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
11.
Clin Sci (Lond) ; 134(8): 985-999, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32297634

ABSTRACT

Small extracellular vesicles (sEVs) as natural membranous vesicles are on the frontiers of nanomedical research, due to their ability to deliver therapeutic molecules such as microRNAs (miRNAs). The miRNA-21 (miR-21) is thought to be involved in the initiation and development of myocardial infarction (MI). Here, we examined whether miR-21 regulation using human peripheral blood-derived sEVs (PB-sEVs) could serve as a potential therapeutic strategy for MI. First, we examined miR-21 levels in hypoxic conditions and validated the ability of PB-sEVs to serve as a potential delivery system for miRNAs. Further, bioinformatics analysis and luciferase assay were performed to identify target genes of miR-21 mechanistically. Among numerous target pathways, we focused on nitrogen metabolism, which remains relatively unexplored compared with other possible miR-21-mediated pathways; hence, we aimed to determine novel target genes of miR-21 related to nitrogen metabolism. In hypoxic conditions, the expression of miR-21 was significantly up-regulated and correlated with nitric oxide synthase 3 (NOS3) levels, which in turn influences cardiac function. The down-regulation of miR-21 expression by PB-sEVs loaded with anti-miR-21 significantly improved survival rates, consistent with the augmentation of cardiac function. However, the up-regulation of miR-21 expression by PB-sEVs loaded with miR-21 reversed these effects. Mechanistically, miR-21 targeted and down-regulated the mRNA and protein expression of striatin (STRN), which could regulate NOS3 expression. In conclusion, we identified a novel therapeutic strategy to improve cardiac function by regulating the expression of miR-21 with PB-sEVs as an miR-21 or anti-miR-21 delivery vehicle and confirmed the miR-21-associated nitrogen metabolic disorders in MI.


Subject(s)
Extracellular Vesicles/chemistry , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Animals , Blood Chemical Analysis , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Female , Genetic Therapy , Humans , Male , Mice, Inbred C57BL , MicroRNAs/administration & dosage , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism
12.
PLoS One ; 14(7): e0220036, 2019.
Article in English | MEDLINE | ID: mdl-31329632

ABSTRACT

Exosomes might have an unimproved potential to serve as effective delivery vehicles. However, when exosomes are developed for therapeutic applications, a method to enhance their delivery is important. This study aimed to evaluate wheather calcium chloride (CaCl2) or other chloride compounds could enhance exosome delivery to various cells without causing toxicity. Exosomes were purified from human serum by using the ExoQuick exosome precipitation kit. Isolated exosomes were mixed with CaCl2 at concentrations ranging from 100 µM to 1 mM, and then washed using Amicon filter for treating the cells. The delivery efficiency of exosomes and the viability of the cells [HEK 293 (human kidney cells) and H9C2 (rat cardiomyocytes)] were evaluated. Cellular uptake of exosomes was observed using a confocal microscope based on PKH26 labeling of exosomes. CaCl2 increased the delivery of exosomes in a dose- and treatment time-dependent manner. In HEK 293 cells, a CaCl2 concentration of 400 µM and exposure time of 12 h increased the delivery of exosomes by >20 times compared with controls. In H9C2 cells, a CaCl2 concentration of 400 µM and exposure time of >24 h increased the delivery of exosomes by >400 times compared with controls. The viability of both cell lines was maintained up to a CaCl2 concentration of 1 mM. However, cobalt chloride, cupric chloride, and magnesium chloride did not change the delivery of exosomes in both cell lines. These results suggest that the use of CaCl2 treatment might be a useful method for enhancing the delivery of exosomes.


Subject(s)
Calcium Chloride/pharmacology , Exosomes/metabolism , Secretory Pathway , Animals , Cells, Cultured , Exocytosis , Exosomes/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats
13.
Int J Mol Med ; 44(1): 358, 2019 07.
Article in English | MEDLINE | ID: mdl-31115503

ABSTRACT

Exosomes serve important functions in cell­to­cell communication and biological functions by serving as a delivery cargo shuttle for various molecules. The application of an improved delivery method for microRNAs (miRNAs/miRs) may enhance their potential as a therapeutic tool in cardiac diseases. Thus, the present study investigated whether human peripheral blood­derived exosomes may be used as a delivery cargo system for miRNAs, and whether the delivery of miR­21 using a human peripheral blood derived­exosome may influence the degree of remodeling following myocardial infarction (MI). In H9C2 and HL­1 cells, miR­21 expression was successfully regulated by treatment with human peripheral blood derived­exosomes loaded with an miR­21 mimic or inhibitor compared with untreated cells. In addition, the mRNA and protein expression levels of SMAD family member 7 (Smad7), phosphatase and tensin homolog (PTEN) and matrix metalloproteinase 2 (MMP2), which are involved in cardiac fibrosis, were associated with the uptake of miR­21 mimic­ or inhibitor­loaded exosomes. Similarly, the in vivo mRNA and protein expression of Smad7, PTEN and MMP2 were altered following treatment with miR­21 mimic­ or inhibitor­loaded exosomes. Furthermore, miR­21 mimic­loaded exosomes enhanced fibrosis, whereas miR­21 inhibitor­loaded exosomes reduced fibrosis in a mouse MI model. These results suggested that miRNA­loaded human peripheral blood derived­exosomes may be used as a therapeutic tool for cardiac diseases. [the original article was published in International Journal of Molecular Medicine 43: 2319­2328, 2019; DOI:10.3892/ijmm.2019.4150].

14.
Int J Mol Med ; 43(6): 2319-2328, 2019 06.
Article in English | MEDLINE | ID: mdl-30942393

ABSTRACT

Exosomes serve important functions in cell­to­cell communication and biological functions by serving as a delivery cargo shuttle for various molecules. The application of an improved delivery method for microRNAs (miRNAs/miRs) may enhance their potential as a therapeutic tool in cardiac diseases. Thus, the present study investigated whether human peripheral blood­derived exosomes may be used as a delivery cargo system for miRNAs, and whether the delivery of miR­21 using a human peripheral blood derived­exosome may influence the degree of remodeling following myocardial infarction (MI). In H9C2 and HL­1 cells, miR­21 expression was successfully regulated by treatment with human peripheral blood derived­exosomes loaded with an miR­21 mimic or inhibitor compared with untreated cells. In addition, the mRNA and protein expression levels of SMAD family member 7 (Smad7), phosphatase and tensin homolog (PTEN) and matrix metalloproteinase 2 (MMP2), which are involved in cardiac fibrosis, were associated with the uptake of miR­21 mimic­ or inhibitor­loaded exosomes. Similarly, the in vivo mRNA and protein expression of Smad7, PTEN and MMP2 were altered following treatment with miR­21 mimic­ or inhibitor­loaded exosomes. Furthermore, miR­21 mimic­loaded exosomes enhanced fibrosis, whereas miR­21 inhibitor­loaded exosomes reduced fibrosis in a mouse MI model. These results suggested that miRNA­loaded human peripheral blood derived­exosomes may be used as a therapeutic tool for cardiac diseases.


Subject(s)
Drug Carriers/chemistry , Exosomes/chemistry , MicroRNAs/administration & dosage , Myocardial Infarction/therapy , Aged , Animals , Cell Line , Female , Fibrosis , Genetic Therapy , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/therapeutic use , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology
15.
FASEB J ; 33(5): 5979-5989, 2019 05.
Article in English | MEDLINE | ID: mdl-30753098

ABSTRACT

Atrial fibrillation (AF), the most common type of cardiac arrhythmia, is thought to be regulated by changes in microRNA (miRNA) expression. However, the evidence for this is inconsistent. The high stability and expression of circulating exosomal miRNAs may allow their use as candidate biomarkers. For the discovery phase, exosomes were isolated from the serum of patients with supraventricular tachycardia (SVT) as the controls (n = 5) and with paroxysmal AF (n = 4) and persistent AF (n = 5) for microarray analysis of miRNAs. Forty-five miRNAs were expressed significantly higher (>1.5-fold) in patients with persistent AF, but not in patients with paroxysmal AF, relative to the levels in patients with SVT control. Notably, expression of 5 miRNAs (miRNA-103a, -107, -320d, -486, and let-7b) was elevated by more than 4.5-fold in patients with persistent AF. For the validation phase, miRNAs were analyzed using quantitative RT-PCR analysis in exosomes from the serum of patients with SVT control (n = 20) and patients with persistent AF (n = 40). These miRNAs and their target genes were involved in atrial function and structure, oxidative stress, and fibrosis pathways. These findings suggest that serum exosomal miRNAs might be used as novel biomarkers to reflect the progression of AF.-Mun, D., Kim, H., Kang, J.-Y., Park, H., Park, H., Lee, S.-H., Yun, N., Joung, B. Expression of miRNAs in circulating exosomes derived from patients with persistent atrial fibrillation.


Subject(s)
Atrial Fibrillation/blood , Biomarkers/blood , Exosomes/metabolism , MicroRNAs/metabolism , Tachycardia, Supraventricular/blood , Aged , Cardiac Catheterization , Disease Progression , Female , Fibrosis/metabolism , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Oxidative Stress
16.
Yonsei Med J ; 59(6): 736-745, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29978610

ABSTRACT

PURPOSE: Bioactive molecules critical to intracellular signaling are contained in extracellular vesicles (EVs) and have cardioprotective effects in ischemia/reperfusion (IR) injured hearts. This study investigated the mechanism of the cardioprotective effects of EVs derived from hypoxia-preconditioned human mesenchymal stem cells (MSCs). MATERIALS AND METHODS: EV solutions (0.4 µg/µL) derived from normoxia-preconditioned MSCs (EV(NM)) and hypoxia-preconditioned MSCs (EV(HM)) were delivered in a rat IR injury model. Successful EV delivery was confirmed by the detection of PKH26 staining in hearts from EV-treated rats. RESULTS: EV(HM) significantly reduced infarct size (24±2% vs. 8±1%, p<0.001), and diminished arrhythmias by recovering electrical conduction, I(Na) current, and Cx43 expression. EV(HM) also reversed reductions in Wnt1 and ß-catenin levels and increases in GSK3ß induced after IR injury. miRNA-26a was significantly increased in EV(HM), compared with EV(NM), in real-time PCR. Finally, in in vitro experiments, hypoxia-induced increases in GSK3ß expression were significantly reduced by the overexpression of miRNA-26a. CONCLUSION: EV(HM) reduced IR injury by suppressing GSK3ß expression via miRNA-26a and increased Cx43 expression. These findings suggest that the beneficial effect of EVHM is related with Wnt signaling pathway.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Hypoxia/physiopathology , Ischemia/physiopathology , MicroRNAs/metabolism , Reperfusion Injury , Animals , Connexin 43 , Extracellular Vesicles , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/genetics , Humans , Hypoxia/metabolism , Immunohistochemistry , Male , Mesenchymal Stem Cells/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA, Small Interfering/metabolism , Rats , beta Catenin
17.
Biochem Biophys Res Commun ; 499(4): 803-808, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29621543

ABSTRACT

Naturally occurring RNA carriers such as exosomes might be an untapped source of effective delivery vehicles. However, if exosomes are to be exploited for therapeutic applications, they must target specific tissues or cell types to avoid off-target effects. This study evaluated whether genetic modification of exosomes could enhance exosome delivery to heart cells and heart tissue without toxicity. Exosomes expressing cardiac-targeting peptide (CTP)-Lamp2b on the exosomal membrane (CTP-Exo) were generated by introducing vectors encoding CTP-Lamp2b into HEK 293 cells. The expression of CTP-Lamp2b peptide on exosomes was stabilized by attaching glycosylation sequences. Exosomes expressing only Lamp2b on exosomal membranes (CTL-Exo) were generated as a control. The in vitro and in vivo uptake of CTL-Exo and CTP-Exo was evaluated in cell lines and mice. Both exosomes were delivered to HEK 293 and H9C2 cells. The delivery of the exosome was not different between CTP-Exo and CTL-Exo in HEK 293 cells, whereas the delivery of CTP-Exo was 16% greater than that of CTL-Exo in H9C2 cells (P = 0.047). Cell viability was maintained at almost 100% with different dosages of both CTL-Exo and CTP-Exo. Moreover, compared with CTL-Exo, the in vivo delivery of exosomes to the hearts of mice was increased by 15% with CTP-Exo (P = 0.035). The delivery to livers and spleens was not different between the two exosomes. Genetic modification of exosomes by expressing CTP-Lamp2b on the exosomal membrane enhanced exosome delivery to heart cells and the heart tissue. These results suggested that CTP-Exo might be used as a therapeutic tool for heart disease.


Subject(s)
Drug Delivery Systems , Exosomes/metabolism , Myocardium/metabolism , Peptides/pharmacology , Animals , Exosomes/drug effects , HEK293 Cells , Humans , Mice , Organ Specificity , Rats
18.
Yonsei Med J ; 59(2): 279-286, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29436197

ABSTRACT

PURPOSE: The mechanisms underlying repolarization abnormalities during pregnancy are not fully understood. Although maternal serotonin (5-hydroxytryptamine, 5-HT) production is an important determinant for normal fetal development in mice, its role in mothers remains unclear. We evaluated the role of serotonin in ventricular repolarization in mice hearts via 5Htr3 receptor (Htr3a) and investigated the mechanism of QT-prolongation during pregnancy. MATERIALS AND METHODS: We measured current amplitudes and the expression levels of voltage-gated K⁺ (Kv) channels in freshly-isolated left ventricular myocytes from wild-type non-pregnant (WT-NP), late-pregnant (WT-LP), and non-pregnant Htr3a homozygous knockout mice (Htr3a(-/-)-NP). RESULTS: During pregnancy, serotonin and tryptophan hydroxylase 1, a rate-limiting enzyme for the synthesis of serotonin, were markedly increased in hearts and serum. Serotonin increased Kv current densities concomitant with the shortening of the QT interval in WT-NP mice, but not in WT-LP and Htr3a(-/-)-NP mice. Ondansetron, an Htr3 antagonist, decreased Kv currents in WT-LP mice, but not in WT-NP mice. Kv4.3 directly interacted with Htr3a, and this binding was facilitated by serotonin. Serotonin increased the trafficking of Kv4.3 channels to the cellular membrane in WT-NP. CONCLUSION: Serotonin increases repolarizing currents by augmenting Kv currents. Elevated serotonin levels during pregnancy counterbalance pregnancy-related QT prolongation by facilitating Htr3-mediated Kv currents.


Subject(s)
Action Potentials , Heart Ventricles/metabolism , Serotonin/metabolism , Action Potentials/drug effects , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Disease Models, Animal , Electrocardiography , Female , HSC70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heart Ventricles/drug effects , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Potassium Channels/metabolism , Pregnancy , Rabbits , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT3/metabolism , Serotonin 5-HT3 Receptor Agonists/pharmacology
19.
Heart Rhythm ; 15(5): 770-779, 2018 05.
Article in English | MEDLINE | ID: mdl-28963014

ABSTRACT

BACKGROUND: Left stellectomy has become an important therapeutic option for patients with potentially fatal arrhythmias. However, the antiarrhythmic mechanism of left stellectomy is not well known. The cholinergic anti-inflammatory pathway (CAIP) is a complex immune mechanism that regulates peripheral inflammatory responses. OBJECTIVE: The purpose of this study was to evaluate the effect of left stellectomy on CAIP using rat experimental autoimmune myocarditis (EAM) models. METHODS: EAM was produced by injecting 2 mg of porcine cardiac myosin into the footpads of rats. Left stellectomy was performed before EAM induction. We evaluated the effect of left stellectomy on arrhythmic events, survival, inflammation, and CAIP in rats without and with EAM. RESULTS: Left stellectomy prevented arrhythmia and improved survival in EAM rats. Left stellectomy decreased the levels of tumor necrosis factor α, interleukin 6, and high mobility group box 1 (P < .05 vs EAM) in serum and heart tissues from EAM rats. In heart rate variability analysis, high-frequency peaks of the power spectrum densities, reflecting parasympathetic cardiovagal tone, were significantly decreased in EAM rats, but increased after left stellectomy. The ratios of phosphorylated STAT3/STAT3 (signal transducer and activator of transcription 3) and phosphorylated JAK2/JAK2 (Janus kinase 2) decreased in cell lysates of the spleen, liver, and heart in EAM rats. However, the same ratios significantly increased after left stellectomy. Nuclear factor κB in cell lysates of the spleen, liver, and heart increased in EAM rats, but decreased after left stellectomy. CONCLUSION: In EAM models, left stellectomy increased survival of the rats while showing antiarrhythmic effects with reduced inflammation via activation of the JAK2-STAT3-mediated signaling cascade. Our findings suggest an exciting opportunity to develop new and novel therapeutics to attenuate cardiac inflammation.


Subject(s)
Autonomic Nerve Block/methods , Janus Kinase 2/metabolism , Myocarditis/therapy , Myocardium/metabolism , STAT3 Transcription Factor/metabolism , Animals , Disease Models, Animal , Heart Rate , Male , Myocarditis/metabolism , Myocarditis/physiopathology , Myocardium/pathology , Rats , Rats, Inbred Lew , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...