusion, we suggest that Let-7b could be developed into a promising anticancer target in glioma.


Asunto(s)
Factor de Transcripción E2F2/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Células Madre Neoplásicas/metabolismo , Neuroglía/metabolismo , Esferoides Celulares/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Biología Computacional , Factor de Transcripción E2F2/metabolismo , Genes Reporteros , Humanos , Luciferasas/genética , Luciferasas/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/patología , Neuroglía/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Esferoides Celulares/patología
17.
J Physiol Biochem ; 72(2): 201-11, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26893028

RESUMEN

Glioblastoma (GBM) is a highly invasive malignant primary brain tumor with neoplastic growth. Despite the progresses made in surgery, chemotherapy, and radiation in recent decade, the prognosis of patients with gliomas remains poor and the average survival time of patients suffering from glioblastoma is still short. As a potential therapy strategy, microRNAs have been considered as new targets for possible cancer treatment. In this study, we found that the miR-92b inhibitors (miR-92b-I) could inhibit the proliferation, migration, invasion, and promote the apoptosis of glioma cells. As a predicted target of miR-92b, phosphatase and tensin homolog (PTEN), also elevated at both mRNA and protein levels. Moreover, the Akt phosphorylation was consistently inhibited. The rescue experiment with miR-92b and PTEN double knockdown resulted in partial reversion of miR-92b-I-induced phenotypes. Taken together, our findings indicated that miR-92b-I could restrain the proliferation, invasion, migration, and stimulate apoptosis of glioma cells by targeting PTEN/Akt signaling pathway. Further investigations will focus on antitumor effect of miR­92b-I in glioma treatment.


Asunto(s)
Antagomirs/farmacología , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Glioma/tratamiento farmacológico , MicroARNs/antagonistas & inhibidores , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal/efectos de los fármacos , Regiones no Traducidas 3'/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Biología Computacional , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Glioma/metabolismo , Glioma/patología , Humanos , MicroARNs/metabolismo , Mutación , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , ARN Neoplásico/antagonistas & inhibidores , ARN Neoplásico/metabolismo
18.
Exp Cell Res ; 344(2): 183-93, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-26844630

RESUMEN

Despite great progress for two decades in microRNAs (miRNAs), the direct regulation of host gene by intragenic (mostly intronic) miRNA is conceptually plausible but evidence-limited. Here, we report that intronic miR-932 could target its host gene via binding with coding sequence (CDS) region rather than regular 3'UTR. The conserved miR-932 is embedded in the fourth intron of Drosophila neuroligin2 (dnlg2), which encodes a synaptic cell adhesion molecule, DNlg2. In silico analysis predicted two putative miR-932 target sites locate in the CDS region of dnlg2 instead of regular 3'-UTR miRNA binding sites. Employing luciferase reporter assay, we further proved that the miR-932 regulates expression of its host gene dnlg2 via the binding CDS region of dnlg2. Consistently, we observed miR-932 downregulated expression of dnlg2 in S2 cell, and the repression of dnlg2 by miR-932 at both protein and RNA level. Furthermore, we found CDS-located site1 is dominant for regulating expression of host dnlg2 by miR-932. In addition to providing thorough examination of one intronic miRNA targeting the CDS region of its host gene, our genome-wide analysis indicated that nearly half of fruitfly and human intronic miRNAs may target their own host gene at coding region. This study would be valuable in elucidating the regulation of intronic miRNA on host gene, and provide new information about the biological context of their genomic arrangements and functions.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Genes de Insecto , Intrones/genética , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/genética , Sistemas de Lectura Abierta/genética , Animales , Secuencia de Bases , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular , Proteínas de Drosophila/genética , Regulación de la Expresión Génica , Genoma Humano , Cabeza , Humanos , Luciferasas/metabolismo , MicroARNs/genética , Proteínas del Tejido Nervioso/metabolismo
19.
PLoS One ; 10(9): e0137432, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26361355

RESUMEN

Parkinson's disease (PD) is a prevalent neurodegenerative disorder with pathological features including death of dopaminergic neurons in the substantia nigra and intraneuronal accumulations of Lewy bodies. As the main component of Lewy bodies, α-synuclein is implicated in PD pathogenesis by aggregation into insoluble filaments. However, the detailed mechanisms underlying α-synuclein induced neurotoxicity in PD are still elusive. MicroRNAs are ~20nt small RNA molecules that fine-tune gene expression at posttranscriptional level. A plethora of miRNAs have been found to be dysregulated in the brain and blood cells of PD patients. Nevertheless, the detailed mechanisms and their in vivo functions in PD still need further investigation. By using Drosophila PD model expressing α-synuclein A30P, we examined brain miRNA expression with high-throughput small RNA sequencing technology. We found that five miRNAs (dme-miR-133-3p, dme-miR-137-3p, dme-miR-13b-3p, dme-miR-932-5p, dme-miR-1008-5p) were upregulated in PD flies. Among them, miR-13b, miR-133, miR-137 are brain enriched and highly conserved from Drosophila to humans. KEGG pathway analysis using DIANA miR-Path demonstrated that neuroactive-ligand receptor interaction pathway was most likely affected by these miRNAs. Interestingly, miR-137 was predicted to regulate most of the identified targets in this pathway, including dopamine receptor (DopR, D2R), γ-aminobutyric acid (GABA) receptor (GABA-B-R1, GABA-B-R3) and N-methyl-D-aspartate (NMDA) receptor (Nmdar2). The validation experiments showed that the expression of miR-137 and its targets was negatively correlated in PD flies. Further experiments using luciferase reporter assay confirmed that miR-137 could act on specific sites in 3' UTR region of D2R, Nmdar2 and GABA-B-R3, which downregulated significantly in PD flies. Collectively, our findings indicate that α-synuclein could induce the dysregulation of miRNAs, which target neuroactive ligand-receptor interaction pathway in vivo. We believe it will help us further understand the contribution of miRNAs to α-synuclein neurotoxicity and provide new insights into the pathogenesis driving PD.


Asunto(s)
MicroARNs/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Transducción de Señal , alfa-Sinucleína/metabolismo , Regiones no Traducidas 3' , Animales , Biología Computacional , Modelos Animales de Enfermedad , Drosophila , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Locomoción/genética , Masculino , Anotación de Secuencia Molecular , Interferencia de ARN , ARN Mensajero/genética , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...