Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int Immunopharmacol ; 136: 112410, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38843641

RESUMEN

Impaired wound healing in diabetes results from a complex interplay of factors that disrupt epithelialization and wound closure. MG53, a tripartite motif (TRIM) family protein, plays a key role in repairing cell membrane damage and facilitating tissue regeneration. In this study, bone marrow-derived mesenchymal stem cells (BMSCs) were transduced with lentiviral vectors overexpressing MG53 to investigate their efficacy in diabetic wound healing. Using a db/db mouse wound model, we observed that BMSCs-MG53 significantly enhanced diabetic wound healing. This improvement was associated with marked increase in re-epithelialization and vascularization. BMSCs-MG53 promoted recruitment and survival of BMSCs, as evidenced by an increase in MG53/Ki67-positive BMSCs and their improved response to scratch wounding. The combination therapy also promoted angiogenesis in diabetic wound tissues by upregulating the expression of angiogenic growth factors. MG53 overexpression accelerated the differentiation of BMSCs into endothelial cells, manifested as the formation of mature vascular network structure and a remarkable increase in DiI-Ac-LDL uptake. Our mechanistic investigation revealed that MG53 binds to caveolin-3 (CAV3) and subsequently increases phosphorylation of eNOS, thereby activating eNOS/NO signaling. Notably, CAV3 knockdown reversed the promoting effects of MG53 on BMSCs endothelial differentiation. Overall, our findings support the notion that MG53 binds to CAV3, activates eNOS/NO signaling pathway, and accelerates the therapeutic effect of BMSCs in the context of diabetic wound healing. These insights hold promise for the development of innovative strategies for treating diabetic-related impairments in wound healing.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Óxido Nítrico Sintasa de Tipo III , Óxido Nítrico , Transducción de Señal , Cicatrización de Heridas , Animales , Células Madre Mesenquimatosas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratones , Óxido Nítrico/metabolismo , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Células Cultivadas , Humanos , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/metabolismo , Diferenciación Celular , Proteínas de la Membrana
2.
Biophys Rev ; 16(1): 13-28, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38495443

RESUMEN

With the rapid advance of single-cell sequencing technology, cell heterogeneity in various biological processes was dissected at different omics levels. However, single-cell mono-omics results in fragmentation of information and could not provide complete cell states. In the past several years, a variety of single-cell multimodal omics technologies have been developed to jointly profile multiple molecular modalities, including genome, transcriptome, epigenome, and proteome, from the same single cell. With the availability of single-cell multimodal omics data, we can simultaneously investigate the effects of genomic mutation or epigenetic modification on transcription and translation, and reveal the potential mechanisms underlying disease pathogenesis. Driven by the massive single-cell omics data, the integration method of single-cell multi-omics data has rapidly developed. Integration of the massive multi-omics single-cell data in public databases in the future will make it possible to construct a cell atlas of multi-omics, enabling us to comprehensively understand cell state and gene regulation at single-cell resolution. In this review, we summarized the experimental methods for single-cell multimodal omics data and computational methods for multi-omics data integration. We also discussed the future development of this field.

3.
Patterns (N Y) ; 5(3): 100915, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38487801

RESUMEN

Understanding tissue architecture and niche-specific microenvironments in spatially resolved transcriptomics (SRT) requires in situ annotation and labeling of cells. Effective spatial visualization of these data demands appropriate colorization of numerous cell types. However, current colorization frameworks often inadequately account for the spatial relationships between cell types. This results in perceptual ambiguity in neighboring cells of biological distinct types, particularly in complex environments such as brain or tumor. To address this, we introduce Spaco, a potent tool for spatially aware colorization. Spaco utilizes the Degree of Interlacement metric to construct a weighted graph that evaluates the spatial relationships among different cell types, refining color assignments. Furthermore, Spaco incorporates an adaptive palette selection approach to amplify chromatic distinctions. When benchmarked on four diverse datasets, Spaco outperforms existing solutions, capturing complex spatial relationships and boosting visual clarity. Spaco ensures broad accessibility by accommodating color vision deficiency and offering open-accessible code in both Python and R.

4.
Cell Physiol Biochem ; 41(5): 1947-1956, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28391274

RESUMEN

BACKGROUND/AIMS: Renal cell carcinoma (RCC) remains an intractable genitourinary malignancy. Resistance to chemotherapy or targeted therapies in RCC is presumably due to the complicated underlying molecular mechanisms and insufficient understanding. The aim of this research was to assess the expression and role of bromodomain-4 protein (BRD4) in RCC and evaluate the effects of BRD4 inhibitor JQ1 for RCC treatment. METHODS: BRD4 expressionlevels were assessed by qRT-PCR and western blot in RCC tissues and cells. The effects of BRD4 knockdown or JQ1 on RCC cells were assessed by MTT assay and flow cytometry. The effects of in vivo treatment were evaluated through xenograft experiments. RESULTS: BRD4 is significantly overexpressed in RCC, and is related to tumor stage and lymph node metastasis. Inhibition of BRD4 suppressed RCC cell proliferation, induced cell apoptosis in vitro and repressed tumor growth in vivo. Inhibition of BRD4 decreased BCL2 and C-MYC expression while increased BAX and cleaved caspase3 expression, and strikingly diminished the recruitment of BRD4 to BCL2 promoter. CONCLUSIONS: Our research reveals that BRD4 probably play a critical role in RCC progression, and is a new promising target for pharmacological treatment directed against this intractable disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Azepinas/farmacología , Carcinoma de Células Renales/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias Renales/metabolismo , Proteínas de Neoplasias , Proteínas Nucleares , Factores de Transcripción , Triazoles/farmacología , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo
5.
Cancer Lett ; 388: 281-291, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27998761

RESUMEN

Emerging evidences have indicated that long non-coding RNAs (LncRNAs) play vital roles in cancer development and progression. Previous studies have suggested that overexpression of SPRY4-IT1 predicates poor prognosis and promotes tumor progress in several cancers. However, the underlying mechanism of SPRY4-IT1 in bladder cancer remains unknown. In this study, we found that SPRY4-IT1 knockdown induced inhibition of cell proliferation, cell migration and invasion ability, and caused promotion of apoptosis in bladder cancer both in vitro and in vivo. Mechanistically, knockdown of SPRY4-IT1 increased the expression of miR-101-3p and subsequently inhibited the expression of EZH2 at posttranscriptional level. Importantly, SPRY4-IT1 could directly interact with miR-101-3p and down-regulation of miR-101-3p efficiently reversed the suppression of EZH2 induced by SPRY4-IT1 shRNA. Thus, SPRY4-IT1 positively regulated the expression of EZH2 through sponging miR-101-3p, and played an oncogenic role in bladder cancer progression. Together, our study elucidates the role of LncRNA SPRY4-IT1 as a miRNA sponge in bladder cancer, and sheds new light on LncRNA-directed diagnostics and therapeutics in bladder cancer.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , ARN Largo no Codificante/genética , Neoplasias de la Vejiga Urinaria/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Ratones Desnudos , MicroARNs , Metástasis de la Neoplasia , Transfección , Regulación hacia Arriba
6.
Oncotarget ; 8(1): 664-678, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27893422

RESUMEN

Pituitary tumor-transforming gene 1 (PTTG1) is identified as an oncogene, and overexpresses in many tumors. However, the role of PTTG1 in bladder cancer (BC) hasn't yet been characterized well. In this study, we showed the expression of PTTG1 mRNA and protein were both significantly increased in BC tissues and cells. The PTTG1 protein levels were positive correlated with increased tumor size, tumor-node-metastasis (TNM) stage, lymphatic invasion and distant metastasis of BC. PTTG1 knockdown dramatically suppressed the migration, invasion, metastasis and growth, and induced senescence and cell-cycle arrest at G0/G1 phase of BC cells. We further identified PTTG1 was the direct target of miR-146a-3p through using target prediction algorithms and luciferase reporter assay. miR-146a-3p was low expressed and negatively correlated with PTTG1 levels in BC tissues and cells. miR-146a-3p overexpression inhibited migration, invasion, metastasis and growth, and induced senescence of BC cells. Rescue experiment suggested ectopic expression of miR-146a-3p and PTTG1 suppressed migration, invasion and induced cell cycle arrest and senescence of BC cells compared to PTTG1 overexpression, confirming miR-146a-3p inhibited BC progression by targeting PTTG1. In summary, our study found miR-146a-3p/PTTG1 axis regulated BC migration, invasion, metastasis and growth, and might be a targets for BC therapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Interferencia de ARN , Securina/genética , Neoplasias de la Vejiga Urinaria/genética , Regiones no Traducidas 3' , Animales , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Senescencia Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Ther ; 15(5): 1029-42, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26939702

RESUMEN

People who develop bladder cancer frequently succumb to the intractable disease. Current treatment strategies are limited presumably due to the underlying molecular complexity and insufficient comprehension. Therefore, exploration of new therapeutic targets in bladder cancer remains necessary. Here, we identify that bromodomain-4 protein (BRD4), an important epigenome reader of bromodomain and extraterminal domain (BET) family member, is a key upstream regulator of enhancer of zeste homologue 2 (EZH2), and represents a novel therapeutic target in bladder cancer. We found that BRD4 was significantly overexpressed in bladder cancer cells and tissues. Inhibition of BRD4 decreased bladder cancer cell proliferation concomitantly with the accumulation of cell apoptosis in vitro and suppressed tumor growth in vivo We further found that suppression of BRD4 decreased the mRNA and protein levels of EZH2, which was reversed by ectopic expression of C-MYC In particular, individual silencing of BRD4 using shRNA or the BET inhibitor JQ1 strikingly diminished the recruitment of C-MYC to EZH2 promoter in bladder cancer. Briefly, our research reveals that BRD4 positively regulates EZH2 transcription through upregulation of C-MYC, and is a novel promising target for pharmacologic treatment in transcriptional program intervention against this intractable disease. Mol Cancer Ther; 15(5); 1029-42. ©2016 AACR.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología
8.
Biochem Biophys Res Commun ; 469(3): 679-85, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26707881

RESUMEN

Thyroid cancer is a common malignancy of the endocrine system. Although radioiodine (131)I treatment on differentiated thyroid cancer is widely used, many patients still fail to benefit from (131)I therapy. Therefore, exploration of novel targeted therapies to suppress tumor growth and improve radioiodine uptake remains necessary. Bromodomain-containing protein 4 (BRD4) is an important member of the bromodomain and extra terminal domain family that influences transcription of downstream genes by binding to acetylated histones. In the present study, we found that BRD4 was up-regulated in thyroid cancer tissues and cell lines. Inhibition of BRD4 in thyroid cancer cells by JQ1 resulted in cell cycle arrest at G0/G1 phase and enhanced (131)I uptake in vitro and suppressed tumor growth in vivo. Moreover, JQ1 treatment suppressed C-MYC but enhanced NIS expression. We further demonstrated that BRD4 was enriched in the promoter region of C-MYC, which could be markedly blocked by JQ1 treatment. In conclusion, our findings revealed that the aberrant expression of BRD4 in thyroid cancer is possibly involved in tumor progression, and JQ1 is potentially an effective chemotherapeutic agent against human thyroid cancer.


Asunto(s)
Radioisótopos de Yodo/farmacocinética , Proteínas Nucleares/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Radioisótopos de Yodo/uso terapéutico , Tasa de Depuración Metabólica , Radiofármacos/farmacocinética , Radiofármacos/uso terapéutico , Neoplasias de la Tiroides/radioterapia , Regulación hacia Arriba
9.
Cell Physiol Biochem ; 37(6): 2209-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26610246

RESUMEN

BACKGROUND/AIMS: Long non-coding RNAs (lncRNAs) play important roles in diverse biological processes, such as cell growth, apoptosis and migration. Although downregulation of lncRNA maternally expressed gene 3 (MEG3) has been identified in several cancers, little is known about its role in prostate cancer progression. The aim of this study was to detect MEG3 expression in clinical prostate cancer tissues, investigate its biological functions in the development of prostate cancer and the underlying mechanism. METHODS: MEG3 expression levels were detected by qRT-PCR in both tumor tissues and adjacent non-tumor tissues from 21 prostate cancer patients. The effects of MEG3 on PC3 and DU145 cells were assessed by MTT assay, colony formation assay, western blot and flow cytometry. Transfected PC3 cells were transplanted into nude mice, and the tumor growth curves were determined. RESULTS: MEG3 decreased significantly in prostate cancer tissues relative to adjacent normal tissues. MEG3 inhibited intrinsic cell survival pathway in vitro and in vivo by reducing the protein expression of Bcl-2, enhancing Bax and activating caspase 3. We further demonstrated that MEG3 inhibited the expression of cell cycle regulatory protein Cyclin D1 and induced cell cycle arrest in G0/G1 phase. CONCLUSIONS: Our study presents an important role of MEG3 in the molecular etiology of prostate cancer and implicates the potential application of MEG3 in prostate cancer therapy.


Asunto(s)
Apoptosis/genética , Proliferación Celular/genética , Neoplasias de la Próstata/patología , ARN Largo no Codificante/fisiología , Animales , Caspasa 3/metabolismo , Ciclina D1/metabolismo , Activación Enzimática , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
10.
Oncotarget ; 6(6): 4066-79, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25714014

RESUMEN

Inactivation of human SET domain containing protein 2 (SETD2) is a common event in clear cell renal cell carcinoma (ccRCC). However, the mechanism underlying loss of SETD2 function, particularly the post-transcriptional regulatory mechanism, still remains unclear. In the present study, we found that SETD2 was downregulated and inversely correlated with high expression of miR-106b-5p in ccRCC tissues and cell lines. Over-expression of miR-106b-5p resulted in the decreased mRNA and protein levels of SETD2 in ccRCC cells. In an SETD2 3'-UTR luciferase reporter system, miR-106b-5p downregulated the luciferase activity, and the effects were abolished by mutating the predicted miR-106b-5p binding site. Moreover, attenuation of miR-106b-5p induced cell cycle arrest at G0/G1 phase, suppressed cell proliferation, enhanced processing of caspase-3, and promoted cell apoptosis in ccRCC cells, whereas these effects were reversed upon knockdown of SETD2. In addition, transfection of miR-106b-5p antagomir resulted in the increased binding of H3K36me3 to the promoter of p53 and enhanced its activity, as well as upregulated the mRNA and protein levels of p53, and the effects were also abolished by cotransfection with si-SETD2. Collectively, our findings extend the knowledge about the regulation of SETD2 at the posttranscriptional level by miRNA and regulatory mechanism downstream of SETD2 in ccRCC.


Asunto(s)
Carcinoma de Células Renales/genética , Genes Supresores de Tumor , N-Metiltransferasa de Histona-Lisina/genética , Neoplasias Renales/genética , MicroARNs/genética , Proteína p53 Supresora de Tumor/metabolismo , Carcinoma de Células Renales/metabolismo , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Neoplasias Renales/metabolismo , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección
11.
Urology ; 84(4): 983.e1-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25260464

RESUMEN

OBJECTIVE: To investigate the feasibility of erectile function restoration by genital branch of genitofemoral nerve (GN) to cavernous nerve (CN) transfer in rats. MATERIALS AND METHODS: Thirty adult (3 months) male Sprague-Dawley rats were divided into 3 groups (n = 10 per group). Rats in sham group underwent sham operation, rats in nerve resection (NR) group underwent bilateral GN and CN resection to make a 2.5-mm gap, and rats in nerve transfer (NT) group underwent nerve anastomosis bilaterally between proximal stump of GN and distal stump of CN after nerve resection. RESULTS: Three months postoperatively, mating test observed 70% rats with intromission behaviors in NT group but only 10% rats in NR group. Electrostimulating the GN of NT group rats resulted in a significant increase in intracavernous pressure, and the ratio of intracavernous pressure increase to mean arterial pressure in NT group was significantly higher than that in NR group. Seven days after Fluoro-Gold injection into the penile crus, Fluoro-Gold-labeled neurons were found in ventral horn of L1 and L2 in NT group, indicating that a new erectile efferent pathway might be established. Axon counting and ultrastructure observation confirmed axonal regeneration in NT group. Furthermore, NT group had a higher expression of nitric oxide synthase in the dorsal penile nerve than that in NR group. CONCLUSION: The results have demonstrated that nerve regeneration can be obtained, and erectile function may be restored after GN to CN nerve transfer in bilateral CN resection rats, which provides an innovative and promising treatment for neurogenic erectile dysfunction.


Asunto(s)
Disfunción Eréctil/cirugía , Transferencia de Nervios , Pene/inervación , Pene/cirugía , Animales , Estudios de Factibilidad , Masculino , Ratas , Ratas Sprague-Dawley , Procedimientos Quirúrgicos Urológicos Masculinos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA