Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
J Control Release ; 370: 798-810, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754633

ABSTRACT

Myocardial infarction (MI) is a major cause of morbidity and mortality worldwide. Although clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) gene editing holds immense potential for genetic manipulation, its clinical application is hindered by the absence of an efficient heart-targeted drug delivery system. Herein, we developed CRISPR-Cas9 ribonucleoprotein (RNP)-loaded extracellular vesicles (EVs) conjugated with cardiac-targeting peptide (T) for precise cardiac-specific genome editing. RNP complexes containing Cas9 and single guide RNA targeting miR-34a, an MI-associated molecular target, were loaded into EVs (EV@RNP). Gene editing by EV@RNP attenuated hydrogen peroxide-induced apoptosis in cardiomyocytes via miR-34a inhibition, evidenced by increased B-cell lymphoma 2 levels, decreased Bcl-2-associated X protein levels, and the cleavage of caspase-3. Additionally, to improve cardiac targeting in vivo, we used click chemistry to form functional T-EV@RNP by conjugating T peptides to EV@RNP. Consequently, T-EV@RNP-mediated miR-34a genome editing might exert a protective effect against MI, reducing apoptosis, ameliorating MI injury, and facilitating the recovery of cardiac function. In conclusion, the genome editing delivery system established by loading CRISPR/Cas9 RNP with cardiac-targeting EVs is a powerful approach for precise and tissue-specific gene therapy for cardiovascular disease.


Subject(s)
CRISPR-Cas Systems , Extracellular Vesicles , Gene Editing , MicroRNAs , Myocardial Infarction , Myocytes, Cardiac , Ribonucleoproteins , Gene Editing/methods , Extracellular Vesicles/metabolism , Animals , Ribonucleoproteins/genetics , Myocytes, Cardiac/metabolism , Myocardial Infarction/therapy , Myocardial Infarction/genetics , MicroRNAs/administration & dosage , MicroRNAs/genetics , Apoptosis/drug effects , Male , Mice, Inbred C57BL , Humans , CRISPR-Associated Protein 9/genetics , Peptides/chemistry , Mice
2.
J Extracell Vesicles ; 12(10): e12371, 2023 10.
Article in English | MEDLINE | ID: mdl-37795828

ABSTRACT

Small-interfering RNA (siRNA) therapy is considered a powerful therapeutic strategy for treating cardiac hypertrophy, an important risk factor for subsequent cardiac morbidity and mortality. However, the lack of safe and efficient in vivo delivery of siRNAs is a major challenge for broadening its clinical applications. Small extracellular vesicles (sEVs) are a promising delivery system for siRNAs but have limited cell/tissue-specific targeting ability. In this study, a new generation of heart-targeting sEVs (CEVs) has been developed by conjugating cardiac-targeting peptide (CTP) to human peripheral blood-derived sEVs (PB-EVs), using a simple, rapid and scalable method based on bio-orthogonal copper-free click chemistry. The experimental results show that CEVs have typical sEVs properties and excellent heart-targeting ability. Furthermore, to treat cardiac hypertrophy, CEVs are loaded with NADPH Oxidase 4 (NOX4) siRNA (siNOX4). Consequently, CEVs@siNOX4 treatment enhances the in vitro anti-hypertrophic effects by CEVs with siRNA protection and heart-targeting ability. In addition, the intravenous injection of CEVs@siNOX4 into angiotensin II (Ang II)-treated mice significantly improves cardiac function and reduces fibrosis and cardiomyocyte cross-sectional area, with limited side effects. In conclusion, the utilization of CEVs represents an efficient strategy for heart-targeted delivery of therapeutic siRNAs and holds great promise for the treatment of cardiac hypertrophy.


Subject(s)
Extracellular Vesicles , Mice , Humans , Animals , RNA, Small Interfering/analysis , RNA, Small Interfering/genetics , NADPH Oxidase 4/genetics , NADPH Oxidase 4/analysis , Extracellular Vesicles/chemistry , Cardiomegaly/therapy , Cardiomegaly/prevention & control , Myocytes, Cardiac
3.
Stem Cell Res ; 65: 102940, 2022 12.
Article in English | MEDLINE | ID: mdl-36260954

ABSTRACT

PITX2 is a homeobox gene located in the human 4q25 locus and is commonly associated with atrial fibrillation (AF). Here, we generated two PITX2 knock-out human induced pluripotent stem cell (iPSC) lines using CRISPR/Cas9 genome editing. The edited iPSCs maintained fullpluripotency, normal karyotype and spontaneousdifferentiation capability. This cell line provides a suitable model for investigating the physiopathologyof PITX2 mutation in atrial fibrillation.


Subject(s)
Atrial Fibrillation , Induced Pluripotent Stem Cells , Humans , CRISPR-Cas Systems/genetics
4.
Stem Cell Res ; 64: 102901, 2022 10.
Article in English | MEDLINE | ID: mdl-36037555

ABSTRACT

TTN mutations are the common genetic cause for various types of cardiomyopathies (e.g., dilated cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, and arrhythmogenic right ventricular cardiomyopathy) and skeletal myopathies. Here, we generated three TTN knock-out human induced pluripotent stem cell (iPSC) lines using CRISPR/Cas9 system. These cell lines, which exhibit normal karyotype, typical morphology and pluripotency, could provide useful platform for investigating the role of TTN in associated disorders.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia , Cardiomyopathies , Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , CRISPR-Cas Systems/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathies/metabolism , Arrhythmogenic Right Ventricular Dysplasia/genetics , Mutation , Connectin/genetics , Connectin/metabolism
5.
Stem Cell Res ; 63: 102878, 2022 08.
Article in English | MEDLINE | ID: mdl-35917600

ABSTRACT

E192K missense mutation of TPM1 has been found in different types of cardiomyopathies (e.g., hypertrophic cardiomyopathy, dilated cardiomyopathy, and left ventricular non-compaction), leading to systolic dysfunction, diastolic dysfunction, and/or tachyarrhythmias. Here, we generated a heterozygous TPM1-E192K knock-in human induced pluripotent stem cell (iPSC) line using CRISPR/Cas9-based genome editing system. The cells exhibit normal karyotype, typical stem cell morphology, expression of pluripotency markers and differentiation ability into three germ layers. Accordingly, this cell line could provide a useful cell resource for exploring the pathogenic role of TPM1-E192K mutation in different types of cardiomyopathies.


Subject(s)
Cardiomyopathies , Induced Pluripotent Stem Cells , CRISPR-Cas Systems/genetics , Cardiomyopathies/metabolism , Gene Editing , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Tropomyosin/genetics
6.
Heart Rhythm ; 19(9): 1450-1458, 2022 09.
Article in English | MEDLINE | ID: mdl-35660473

ABSTRACT

BACKGROUND: Exosomal long noncoding RNAs (lncRNAs) are known as ideal diagnostic biomarkers of various diseases. However, there are no reports on the use of serum exosomal lncRNAs as diagnostic biomarkers for atrial fibrillation (AF). OBJECTIVE: The purpose of this study was to explore serum exosomal lncRNAs as a useful tool for diagnosing AF. METHODS: Serum exosomes from patients with persistent AF and controls were isolated using a polymer-based exosome precipitation kit. We conducted a multiphase process including screening and 2 independent validation phases. In the screening phase, serum exosomal lncRNA expression profiles were examined using RNA sequencing analysis. In 2 validation phases, we evaluated the expression levels of candidate exosomal lncRNAs using quantitative reverse transcription polymerase chain reaction. Finally, we performed different statistical and functional analyses. RESULTS: After the screening phase, we identified 26 differentially expressed lncRNAs (ie, 15 upregulated and 11 downregulated lncRNAs with a |fold change| ≥2 and P <.05) in serum exosomes from patients with persistent AF compared with controls. We then screened out 6 exosomal lncRNAs as biomarker candidates following parameters: read length ≥200 nucleotides; exon number ≥2; and coding potential score <0.1. In 2 validation phases, exosomal lncRNAs LOC105377989 and LOC107986997 were consistently upregulated in the serum of patients with persistent AF compared with controls (P <.0001). Moreover, both exosomal lncRNAs exhibited significant diagnostic validity for AF. Notably, exosomal lncRNA LOC107986997 was involved in AF-related pathophysiological mechanisms. CONCLUSION: Serum-derived exosomal lncRNA LOC107986997 could serve as a potential biomarker for AF diagnosis.


Subject(s)
Atrial Fibrillation , Exosomes , RNA, Long Noncoding , Atrial Fibrillation/diagnosis , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Biomarkers/metabolism , Exosomes/genetics , Exosomes/metabolism , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
7.
Clin Sci (Lond) ; 136(8): 621-637, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35411927

ABSTRACT

Small extracellular vesicles (sEVs) are nanometer-sized membranous vesicles that contribute to the pathogenesis of atrial fibrillation (AF). Here, we investigated the role of sEVs derived from patients with persistent AF in the pathophysiology of AF. First, we evaluated the pathological effects of sEVs derived from the peripheral blood of patients with persistent AF (AF-sEVs). AF-sEVs treatment reduced cell viability, caused abnormal Ca2+ handling, induced reactive oxygen species (ROS) production and led to increased CaMKII activation of non-paced and paced atrial cardiomyocytes. Next, we analyzed the miRNA profile of AF-sEVs to investigate which components of AF-sEVs promote arrhythmias, and we selected six miRNAs that correlated with CaMKII activation. qRT-PCR experiment identified that miR-30a-5p was significantly down-regulated in AF-sEVs, paced cardiomyocytes, and atrial tissues of patients with persistent AF. CaMKII was predicted by bioinformatics analysis as a miR-30a-5p target gene and validated by a dual luciferase reporter; hence, we evaluated the effects of miR-30a-5p on paced cardiomyocytes and validated miR-30a-5p as a pro-arrhythmic signature of AF-sEVs. Consequently, AF-sEVs-loaded with miR-30a-5p attenuated pacing-induced Ca2+-handling abnormalities, whereas AF-sEVs-loaded with anti-miR-30a-5p reversed the change in paced cardiomyocytes. Taken together, the regulation of CaMKII by miR-30a-5p revealed that miR-30a-5p is a major mediator for AF-sEVs-mediated AF pathogenesis. Accordingly, these findings suggest that sEVs derived from patients with persistent AF exacerbate arrhythmogenesis via miR-30a-5p.


Subject(s)
Atrial Fibrillation , Extracellular Vesicles , MicroRNAs , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Extracellular Vesicles/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism
8.
Mol Ther Nucleic Acids ; 24: 1024-1032, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34141457

ABSTRACT

Small extracellular vesicles (sEVs) are nanometer-sized membranous vesicles secreted by cells, with important roles in physiological and pathological processes. Recent research has established the application of sEVs as therapeutic vehicles in various conditions, including heart disease. However, the high risk of off-target effects is a major barrier for their introduction into the clinic. This study evaluated the use of modified sEVs expressing high levels of cardiac-targeting peptide (CTP) for therapeutic small interfering RNA (siRNA) delivery in myocarditis, an inflammatory disease of heart. sEVs were extracted from the cell culture medium of HEK293 cells stably expressing CTP-LAMP2b (referred to as C-sEVs). The cardiac targeting ability of C-sEVs with the highest CTP-LAMP2b expression was >2-fold greater than that of normal sEVs (N-sEVs). An siRNA targeting the receptor for advanced glycation end products (RAGE) (siRAGE) was selected as a therapeutic siRNA and loaded into C-sEVs. The efficiency of cardiac-specific siRNA delivery via C-sEVs was >2-fold higher than that via N-sEVs. Furthermore, siRAGE-loaded C-sEVs attenuated inflammation in both cell culture and an in vivo model of myocarditis. Taken together, C-sEVs may be a useful drug delivery vehicle for the treatment of heart disease.

9.
Korean Circ J ; 51(2): 157-170, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33525071

ABSTRACT

BACKGROUND AND OBJECTIVES: Ambient particulate matter (PM) in real urban air pollution (RUA) is an environmental health risk factor associated with increased cardiac events. This study investigated the threshold level to induce arrhythmia, as well as arrhythmogenic mechanism of RUA that mainly consisted of PM <2.5 µm in aerodynamic diameter close to ultrafine particles. METHODS: RUA was artificially produced by a lately developed pyrolysis based RUA generator. C57BL/6 mice were divided into 4 groups: a control group (control, n=12) and three groups with exposure to RUA with the concentration of 200 µg/m³ (n=12), 400 µg/m³ (n=12), and 800 µg/m³ (n=12). Mice were exposed to RUA at each concentration for 8 hr/day and 5 day/week to mimic ordinary human activity during 3 weeks. RESULTS: The QRS and QTc intervals, as well as intracellular Ca2+ duration, apicobasal action potential duration (APD) gradient, fibrosis, and inflammation of left ventricle of mouse hearts were increased dose-dependently with the increase of RUA concentration, and significantly increased at RUA concentration of 400 µg/m³ compared to control (all p<0.001). In mice exposed to RUA concentration of 800 µg/m³, spontaneous ventricular arrhythmia was observed in 42%, with significant increase of inflammatory markers, phosphorylated Ca2+/calmodulin-dependent protein kinase II (CaMKII), and phospholamban (PLB) compared to control. CONCLUSIONS: RUA could induce electrophysiological changes such as APD and QT prolongation, fibrosis, and inflammation dose-dependently, with significant increase of ventricular arrhythmia at the concentration of 400 µg/m³. RUA concentration of 800 µg/m³ increased phosphorylation of CaMKII and PLB.

10.
J Control Release ; 331: 62-73, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33460670

ABSTRACT

Curcumin exerts therapeutic effects in heart disease, but has limited bioavailability. Extracellular vesicles (EVs) have gained attention as nanovehicles; however, the poor targeting ability of systemically administered EVs still remains a crucial issue. Herein, we generated heart-targeted EVs (CTP-EVs) by functionalizing EVs surface with cardiac targeting peptide (CTP) using genetic modification of EVs-secreting cells, and further loaded curcumin into CTP-EVs (CTP-EVs-Cur). Consequently, CTP-EVs were able to specifically deliver curcumin to the heart. In addition, curcumin-loaded CTP-EVs possess improved bioavailability, and are fully functional with a high cardioprotective efficiency. Moreover, we loaded miR-144-3p in CTP-EVs-Cur following validation of miR-144-3p as a major contributor in curcumin-mediated therapeutic effects. The simultaneous packing of curcumin and miR-144-3p in CTP-EVs not only retains the active heart-targeting ability but also achieves enhanced cardioprotective effects both in vitro and in vivo, indicating the possibility of combining and sustaining their therapeutic potential by simultaneously loading in CTP-EVs. Therefore, CTP-EVs could be a potential and effective strategy for the delivery of therapeutic molecules, thereby providing a promising nanomedicine for MI therapy.


Subject(s)
Curcumin , Extracellular Vesicles , MicroRNAs , Myocardial Infarction , Heart , Humans , Myocardial Infarction/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL