Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 28
Filtrer
1.
Front Cell Dev Biol ; 12: 1423553, 2024.
Article de Anglais | MEDLINE | ID: mdl-39045460

RÉSUMÉ

Alternative splicing is a complex gene regulatory process that distinguishes itself from canonical splicing by rearranging the introns and exons of an immature pre-mRNA transcript. This process plays a vital role in enhancing transcriptomic and proteomic diversity from the genome. Alternative splicing has emerged as a pivotal mechanism governing complex biological processes during both heart development and the development of cardiovascular diseases. Multiple alternative splicing factors are involved in a synergistic or antagonistic manner in the regulation of important genes in relevant physiological processes. Notably, circular RNAs have only recently garnered attention for their tissue-specific expression patterns and regulatory functions. This resurgence of interest has prompted a reevaluation of the topic. Here, we provide an overview of our current understanding of alternative splicing mechanisms and the regulatory roles of alternative splicing factors in cardiovascular development and pathological process of different cardiovascular diseases, including cardiomyopathy, myocardial infarction, heart failure and atherosclerosis.

2.
Stem Cell Res ; 78: 103467, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38861774

RÉSUMÉ

Dilated cardiomyopathy (DCM) is one of the main causes of sudden cardiac death and heart failure and is the leading indication for cardiac transplantation worldwide. Mutations in dozens of cardiac genes have been connected to the development of DCM including the Troponin T2 gene (TNNT2). Here, we generated a human induced pluripotent stem cells (hiPSCs) from a DCM patient with a familial history that carries a missense mutation in TNNT2. The hiPSCs show typical morphology of pluripotent stem cells, expression of pluripotency markers, normal karyotype, and in vitro capacity to differentiate into all three germ layers.


Sujet(s)
Cardiomyopathie dilatée , Cellules souches pluripotentes induites , Troponine T , Humains , Cardiomyopathie dilatée/anatomopathologie , Cellules souches pluripotentes induites/métabolisme , Troponine T/métabolisme , Troponine T/génétique , Différenciation cellulaire , Lignée cellulaire , Mâle , Caryotype
3.
Mol Ther Nucleic Acids ; 32: 582-593, 2023 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-37200858

RÉSUMÉ

Human pluripotent stem cell-derived endothelial cells (hPSC-ECs) represent a promising source of human ECs urgently needed for the study of cardiovascular disease mechanisms, cell therapy, and drug screening. This study aims to explore the function and regulatory mechanism of the miR-148/152 family consisting of miR-148a, miR-148b, and miR-152 in hPSC-ECs, so as to provide new targets for improving EC function during the above applications. In comparison with the wild-type (WT) group, miR-148/152 family knockout (TKO) significantly reduced the endothelial differentiation efficiency of human embryonic stem cells (hESCs), and impaired the proliferation, migration, and capillary-like tube formatting abilities of their derived ECs (hESC-ECs). Overexpression of miR-152 partially restored the angiogenic capacity of TKO hESC-ECs. Furthermore, the mesenchyme homeobox 2 (MEOX2) was validated as the direct target of miR-148/152 family. MEOX2 knockdown resulted in partial restoration of the angiogenesis ability of TKO hESC-ECs. The Matrigel plug assay further revealed that the in vivo angiogenic capacity of hESC-ECs was impaired by miR-148/152 family knockout, and increased by miR-152 overexpression. Thus, the miR-148/152 family is crucial for maintaining the angiogenesis ability of hPSC-ECs, and might be used as a target to enhance the functional benefit of EC therapy and promote endogenous revascularization.

4.
Circulation ; 147(22): 1684-1704, 2023 05 30.
Article de Anglais | MEDLINE | ID: mdl-37066795

RÉSUMÉ

BACKGROUND: A large portion of idiopathic and familial dilated cardiomyopathy (DCM) cases have no obvious causal genetic variant. Although altered response to metabolic stress has been implicated, the molecular mechanisms underlying the pathogenesis of DCM remain elusive. The JMJD family proteins, initially identified as histone deacetylases, have been shown to be involved in many cardiovascular diseases. Despite their increasingly diverse functions, whether JMJD family members play a role in DCM remains unclear. METHODS: We examined Jmjd4 expression in patients with DCM, and conditionally deleted and overexpressed Jmjd4 in cardiomyocytes in vivo to investigate its role in DCM. RNA sequencing, metabolites profiling, and mass spectrometry were used to dissect the molecular mechanism of Jmjd4-regulating cardiac metabolism and hypertrophy. RESULTS: We found that expression of Jmjd4 is significantly decreased in hearts of patients with DCM. Induced cardiomyocyte-specific deletion of Jmjd4 led to spontaneous DCM with severely impaired mitochondrial respiration. Pkm2, the less active pyruvate kinase compared with Pkm1, which is normally absent in healthy adult cardiomyocytes but elevated in cardiomyopathy, was found to be drastically accumulated in hearts with Jmjd4 deleted. Jmjd4 was found mechanistically to interact with Hsp70 to mediate degradation of Pkm2 through chaperone-mediated autophagy, which is dependent on hydroxylation of K66 of Pkm2 by Jmjd4. By enhancing the enzymatic activity of the abundant but less active Pkm2, TEPP-46, a Pkm2 agonist, showed a significant therapeutic effect on DCM induced by Jmjd4 deficiency, and heart failure induced by pressure overload, as well. CONCLUSIONS: Our results identified a novel role of Jmjd4 in maintaining metabolic homeostasis in adult cardiomyocytes by degrading Pkm2 and suggest that Jmjd4 and Pkm2 may be therapeutically targeted to treat DCM, and other cardiac diseases with metabolic dysfunction, as well.


Sujet(s)
Cardiomyopathie dilatée , Défaillance cardiaque , Humains , Myocytes cardiaques/métabolisme , Cardiomyopathie dilatée/anatomopathologie , Défaillance cardiaque/anatomopathologie
5.
Acta Pharmacol Sin ; 43(1): 240-250, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-33686244

RÉSUMÉ

Cardiovascular safety assessment is vital for drug development, yet human cardiovascular cell models are lacking. In vitro mass-generated human pluripotent stem cell (hPSC)-derived cardiovascular cells are a suitable cell model for preclinical cardiovascular safety evaluations. In this study, we established a preclinical toxicology model using same-origin hPSC-differentiated cardiomyocytes (hPSC-CMs) and endothelial cells (hPSC-ECs). For validation of this cell model, alirocumab, a human antibody against proprotein convertase subtilisin kexin type 9 (PCSK9), was selected as an emerging safe lipid-lowering drug; atorvastatin, a common statin (the most effective type of lipid-lowering drug), was used as a drug with reported side effects at high concentrations, while doxorubicin was chosen as a positive cardiotoxic drug. The cytotoxicity of these drugs was assessed using CCK8, ATP, and lactate dehydrogenase release assays at 24, 48, and 72 h. The influences of these drugs on cardiomyocyte electrophysiology were detected using the patch-clamp technique, while their effects on endothelial function were determined by tube formation and Dil-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake assays. We showed that alirocumab did not affect the cell viability or cardiomyocyte electrophysiology in agreement with the clinical results. Atorvastatin (5-50 µM) dose-dependently decreased cardiovascular cell viability over time, and at a high concentration (50 µM, ~100 times the normal peak serum concentration in clinic), it affected the action potentials of hPSC-CMs and damaged tube formation and Dil-Ac-LDL uptake of hPSC-ECs. The results demonstrate that the established same-origin hPSC-derived cardiovascular cell model can be used to evaluate lipid-lowering drug safety in cardiovascular cells and allow highly accurate preclinical assessment of potential drugs.


Sujet(s)
Anticholestérolémiants/pharmacologie , Atorvastatine/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Anticholestérolémiants/composition chimique , Atorvastatine/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Humains , Modèles moléculaires , Structure moléculaire , Relation structure-activité
6.
Small ; 17(40): e2102884, 2021 Oct.
Article de Anglais | MEDLINE | ID: mdl-34480523

RÉSUMÉ

Chiral hybrid organic-inorganic perovskites (HOIPs) have been well developed for circularly polarized light (CPL) detection, while new members that target at solar-blind ultraviolet (UV) region remain completely unexplored. Here, an effective design strategy to demonstrate circular polarization-sensitive solar-blind UV photodetection by growing wide-bandgap chiral HOIP [(R)-MPA]2 PbCl4 ((R)-MPA = methylphenethylammonium) single crystals onto silicon wafers, with well-defined heterostructures, is reported. The solid mechanical and electrical connection between the chiral HOIP and silicon wafer results in strong built-in electric field at heterojunction, providing a desirable driving force for separating/transporting carriers generated under CPL excitation at 266 nm. Unexpectedly, during such a transport process, not only the chirality of HOIP crystal is transferred to the heterostructure, but also the circular polarization sensitivity is significantly amplified. Consequently, anisotropy factor of the resultant detectors can reach up to 0.4 at zero bias, which is much higher than that of the pristine single-phase chiral HOIP (≈0.1), reaching the highest among the reported CPL-UV photodetectors. As far as we know, the integration of chiral HOIP crystals with silicon technology is unprecedent, which paves a way for designing boosted-performance CPL detectors in solar-blind UV region as well as for other advanced optoelectronic devices.

7.
Stem Cell Res ; 52: 102241, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33611045

RÉSUMÉ

Ephrin B2 (EFNB2) is the first identified and most widely used marker for arterial endothelial cells (AECs). We generated a heterozygous EFNB2-2A-mCherry reporter H1 cell line, H1-EFNB2-2A-mCherry+/- (WAe001-A-57), by CRISPR/Cas9-mediated insertion of 2A-mCherry cassette into the EFNB2 gene locus, immediately before the translation stop codon. The H1-EFNB2-2A-mCherry reporter cells were pluripotent and could differentiate into all three germ layer lineages. Simultaneous expression of mCherry was observed when expression of EFNB2 was increased during endothelial cell differentiation. Thus, the generated reporter cells enable live identification of EFNB2-positive AECs, and screening of small molecule compound and target genes that promote AEC differentiation.


Sujet(s)
Éphrine B2 , Cellules souches embryonnaires humaines , Systèmes CRISPR-Cas/génétique , Lignée cellulaire , Cellules endothéliales , Recombinaison homologue , Humains
8.
J Cell Mol Med ; 24(23): 13775-13788, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33084169

RÉSUMÉ

Myocardial infarction (MI) results in cardiomyocyte death and ultimately leads to heart failure. Pyroptosis is a type of the inflammatory programmed cell death that has been found in various diseased tissues. However, the role of pyroptosis in MI heart remains unknown. Here, we showed that CXADR-like membrane protein (CLMP) was involved in pyroptosis in the mouse MI heart. Our data showed that CLMP was strongly expressed in fibroblasts of the infarcted mouse hearts. The Clmp+/- mice showed more serious myocardial fibrosis and ventricular dysfunction post-MI than wild-type (Clmp+/+ ) mice, indicating a protective effect of the fibroblast-expressed CLMP against MI-induced heart damage. Transcriptome analyses by RNA sequencing indicated that Il-1ß mRNA was significantly increased in the MI heart of Clmp+/- mouse, which indicated a more serious inflammatory response. Meanwhile, cleaved caspase-1 and Gasdermin D were significantly increased in the Clmp+/- MI heart, which demonstrated enhanced pyroptosis in the Clmp knockdown heart. Further analysis revealed that the pyroptosis mainly occurred in cardiac fibroblasts (CFs). Compared to wild-type fibroblasts, Clmp+/- CFs showed more serious pyroptosis and inflammatory after LPS plus nigericin treatment. Collectively, our results indicate that CLMP participates in the pyroptotic and inflammatory response of CFs in MI heart. We have provided a novel pyroptotic insight into the ischaemic heart, which might hold substantial potential for the treatment of MI.


Sujet(s)
Protéine membranaire apparentée au récepteur des coxsackievirus et adénovirus/génétique , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Pyroptose/génétique , Animaux , Marqueurs biologiques , Protéine membranaire apparentée au récepteur des coxsackievirus et adénovirus/métabolisme , Analyse de mutations d'ADN , Modèles animaux de maladie humaine , Échocardiographie , Fibroblastes/métabolisme , Expression des gènes , Génotype , Immunohistochimie , Médiateurs de l'inflammation/métabolisme , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Souris , Souris knockout , Souris transgéniques , Modèles biologiques , Mutation , Infarctus du myocarde/imagerie diagnostique , Infarctus du myocarde/étiologie , Phénotype
9.
Theranostics ; 10(21): 9686-9701, 2020.
Article de Anglais | MEDLINE | ID: mdl-32863954

RÉSUMÉ

Cardiomyocytes differentiated from human embryonic stem cells (hESCs) represent a promising cell source for heart repair, disease modeling and drug testing. However, improving the differentiation efficiency and maturation of hESC-derived cardiomyocytes (hESC-CMs) is still a major concern. Retinoic acid (RA) signaling plays multiple roles in heart development. However, the effects of RA on cardiomyocyte differentiation efficiency and maturation are still unknown. Methods: RA was added at different time intervals to identify the best treatment windows for cardiomyocyte differentiation and maturation. The efficiency of cardiomyocyte differentiation was detected by quantitative real-time PCR and flow cytometry. Cardiomyocytes maturation was detected by immunofluorescence staining, metabolic assays and patch clamp to verify structural, metabolic and electrophysiological maturation, respectively. RNA sequencing was used for splicing analysis. Results: We found that RA treatment at the lateral mesoderm stage (days 2-4) significantly improved cardiomyocyte differentiation, as evidenced by the upregulation of TNNT2, NKX2.5 and MYH6 on day 10 of differentiation. In addition, flow cytometry showed that the proportion of differentiated cardiomyocytes in the RA-treated group was significantly higher than that in control group. RA treatment on days 15-20 increased cardiomyocyte area, sarcomere length, multinucleation and mitochondrial copy number. RNA sequencing revealed RA promoted RNA isoform switch to the maturation-related form. Meanwhile, RA promoted electrophysiological maturation and calcium handling of hESC-CMs. Importantly, RA-treated cardiomyocytes showed decreased glycolysis and enhanced mitochondrial oxidative phosphorylation, with the increased utilization of fatty acid and exogenous pyruvate but not glutamine. Conclusion: Our data indicated that RA treatment at an early time window (days 2-4) promotes the efficiency of cardiomyocyte differentiation and that RA treatment post beating (days 15-20) promotes cardiomyocyte maturation. The biphasic effects of RA provide new insights for improving cardiomyocyte differentiation and quality.


Sujet(s)
Cellules souches embryonnaires humaines/effets des médicaments et des substances chimiques , Cellules souches embryonnaires humaines/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Trétinoïne/pharmacologie , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Acides gras/métabolisme , Humains , Souris , Souris de lignée ICR , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Acide pyruvique/métabolisme , Analyse de séquence d'ARN/méthodes , Transduction du signal/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques
10.
Opt Lett ; 45(8): 2291-2294, 2020 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-32287216

RÉSUMÉ

We report on the development and application of a novel, to the best of our knowledge, all-solid-state tunable narrow-linewidth 226 nm UV laser system. The laser system consists of three parts: a tunable single-frequency Ti:sapphire 787 nm laser, a single-frequency long-pulse-width flattop-shaped 532 nm laser, and a nonlinear frequency transformation system. The 532 nm laser is a sum-frequency mixed with the second harmonic of the 787 nm laser to produce the 226 nm laser. The maximum output pulse energy at 226 nm is 3 mJ. Nitric oxide planar laser-induced fluorescence velocimetry is demonstrated in the China Aerodynamics Research and Development Center's FD14 hypersonic shock tunnel using this 226 nm laser system. It is proven that this laser is convenient for high-resolution molecular tagging fluorescence spectroscopy.

11.
Theranostics ; 9(24): 7282-7297, 2019.
Article de Anglais | MEDLINE | ID: mdl-31695768

RÉSUMÉ

Rationale: As a hallmark of various heart diseases, cardiac fibrosis ultimately leads to end-stage heart failure. Anti-fibrosis is a potential therapeutic strategy for heart failure. Long noncoding RNAs (lncRNAs) have emerged as critical regulators of heart diseases that promise to serve as therapeutic targets. However, few lncRNAs have been directly implicated in cardiac fibrosis. Methods: The lncRNA expression profiles were assessed by microarray in cardiac fibrotic and remote ventricular tissues in mice with myocardial infarction. The mechanisms and functional significance of lncRNA-AK137033 in cardiac fibrosis were further investigated with both in vitro and in vivo models. Results: We identified 389 differentially expressed lncRNAs in cardiac fibrotic and remote ventricular tissues in mice with myocardial infarction. Among them, a lncRNA (AK137033) we named Safe was enriched in the nuclei of fibroblasts, and elevated in both myocardial infarction and TGF-ß-induced cardiac fibrosis. Knockdown of Safe prevented TGF-ß-induced fibroblast-myofibroblast transition, aberrant cell proliferation and secretion of extracellular matrix proteins in vitro, and mended the impaired cardiac function in mice suffering myocardial infarction. In vitro studies indicated that knockdown of Safe significantly inhibited the expression of its neighboring gene Sfrp2, and vice versa. The Sfrp2 overexpression obviously disturbed the regulatory effects of Safe shRNAs in both the in vitro cultured cardiac fibroblasts and myocardial infarction-induced fibrosis. Dual-Luciferase assay demonstrated that Safe and Sfrp2 mRNA stabilized each other via their complementary binding at the 3'-end. RNA electrophoretic mobility shift assay and RNA immunoprecipitation assay indicated that RNA binding protein HuR could bind to Safe-Sfrp2 RNA duplex, whereas the knockdown of HuR dramatically reduced the stabilization of Safe and Sfrp2 mRNAs, down-regulated their expression in cardiac fibroblasts, and thus inhibited TGF-ß-induced fibrosis. The Safe overexpression partially restrained the phenotype change of cardiac fibroblasts induced by Sfrp2 shRNAs, but not that induced by HuR shRNAs. Conclusions: Our study identifies Safe as a critical regulator of cardiac fibrosis, and demonstrates Safe-Sfrp2-HuR complex-mediated Sfrp2 mRNA stability is the underlying mechanism of Safe-regulated cardiac fibrosis. Fibroblast-enriched Safe could represent a novel target for anti-fibrotic therapy in heart diseases.


Sujet(s)
Protéine-1 similaire à ELAV/métabolisme , Protéines membranaires/métabolisme , Infarctus du myocarde/métabolisme , ARN long non codant/métabolisme , Animaux , Protéine-1 similaire à ELAV/génétique , Femelle , Fibroblastes/métabolisme , Fibrose/génétique , Fibrose/métabolisme , Humains , Protéines membranaires/génétique , Souris , Infarctus du myocarde/génétique , Infarctus du myocarde/anatomopathologie , Liaison aux protéines , Stabilité de l'ARN , ARN long non codant/génétique
12.
J Mol Cell Cardiol ; 134: 1-12, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31233755

RÉSUMÉ

MicroRNAs (miRNAs), as a class of naturally occurring RNAs, play important roles in cardiac physiology and pathology. There are many miRNAs that show multifarious expression patterns during cardiomyocyte genesis. Here, we focused on the MIR148A family, which is composed of MIR148A, MIR148B and MIR152, and shares the same seed sequences. The expression levels of all MIR148A family members progressively increased during the differentiation of human embryonic stem cells (hESCs) into cardiomyocytes. The deletion of MIR148A family (MIR148A-TKO) resulted in a decreased proportion of cardiomyocytes after cardiac induction, which was restored by the ectopic expression of MIR148A family members. Transcriptome analyses indicated that the MIR148A family could partially repress paraxial mesodermal differentiation from primitive streak cells. In turn, these miRNAs promoted lateral mesoderm and cardiomyocyte differentiation. Furthermore, the NOTCH ligand Delta-like 1 (DLL1) was validated as the target gene of MIR148A family, and knockdown of DLL1 could promote the cardiomyocyte differentiation of MIR148A-TKO hESCs. Thus, our results demonstrate MIR148A family could promote cardiomyocyte differentiation by inhibiting undesired paraxial mesoderm lineage commitment, which improves our understanding on cardiomyocyte differentiation from hESCs.


Sujet(s)
Protéines de liaison au calcium/génétique , Différenciation cellulaire/génétique , Cellules souches embryonnaires humaines/physiologie , Protéines membranaires/génétique , microARN/génétique , Myocytes cardiaques/physiologie , Récepteurs Notch/génétique , Transduction du signal/génétique , Protéines de liaison au calcium/métabolisme , Lignée cellulaire , Analyse de profil d'expression de gènes/méthodes , Cellules HEK293 , Humains , Mésoderme/physiologie , Transcriptome/génétique
13.
Gene Ther ; 25(8): 511-523, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30287867

RÉSUMÉ

Myocardial infarction (MI), with a major process of cardiomyocyte death, remains a leading cause of morbidity and mortality worldwide. To date, it has been shown that lncRNAs play important roles in cardiovascular pathology. However, the detailed studies on lncRNAs regulating cardiomyocyte death in myocardial infarction are still limited. In this study, we found a progressively upregulated expression of Meg3 in mouse injured heart after MI. Gain-of-function and loss-of-function approaches further revealed pro-apoptotic functions of Meg3 in rodent cardiomyocytes. Moreover, Meg3 was directly upregulated by p53 in hypoxic condition, and involved in apoptotic regulation via its direct binding with RNA-binding protein FUS (fused in sarcoma). Afterwards, adult MI mice that underwent intramyocardial injection with adeno-associated virus serotype 9 (AAV9) system carrying Meg3 shRNA showed a significant improvement of cardiac function. Moreover, we also found that MEG3 was increased in clinical heart failure samples, and had conservatively pro-apoptotic function in human cardiomyocytes that were differentiated from the human embryonic stem cells. Together, these results indicate that p53-induced Meg3-FUS complex plays an important role in cardiomyocyte apoptosis post-MI, and its specific knockdown in cardiomyocytes with AAV9 system represents a promising method to treat MI for preclinical investigation.


Sujet(s)
Apoptose , Infarctus du myocarde/génétique , Myocytes cardiaques/métabolisme , ARN long non codant/génétique , Thérapie par l'interférence par ARN/méthodes , Animaux , Hypoxie cellulaire , Cellules cultivées , Dependovirus/génétique , Femelle , Humains , Souris , Infarctus du myocarde/métabolisme , Infarctus du myocarde/thérapie , ARN long non codant/métabolisme , Protéine FUS de liaison à l'ARN/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme
14.
Opt Lett ; 43(6): 1271-1274, 2018 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-29543269

RÉSUMÉ

We realize Ho laser operation in a composite Tm/Ho:YAG gain medium for the first time, to the best of our knowledge, which was integrated via diffusion bonding the Tm-doped and Ho-doped crystals into a single bulk structure. A maximum output power around 6 W was obtained with a slope efficiency of 40.1% and conversion efficiency (CE) of 33.6% from the absorbed 785 nm laser diode to 2122 nm Ho laser, which is comparable with CE in 1.9 µm LD pumped Ho lasers. Such a scheme is demonstrated to be another valid way for Ho laser generation here, which is of significance to be adopted in other host media or waveguide structures for an assessable, compact, and efficient Ho laser.

15.
ACS Appl Mater Interfaces ; 8(27): 17118-26, 2016 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-27315327

RÉSUMÉ

Silk is useful as a drug carrier due to its biocompatibility, tunable degradation, and outstanding capacity in maintaining the function of drugs. Injectable silk hydrogels could deliver doxorubicin (DOX) for localized chemotherapy for breast cancer. To improve hydrogel properties, thixotropic silk nanofiber hydrogels in an all-aqueous solution were prepared and used to locally deliver DOX. The silk hydrogels displayed thixotropic capacity, allowing for easy injectability followed by solidification in situ. The hydrogels were loaded with DOX and released the drug over eight weeks with pH- and concentration-dependent release kinetics. In vitro and in vivo studies demonstrated that DOX-loaded silk hydrogels had good antitumor response, outperforming the equivalent dose of free DOX administered intravenously. Thixotropic silk hydrogels provide improved injectability to support sustained release, suggesting promising applications for localized chemotherapy.


Sujet(s)
Nanofibres , Antinéoplasiques , Doxorubicine , Systèmes de délivrance de médicaments , Hydrogels , Concentration en ions d'hydrogène , Soie
16.
Mol Carcinog ; 54(2): 102-10, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-24038513

RÉSUMÉ

CD44 is such one adhesion molecule that mediates interactions between acute myeloid leukemia (AML) cells and stromal. It has been demonstrated that CD4 plays a critical role in AML development. However, studies of functional single nucleotide polymorphisms (SNPs) in CD44 gene have not touched upon AML. This case-control study probed the contribution of functional SNPs in CD44 gene to AML susceptibility in eastern Chinese population. Five representative SNPs of CD44 (rs10836347C>T, rs13347C>T, rs1425802A>G, rs11821102G>A, rs713330T>C) were opted and genotyped in 421 AML patients and 461 healthy subjects and the association with risk of AML was estimated by logistic regression. Moreover, the potential role of rs13347C > T in AML was further explored. Compared with the rs13347CC genotype, CT carriers had a significant increase in AML susceptibility (adjusted odds ratio [OR] = 1.76; 95% confidence interval [CI] = 1.32-2.34), TT carriers had a further increased risk of AML (OR = 2.67; 95% CI = 1.69-4.21). Furthermore, our transient transfection assay and Western blot results demonstrated that the presence of rs13347T allele led to more CD44 expression. Yet, there exists no significant difference in genotype frequencies of the other four sites between cases and controls. Above findings suggest that rs13347C>T in 3'UTR of CD44 may be a genetic modifier for developing AML.


Sujet(s)
Asiatiques/génétique , Antigènes CD44/génétique , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/génétique , Polymorphisme de nucléotide simple , Régions 3' non traduites , Adulte , Études cas-témoins , Lignée cellulaire tumorale , Femelle , Fréquence d'allèle , Études d'associations génétiques , Prédisposition génétique à une maladie , Cellules HL-60 , Humains , Antigènes CD44/métabolisme , Mâle , microARN/génétique , microARN/métabolisme , Adulte d'âge moyen
17.
Carcinogenesis ; 35(12): 2687-97, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25239642

RÉSUMÉ

Gene fusion is among the primary processes that generate new genes and has been well characterized as potent pathway of oncogenesis. Here, by high-throughput RNA sequencing in nine paired human endometrial carcinoma (EC) and matched non-cancerous tissues, we obtained that chimeric translin-associated factor X-disrupted-in-schizophrenia 1 (TSNAX-DISC1) occurred significantly upregulated in multiple EC samples. Experimental investigation showed that TSNAX-DISC1 appears to be formed by splicing without chromosomal rearrangement. The chimera expression inversely correlated with the binding of CCCTC-binding factor (CTCF) to the insulators. Subsequent investigations indicate that long intergenic non-coding RNA lincRNA-NR_034037, separating TSNAX from DISC1, regulates TSNAX -DISC1 production and TSNAX/DISC1 expression levels by extricating CTCF from insulators. Dysregulation of TSNAX influences steroidogenic factor-1-stimulated transcription on the StAR promoter, altering progesterone actions, implying the association with cancer. Together, these results advance our understanding of the mechanism in which lincRNA-NR_034037 regulates TSNAX-DISC1 formation programs that tightly regulate EC development.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/anatomopathologie , Séquençage nucléotidique à haut débit , Protéines de tissu nerveux/génétique , Épissage des ARN/génétique , Protéines de répression/métabolisme , Adolescent , Sujet âgé , Animaux , Apoptose , Technique de Western , Facteur de liaison à la séquence CCCTC , Cycle cellulaire , Prolifération cellulaire , Enfant , Immunoprécipitation de la chromatine , Protéines de liaison à l'ADN/métabolisme , Tumeurs de l'endomètre/métabolisme , Endomètre/métabolisme , Femelle , Réarrangement des gènes , Humains , Techniques immunoenzymatiques , Souris , Souris nude , Grading des tumeurs , Protéines de tissu nerveux/métabolisme , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , Protéines de répression/génétique , RT-PCR , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
18.
PLoS One ; 9(5): e98251, 2014.
Article de Anglais | MEDLINE | ID: mdl-24879036

RÉSUMÉ

Breast cancer, one of the most common malignancies diagnosed among women worldwide, is a complex polygenic disease in the etiology of which genetic factors play an important role. Thus far, a subset of breast cancer genetic susceptibility loci has been addressed among Asian woman through genome-wide association studies (GWASs). In this study, we identified numerous long, intergenic, noncoding RNAs (lincRNAs) enriched in these breast cancer risk-related loci and identified 16 single nucleotide polymorphisms (SNPs) located within the sequences of lincRNA exonic regions. We examined whether these 16 SNPs are associated with breast cancer risk in 2539 cancer patients and 2818 control subjects from eastern, southern, and northern Chinese populations. We found that the C allele of the rs12325489C>T polymorphism in the exonic regions of lincRNA-ENST00000515084 was associated with a significantly increased risk of breast cancer (adjusted odds ratio [OR] = 1.79; 95% confidence interval [CI] = 1.50-2.12), compared with the rs12325489TT genotype. Biochemical analysis demonstrated that the C to T base change at rs12325489C>T disrupts the binding site for miRNA-370, thereby influencing the transcriptional activity of lincRNA-ENST00000515084 in vitro and in vivo, and affecting cell proliferation and tumor growth. Our findings indicate that the rs12325489C>T polymorphism in the lincRNA-ENST00000515084 exon may be a genetic modifier in the development of breast cancer.


Sujet(s)
Tumeurs du sein/génétique , Prédisposition génétique à une maladie/génétique , Étude d'association pangénomique , Polymorphisme de nucléotide simple , ARN long non codant/génétique , Adolescent , Animaux , Asiatiques/génétique , Études cas-témoins , Lignée cellulaire tumorale , Biologie informatique , Exons/génétique , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Souris , microARN/génétique , Adulte d'âge moyen
19.
Gastroenterology ; 146(7): 1714-26.e5, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24631494

RÉSUMÉ

BACKGROUND & AIMS: Thousands of long intergenic non-protein coding RNAs (lincRNAs) have been identified in mammals via genome-wide sequencing studies. Many are functional, but are expressed aberrantly by cancer cells. We investigated whether levels of lincRNAs are altered during the development of esophageal squamous cell carcinoma (ESCC). METHODS: We used quantitative real-time polymerase chain reaction to measure levels of 26 highly conserved lincRNAs in ESCC and surrounding nontumor tissues. A total of 182 ESCC and paired adjacent nontumor tissue samples were collected from patients undergoing tylectomy at The First Affiliate Hospital of Soochow University from 2001 through 2009; another 178 ESCC tissue pairs were collected from Guangzhou Medical University from 2002 through 2009. LincRNAs were expressed from lentiviral vectors or knocked down with small hairpin RNAs in Eca-109 and TE-1 cells. RESULTS: Levels of a lincRNA encoded by a gene located next to POU3F3 (linc-POU3F3) were significantly higher in ESCC than neighboring nontumor tissues. In RNA immunoprecipitation assays, linc-POU3F3 was associated with the EZH2 messenger RNA (mRNA). Overexpression of linc-POU3F3 in cell lines increased their proliferation and ability to form colonies, and reduced the expression of POU3F3 mRNA, whereas knockdown of linc-POU3F3 increased the levels of POU3F3 mRNA. CpG islands in POU3F3 were densely hypermethylated in cell lines that overexpressed linc-POU3F3; methylation at these sites was reduced by knockdown of linc-POU3F3. Pharmacologic inhibition of EZH2 increased the levels of POU3F3 mRNA and significantly reduced binding of DNA methyltransferase (DNMT)1, DNMT3A, and DNMT3B to POU3F3. ESCC cells with knockdown of linc-POU3F3 formed xenograft tumors more slowly in mice than control ESCC cells. CONCLUSIONS: Levels of linc-POU3F3 are increased in ESCC samples from patients compared with nontumor tissues. This noncoding RNA contributes to the development of ESCC by interacting with EZH2 to promote methylation of POU3F3, which encodes a transcription factor.


Sujet(s)
Carcinome épidermoïde/métabolisme , Ilots CpG , Méthylation de l'ADN , Tumeurs de l'oesophage/métabolisme , Facteurs de transcription à domaine POU/métabolisme , ARN long non codant/métabolisme , Adulte , Animaux , Séquence nucléotidique , Sites de fixation , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/chirurgie , Lignée cellulaire tumorale , Prolifération cellulaire , Chine , DNA (Cytosine-5-)-methyltransferase 1 , DNA (cytosine-5-)-methyltransferase/métabolisme , Méthylation de l'ADN/effets des médicaments et des substances chimiques , DNA methyltransferase 3A , Protéine-2 homologue de l'activateur de Zeste , Antienzymes/pharmacologie , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/chirurgie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Souris , Souris de lignée BALB C , Souris nude , Adulte d'âge moyen , Données de séquences moléculaires , Facteurs de transcription à domaine POU/génétique , Complexe répresseur Polycomb-2/antagonistes et inhibiteurs , Complexe répresseur Polycomb-2/génétique , Complexe répresseur Polycomb-2/métabolisme , ARN messager/métabolisme , Facteurs temps , Transfection , Charge tumorale , Régulation positive ,
20.
Carcinogenesis ; 34(12): 2908-17, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-23872665

RÉSUMÉ

The development of esophageal squamous cell carcinoma (ESCC) is a multifactorial process, and associations between genetic variants and ESCC have been identified in genome-wide association studies. The aim of this study was to evaluate the effects of single nucleotide polymorphisms (SNPs) of long intergenic non-coding RNAs (lincRNAs) on ESCC susceptibility in Chinese populations. We scoured exons of lincRNAs located in ESCC susceptibility loci for all probable functional SNPs. These 52 SNPs were opted for and genotyped in 1493 ESCC patients and 1553 cancer-free controls from eastern and southern Chinese populations, and their associations with the risk for ESCC were estimated using logistic regression. Functional relevance was further examined by biochemical assays. Significant differences were found between patients and controls in the genotype frequencies for the rs11752942A>G site in the lincRNA-uc003opf.1 exon. Compared with the rs11752942AA genotype, AG and GG genotypes had a significantly reduced risk of ESCC (adjusted odds ratio = 0.73; 95% confidence interval = 0.63-0.84). Biochemical analysis demonstrated that, when compared with the A allele, the rs11752942G allele could markedly attenuate the level of lincRNA-uc003opf.1 both in vivo and in vitro by binding micro-RNA-149*, thereby affecting cell proliferation and tumor growth. These findings indicated that functional polymorphism rs11752942A>G in lincRNA-uc003opf.1 exon might be a genetic modifier for the development of ESCC.


Sujet(s)
Asiatiques/génétique , Carcinome épidermoïde/génétique , Tumeurs de l'oesophage/génétique , Prédisposition génétique à une maladie/génétique , Polymorphisme de nucléotide simple/génétique , ARN long non codant/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Allèles , Études cas-témoins , Prolifération cellulaire , Carcinome épidermoïde de l'oesophage , Exons/génétique , Fréquence d'allèle/génétique , Génotype , Humains , microARN/génétique , Adulte d'âge moyen , Risque , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE