Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Cell Mol Life Sci ; 81(1): 196, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658440

ABSTRACT

Telomeres as the protective ends of linear chromosomes, are synthesized by the enzyme telomerase (TERT). Critically short telomeres essentially contribute to aging-related diseases and are associated with a broad spectrum of disorders known as telomeropathies. In cardiomyocytes, telomere length is strongly correlated with cardiomyopathies but it remains ambiguous whether short telomeres are the cause or the result of the disease. In this study, we employed an inducible CRISPRi human induced pluripotent stem cell (hiPSC) line to silence TERT expression enabling the generation of hiPSCs and hiPSC-derived cardiomyocytes with long and short telomeres. Reduced telomerase activity and shorter telomere lengths of hiPSCs induced global transcriptomic changes associated with cardiac developmental pathways. Consequently, the differentiation potential towards cardiomyocytes was strongly impaired and single cell RNA sequencing revealed a shift towards a more smooth muscle cell like identity in the cells with the shortest telomeres. Poor cardiomyocyte function and increased sensitivity to stress directly correlated with the extent of telomere shortening. Collectively our data demonstrates a TERT dependent cardiomyogenic differentiation defect, highlighting the CRISPRi TERT hiPSCs model as a powerful platform to study the mechanisms and consequences of short telomeres in the heart and also in the context of telomeropathies.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Telomerase , Telomere , Telomerase/metabolism , Telomerase/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Telomere/metabolism , Telomere Shortening , Cell Line
2.
Basic Res Cardiol ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38639887

ABSTRACT

Hypertrophic cardiomyopathy (HCM) constitutes the most common genetic cardiac disorder. However, current pharmacotherapeutics are mainly symptomatic and only partially address underlying molecular mechanisms. Circular RNAs (circRNAs) are a recently discovered class of non-coding RNAs and emerged as specific and powerful regulators of cellular functions. By performing global circRNA-specific next generation sequencing in cardiac tissue of patients with hypertrophic cardiomyopathy compared to healthy donors, we identified circZFPM2 (hsa_circ_0003380). CircZFPM2, which derives from the ZFPM2 gene locus, is a highly conserved regulatory circRNA that is strongly induced in HCM tissue. In vitro loss-of-function experiments were performed in neonatal rat cardiomyocytes, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), and HCM-patient-derived hiPSC-CMs. A knockdown of circZFPM2 was found to induce cardiomyocyte hypertrophy and compromise mitochondrial respiration, leading to an increased production of reactive oxygen species and apoptosis. In contrast, delivery of recombinant circZFPM2, packaged in lipid-nanoparticles or using AAV-based overexpression, rescued cardiomyocyte hypertrophic gene expression and promoted cell survival. Additionally, HCM-derived cardiac organoids exhibited improved contractility upon CM-specific overexpression of circZFPM2. Multi-Omics analysis further promoted our hypothesis, showing beneficial effects of circZFPM2 on cardiac contractility and mitochondrial function. Collectively, our data highlight that circZFPM2 serves as a promising target for the treatment of cardiac hypertrophy including HCM.

3.
Stem Cell Res ; 77: 103404, 2024 Mar 24.
Article in English | MEDLINE | ID: mdl-38552356

ABSTRACT

Fabry disease (FD) is a rare and inherited monogenetic disease caused by mutations in the X-chromosomal alpha-galactosidase A gene GLA concomitant with accumulation of its substrate globotriaosylceramide (Gb3) and multi-organ symptoms. We derived an induced pluripotent stem cell line, MHHi029-A, from a male FD patient carrying a c.959A > T missense mutation in the GLA gene. The hiPSCs show a normal karyotype, expression of pluripotency markers and trilineage differentiation capacity. Importantly, they present the patient-specific mutation in the GLA gene and are therefore a valuable resource for investigating the FD mechanism and identifying novel therapies.

4.
Mol Ther Nucleic Acids ; 35(1): 102118, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38314095

ABSTRACT

Elucidating the pathobiological mechanisms underlying post-acute pulmonary sequelae following SARS-CoV-2 infection is essential for early interventions and patient stratification. Here, we investigated the potential of microRNAs (miRNAs) as theranostic agents for pulmoprotection in critical illness survivors. Multicenter study including 172 ICU survivors. Diffusion impairment was defined as a lung-diffusing capacity for carbon monoxide (DLCO) <80% within 12 months postdischarge. A disease-associated 16-miRNA panel was quantified in plasma samples collected at ICU admission. Bioinformatic analyses were conducted using KEGG, Reactome, GTEx, and Drug-Gene Interaction databases. The results were validated using an external RNA-seq dataset. A 3-miRNA signature linked to diffusion impairment (miR-27a-3p, miR-93-5p, and miR-199a-5p) was identified using random forest. Levels of miR-93-5p and miR-199a-5p were independently associated with the outcome, improving patient classification provided by the electronic health record. The experimentally validated targets of these miRNAs exhibited enrichment across diverse pathways, with telomere length quantification in an additional set of samples (n = 83) supporting the role of cell senescence in sequelae. Analysis of an external dataset refined the pathobiological fingerprint of pulmonary sequelae. Gene-drug interaction analysis revealed four FDA-approved drugs. Overall, this study advances our understanding of lung recovery in postacute respiratory infections, highlighting the potential of miRNAs and their targets for pulmoprotection.

5.
Sci Rep ; 14(1): 3799, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38360886

ABSTRACT

Ventricular tachyarrhythmia (VTA) are frequent arrhythmias in patients with hypertrophic cardiomyopathy (HCM). Representing a major risk factor for sudden cardiac death, Holter ECG at first clinical presentation appears insufficient. This study aims to investigate the ability of routinely obtained parameters associated with myocardial remodeling in stratifying for VTA in HCM. In this monocentric analysis, patients with HCM underwent 12-channel electrocardiography and echocardiography, including tissue doppler imaging. The study's primary endpoint was the documentation of non-sustained and sustained ventricular tachycardia-summarized as ventricular tachyarrhythmias (VTA) on Holter ECG or active devices. The occurrence of VTA was exploratory. Based on our collective, we developed a risk model regarding VTA. Of 140 HCM patients, 38 (27.1%) had an episode of VTA. Patients with VTA were likelier to have a history of atrial fibrillation (p < 0.001), a thicker interventricular septum (p < 0.001) and lower peak systolic mitral annular velocity (p < 0.001). The parameters were independently associated with endpoint in univariate and multivariate logistic regression. We created a logistic equation and calculated a cut-off value. The resulting ROC curve revealed a discriminative ability with AUC of 0.80 (sensitivity, 63%; specificity, 88%). Our risk model including these widely available parameters is able to distinguish low and high-risk of VTA in patients with HCM.


Subject(s)
Cardiomyopathy, Hypertrophic , Tachycardia, Ventricular , Humans , Pilot Projects , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/diagnostic imaging , Echocardiography/adverse effects , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/complications , Risk Factors , Risk Assessment , Death, Sudden, Cardiac/etiology
6.
Methods Mol Biol ; 2765: 247-260, 2024.
Article in English | MEDLINE | ID: mdl-38381344

ABSTRACT

This chapter serves as a guide for researchers embarking on circular RNA-based translational studies. It provides a foundation for the successful encapsulation of circular RNA into lipid nanoparticles (LNPs) and facilitates progress in this emerging field. Crucial scientific methods and techniques involved in the formulation process, particle characterization, and downstream processing of circ-LNPs are covered. The production of in vitro transcribed circular RNA-containing LNPs based on a commercially available lipid mix is provided, in addition to the fundamentals for successful encapsulation based on lipid mixes composed of single components. Furthermore, the transfection and validation protocols for the identification of a functional and potentially therapeutic circRNA candidate for initial in vitro verification, before subsequent LNP studies, are explained.

7.
J Transl Med ; 21(1): 742, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37864227

ABSTRACT

BACKGROUND: Patients with heart failure with reduced ejection fraction (HFrEF) and central sleep apnea (CSA) are at a very high risk of fatal outcomes. OBJECTIVE: To test whether the circulating miRNome provides additional information for risk stratification on top of clinical predictors in patients with HFrEF and CSA. METHODS: The study included patients with HFrEF and CSA from the SERVE-HF trial. A three-step protocol was applied: microRNA (miRNA) screening (n = 20), technical validation (n = 60), and biological validation (n = 587). The primary outcome was either death from any cause, lifesaving cardiovascular intervention, or unplanned hospitalization for worsening of heart failure, whatever occurred first. MiRNA quantification was performed in plasma samples using miRNA sequencing and RT-qPCR. RESULTS: Circulating miR-133a-3p levels were inversely associated with the primary study outcome. Nonetheless, miR-133a-3p did not improve a previously established clinical prognostic model in terms of discrimination or reclassification. A customized regression tree model constructed using the Classification and Regression Tree (CART) algorithm identified eight patient subphenotypes with specific risk patterns based on clinical and molecular characteristics. MiR-133a-3p entered the regression tree defining the group at the lowest risk; patients with log(NT-proBNP) ≤ 6 pg/mL (miR-133a-3p levels above 1.5 arbitrary units). The overall predictive capacity of suffering the event was highly stable over the follow-up (from 0.735 to 0.767). CONCLUSIONS: The combination of clinical information, circulating miRNAs, and decision tree learning allows the identification of specific risk subphenotypes in patients with HFrEF and CSA.


Subject(s)
Heart Failure , MicroRNAs , Sleep Apnea, Central , Ventricular Dysfunction, Left , Humans , Sleep Apnea, Central/complications , Biomarkers , Stroke Volume , MicroRNAs/genetics , Decision Trees
8.
Science ; 381(6660): 897-906, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37616346

ABSTRACT

Aging is a major risk factor for impaired cardiovascular health. Because the aging myocardium is characterized by microcirculatory dysfunction, and because nerves align with vessels, we assessed the impact of aging on the cardiac neurovascular interface. We report that aging reduces nerve density in the ventricle and dysregulates vascular-derived neuroregulatory genes. Aging down-regulates microRNA 145 (miR-145) and derepresses the neurorepulsive factor semaphorin-3A. miR-145 deletion, which increased Sema3a expression or endothelial Sema3a overexpression, reduced axon density, mimicking the aged-heart phenotype. Removal of senescent cells, which accumulated with chronological age in parallel to the decline in nerve density, rescued age-induced denervation, reversed Sema3a expression, preserved heart rate patterns, and reduced electrical instability. These data suggest that senescence-mediated regulation of nerve density contributes to age-associated cardiac dysfunction.


Subject(s)
Aging , Cellular Senescence , Heart , MicroRNAs , Microvascular Density , Myocardium , Semaphorin-3A , Heart/innervation , Microcirculation , MicroRNAs/genetics , MicroRNAs/metabolism , Semaphorin-3A/genetics , Animals , Mice , Aging/genetics , Aging/pathology , Male , Mice, Inbred C57BL , Cellular Senescence/genetics , Myocardium/pathology , Axons
9.
Cardiovasc Res ; 119(10): 1969-1980, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37315201

ABSTRACT

AIMS: Heart failure (HF) after myocardial infarction (MI) is a major cause of morbidity and mortality. We sought to investigate the functional importance of cardiac iron status after MI and the potential of pre-emptive iron supplementation in preventing cardiac iron deficiency (ID) and attenuating left ventricular (LV) remodelling. METHODS AND RESULTS: MI was induced in C57BL/6J male mice by left anterior descending coronary artery ligation. Cardiac iron status in the non-infarcted LV myocardium was dynamically regulated after MI: non-haem iron and ferritin increased at 4 weeks but decreased at 24 weeks after MI. Cardiac ID at 24 weeks was associated with reduced expression of iron-dependent electron transport chain (ETC) Complex I compared with sham-operated mice. Hepcidin expression in the non-infarcted LV myocardium was elevated at 4 weeks and suppressed at 24 weeks. Hepcidin suppression at 24 weeks was accompanied by more abundant expression of membrane-localized ferroportin, the iron exporter, in the non-infarcted LV myocardium. Notably, similarly dysregulated iron homeostasis was observed in LV myocardium from failing human hearts, which displayed lower iron content, reduced hepcidin expression, and increased membrane-bound ferroportin. Injecting ferric carboxymaltose (15 µg/g body weight) intravenously at 12, 16, and 20 weeks after MI preserved cardiac iron content and attenuated LV remodelling and dysfunction at 24 weeks compared with saline-injected mice. CONCLUSION: We demonstrate, for the first time, that dynamic changes in cardiac iron status after MI are associated with local hepcidin suppression, leading to cardiac ID long term after MI. Pre-emptive iron supplementation maintained cardiac iron content and attenuated adverse remodelling after MI. Our results identify the spontaneous development of cardiac ID as a novel disease mechanism and therapeutic target in post-infarction LV remodelling and HF.


Subject(s)
Heart Failure , Iron Deficiencies , Myocardial Infarction , Male , Mice , Humans , Animals , Hepcidins/metabolism , Hepcidins/therapeutic use , Iron/metabolism , Iron/therapeutic use , Mice, Inbred C57BL , Myocardium/metabolism , Heart Failure/metabolism , Dietary Supplements , Ventricular Remodeling
10.
Biomolecules ; 13(6)2023 06 15.
Article in English | MEDLINE | ID: mdl-37371575

ABSTRACT

Takotsubo syndrome (TTS), an acute cardiac condition characterized by transient wall motion abnormalities mostly of the left ventricle, results in difficulties in diagnosing patients. We set out to present a detailed blood analysis of TTS patients analyzing novel markers to understand the development of TTS. Significant differences in proinflammatory cytokine expression patterns and sex steroid and glucocorticoid receptor (GR) expression levels were observed in the TTS patient collected. Remarkably, the measured catecholamine serum concentrations determined from TTS patient blood could be shown to be two orders of magnitude lower than the levels determined from experimentally induced TTS in laboratory animals. Consequently, the exposure of endothelial cells and cardiomyocytes in vitro to such catecholamine concentrations did not damage the cellular integrity or function of either endothelial cells forming the blood-brain barrier, endothelial cells derived from myocardium, or cardiomyocytes in vitro. Computational analysis was able to link the identified blood markers, specifically, the proinflammatory cytokines and glucocorticoid receptor GR to microRNA (miR) relevant in the ontogeny of TTS (miR-15) and inflammation (miR-21, miR-146a), respectively. Amongst the well-described risk factors of TTS (older age, female sex), inflammaging-related pathways were identified to add to these relevant risk factors or prediagnostic markers of TTS.


Subject(s)
MicroRNAs , Takotsubo Cardiomyopathy , Vascular Diseases , Animals , Female , Takotsubo Cardiomyopathy/diagnosis , Endothelial Cells , Receptors, Glucocorticoid , Myocytes, Cardiac , MicroRNAs/genetics , Biomarkers , Catecholamines
11.
J Sport Health Sci ; 12(4): 438-463, 2023 07.
Article in English | MEDLINE | ID: mdl-36868374

ABSTRACT

Coronaviruses are pathogens thought to primarily affect the respiratory tracts of humans. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 was also marked mainly by its symptoms of respiratory illness, which were named coronavirus disease 2019 (COVID-19). Since its initial discovery, many other symptoms have been linked to acute SARS-CoV-2 infections as well as to the long-term outcomes of COVID-19 patients. Among these symptoms are different categories of cardiovascular diseases (CVDs), which continue to be the main cause of death worldwide. The World Health Organization estimates that 17.9 million people die from CVDs each year, accounting for ∼32% of all deaths globally. Physical inactivity is one of the most important behavioral risk factors for CVDs. The COVID-19 pandemic has affected CVDs as well as the physical activity in different ways. Here, we provide an overview of the current status as well as future challenges and possible solutions.


Subject(s)
COVID-19 , Middle East Respiratory Syndrome Coronavirus , Humans , COVID-19/epidemiology , SARS-CoV-2 , Pandemics , Communicable Disease Control
12.
J Appl Physiol (1985) ; 134(4): 799-809, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36759165

ABSTRACT

Electromyostimulation (EMS) is used to maintain or build skeletal muscle and to increase cardiopulmonary fitness. Only limited data on the molecular mechanisms induced by EMS are available and effects on circulating microRNAs (c-miRNAs) have not been reported. This study aimed to evaluate whether EMS induces long-term changes in muscle- and cardiovascular-specific c-miRNA levels. Twelve healthy participants (33.0 ± 12.0 yr, 7 women) performed a 20-min whole body EMS training and a time- and intensity-matched whole body circuit training (CT) in random order. Blood samples were drawn pre-/posttraining and at 1.5, 3, 24, 48, and 72 h to determine creatine kinase (CK) and miRNA-21-5p, -126-3p, -133a-3p, -146a-5p, -206-3p, -222-3p, and -499a-5p levels. Muscular exertion was determined using an isometric strength test, and muscle soreness/pain was assessed by questionnaire. EMS participants reported higher muscle soreness 48 and 72 h postexercise and mean CK levels after EMS increased compared with CT at 48 and 72 h (time × group P ≤ 0.01). The EMS session induced a significant elevation of myomiR-206 and -133a levels starting at 1.5 and 3 h after exercise. Both miRNAs remained elevated for 72 h with significant differences between 24 and 72 h (time × group P ≤ 0.0254). EMS did not induce changes in cardiovascular miRNAs and no elevation in any miRNA was detected following CT. Time-course analysis of muscle damage marker CK and c-miR-133a and -206 levels did not suggest a common scheme (P ≥ 0.277). We conclude that a single EMS session induces specific long-lasting changes of miR-206 and miR-133 involved in muscle proliferation and differentiation. A single EMS session does not affect primary cardiovascular miRNA-21-5p, -126-3p, -146a-5p, and -222-3p levels.NEW & NOTEWORTHY Our study describes the long-term effects of electromyostimulation (EMS) on circulating miRNA levels. The observed increase of functional myomiR-206 and -133a levels over 72 h suggests long-lasting effects on muscle proliferation and differentiation, whereas cardiovascular miRNAs appear unaffected. Our findings suggest that circulating miRNAs provide useful insight into muscle regeneration processes after EMS and may thus be used to optimize EMS training effects.


Subject(s)
MicroRNAs , Humans , Female , MicroRNAs/genetics , Myalgia , Cross-Over Studies , Muscle, Skeletal , Exercise/physiology
13.
Cardiovasc Res ; 119(7): 1495-1508, 2023 07 04.
Article in English | MEDLINE | ID: mdl-36651915

ABSTRACT

Considerable progress has been made in managing cancer; however, with these advancements comes the discovery of previously unknown adverse events. In particular, the prolonged lifespan of patients has uncovered severe cardiotoxic side effects of widely used anti-cancer therapies, which restrict their administration and thus compromise the success of the seemingly most suitable treatments in large cancer patient cohorts. Vice versa, cardiovascular diseases can also promote both the onset and progression of different cancers, highlighting that both conditions are deeply interlinked. Recognizing these close interactions, the novel interdisciplinary field of cardio-oncology has emerged to closely study these uniquely correlating diseases. In this regard, non-coding RNAs (ncRNAs) are gaining increasing attention since they constitute crucial regulators in many physiological but also pathological signalling pathways, including those of cancer and cardiac dysfunction. In this review, we focus on the new subtype of ncRNA, circular RNAs, in their distinct exchange within cardio-oncology and discuss their suitability as potent targets for the simultaneous treatment of cardiac dysfunction and cancer.


Subject(s)
Cardiovascular Diseases , Heart Diseases , Neoplasms , Humans , RNA, Circular/genetics , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/therapy , Neoplasms/drug therapy , Neoplasms/genetics , Heart , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/genetics , RNA, Untranslated/genetics , RNA, Untranslated/therapeutic use
14.
Adv Exp Med Biol ; 1396: 235-254, 2023.
Article in English | MEDLINE | ID: mdl-36454471

ABSTRACT

Cardiovascular diseases (CVDs) are the leading causes of death globally and urgently require new novel therapeutic strategies. Gene therapy is the application of gene modulation technology to treat abnormal gene expression under disease conditions. Viral- and nonviral-based gene delivery systems are the foundation of gene modulation in target cells. Moreover, plasmid- or oligo-based gene modulation tools as well as new advancements in gene editing using CRISPR/Cas technology are currently being tested in a variety of clinical trials. Here, we summarized state-of-the-art gene therapy technologies as well as recent clinical trials and discuss the applications and lessons of gene therapy in CVDs.


Subject(s)
Cardiovascular Diseases , Humans , Cardiovascular Diseases/genetics , Cardiovascular Diseases/therapy , Genetic Therapy , Gene Editing , Cloning, Molecular , Erythrocyte Count
15.
Mol Ther ; 30(12): 3601-3618, 2022 12 07.
Article in English | MEDLINE | ID: mdl-35810332

ABSTRACT

AAV vectors are promising delivery tools for human gene therapy. However, broad tissue tropism and pre-existing immunity against natural serotypes limit their clinical use. We identified two AAV capsid variants, AAV2-THGTPAD and AAV2-NLPGSGD, by in vivo AAV2 peptide display library screening in a murine model of pressure overload-induced cardiac hypertrophy. Both variants showed significantly improved efficacy in in vivo cardiomyocyte transduction compared with the parental serotype AAV2 as indicated by a higher number of AAV vector episomes in the nucleus and significant improved transduction efficiency. Both variants also outcompeted the reference serotype AAV9 regarding cardiomyocyte tropism, reaching comparable cardiac transduction efficiencies accompanied with liver de-targeting and decreased transduction efficiency of non-cardiac cells. Capsid modification influenced immunogenicity as sera of mice treated with AAV2-THGTPAD and AAV2-NLPGSGD demonstrated a poor neutralization capacity for the parental serotype and the novel variants. In a therapeutic setting, using the long non-coding RNA H19 in low vector dose conditions, novel AAV variants mediated superior anti-hypertrophic effects and revealed a further improved target-to-noise ratio, i.e., cardiomyocyte tropism. In conclusion, AAV2-THGTPAD and AAV2-NLPGSGD are promising novel tools for cardiac-directed gene therapy outperforming AAV9 regarding the specificity and therapeutic efficiency of in vivo cardiomyocyte transduction.


Subject(s)
Myocytes, Cardiac , RNA, Long Noncoding , Animals , Humans , Mice , Tropism , Capsid
16.
Eur J Heart Fail ; 24(10): 1778-1791, 2022 10.
Article in English | MEDLINE | ID: mdl-35867781

ABSTRACT

In vitro modelling the complex (patho-) physiological conditions of the heart is a major challenge in cardiovascular research. In recent years, methods based on three-dimensional (3D) cultivation approaches have steadily evolved to overcome the major limitations of conventional adherent two-dimensional (2D) monolayer cultivation. These 3D approaches aim to study, reproduce or modify fundamental native features of the heart such as tissue organization and cardiovascular microenvironment. Therefore, these systems have great potential for (patient-specific) disease research, for the development of new drug screening platforms, and for the use in regenerative and replacement therapy applications. Consequently, continuous improvement and adaptation is required with respect to fundamental limitations such as cardiomyocyte maturation, scalability, heterogeneity, vascularization, and reproduction of native properties. In this review, 2D monolayer culturing and the 3D in vitro systems of cardiac spheroids, organoids, engineered cardiac microtissue and bioprinting as well as the ex vivo technique of myocardial slicing are introduced with their basic concepts, advantages, and limitations. Furthermore, recent advances of various new approaches aiming to extend as well as to optimize these in vitro and ex vivo systems are presented.


Subject(s)
Bioprinting , Heart Failure , Humans , Bioprinting/methods , Organoids , Myocardium , Myocytes, Cardiac , Tissue Engineering/methods
17.
Eur Heart J ; 43(42): 4496-4511, 2022 11 07.
Article in English | MEDLINE | ID: mdl-35758064

ABSTRACT

AIMS: Cardiotoxicity leading to heart failure (HF) is a growing problem in many cancer survivors. As specific treatment strategies are not available, RNA discovery pipelines were employed and a new and powerful circular RNA (circRNA)-based therapy was developed for the treatment of doxorubicin-induced HF. METHODS AND RESULTS: The circRNA sequencing was applied and the highly species-conserved circRNA insulin receptor (Circ-INSR) was identified, which participates in HF processes, including those provoked by cardiotoxic anti-cancer treatments. Chemotherapy-provoked cardiotoxicity leads to the down-regulation of Circ-INSR in rodents and patients, which mechanistically contributes to cardiomyocyte cell death, cardiac dysfunction, and mitochondrial damage. In contrast, Circ-INSR overexpression prevented doxorubicin-mediated cardiotoxicity in both rodent and human cardiomyocytes in vitro and in a mouse model of chronic doxorubicin cardiotoxicity. Breast cancer type 1 susceptibility protein (Brca1) was identified as a regulator of Circ-INSR expression. Detailed transcriptomic and proteomic analyses revealed that Circ-INSR regulates apoptotic and metabolic pathways in cardiomyocytes. Circ-INSR physically interacts with the single-stranded DNA-binding protein (SSBP1) mediating its cardioprotective effects under doxorubicin stress. Importantly, in vitro transcribed and circularized Circ-INSR mimics also protected against doxorubicin-induced cardiotoxicity. CONCLUSION: Circ-INSR is a highly conserved non-coding RNA which is down-regulated during cardiotoxicity and cardiac remodelling. Adeno-associated virus and circRNA mimics-based Circ-INSR overexpression prevent and reverse doxorubicin-mediated cardiomyocyte death and improve cardiac function. The results of this study highlight a novel and translationally important Circ-INSR-based therapeutic approach for doxorubicin-induced cardiac dysfunction.


Subject(s)
Cardiotoxicity , Heart Diseases , Mice , Animals , Humans , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , RNA, Circular/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptor, Insulin/pharmacology , Proteomics , Apoptosis , Doxorubicin/toxicity , Myocytes, Cardiac/metabolism , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/prevention & control , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Mitochondrial Proteins
18.
Basic Res Cardiol ; 117(1): 26, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35503383

ABSTRACT

Myocardial injury often leads to heart failure due to the loss and insufficient regeneration of resident cardiomyocytes. The low regenerative potential of the mammalian heart is one of the main drivers of heart failure progression, especially after myocardial infarction accompanied by large contractile muscle loss. Preclinical therapies for cardiac regeneration are promising, but clinically still missing. Mammalian models represent an excellent translational in vivo platform to test drugs and treatments for the promotion of cardiac regeneration. Particularly, short-lived mice offer the possibility to monitor the outcome of such treatments throughout the life span. Importantly, there is a short period of time in newborn mice in which the heart retains full regenerative capacity after cardiac injury, which potentially also holds true for the neonatal human heart. Thus, in vivo neonatal mouse models of cardiac injury are crucial to gain insights into the molecular mechanisms underlying the cardiac regenerative processes and to devise novel therapeutic strategies for the treatment of diseased adult hearts. Here, we provide an overview of the established injury models to study cardiac regeneration. We summarize pioneering studies that demonstrate the potential of using neonatal cardiac injury models to identify factors that may stimulate heart regeneration by inducing endogenous cardiomyocyte proliferation in the adult heart. To conclude, we briefly summarize studies in large animal models and the insights gained in humans, which may pave the way toward the development of novel approaches in regenerative medicine.


Subject(s)
Heart Failure , Myocardial Infarction , Animals , Animals, Newborn , Cell Proliferation , Heart/physiology , Mammals , Mice , Myocytes, Cardiac/physiology , Regeneration/physiology
19.
Mol Ther ; 30(4): 1675-1691, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35077859

ABSTRACT

Exercise and its regulated molecules have myocardial protective effects against cardiac ischemia/reperfusion (I/R) injury. The muscle-enriched miR-486 was previously identified to be upregulated in the exercised heart, which prompted us to investigate the functional roles of miR-486 in cardiac I/R injury and to further explore its potential in contributing to exercise-induced protection against I/R injury. Our data showed that miR-486 was significantly downregulated in the heart upon cardiac I/R injury. Both preventive and therapeutic interventions of adeno-associated virus 9 (AAV9)-mediated miR-486 overexpression could reduce cardiac I/R injury. Using AAV9 expressing miR-486 with a cTnT promoter, we further demonstrated that cardiac muscle cell-targeted miR-486 overexpression was also sufficient to protect against cardiac I/R injury. Consistently, miR-486 was downregulated in oxygen-glucose deprivation/reperfusion (OGDR)-stressed cardiomyocytes, while upregulating miR-486 inhibited cardiomyocyte apoptosis through PTEN and FoxO1 inhibition and AKT/mTOR activation. Finally, we observed that miR-486 was necessary for exercise-induced protection against cardiac I/R injury. In conclusion, miR-486 is protective against cardiac I/R injury and myocardial apoptosis through targeting of PTEN and FoxO1 and activation of the AKT/mTOR pathway, and mediates the beneficial effect of exercise for myocardial protection. Increasing miR-486 might be a promising therapeutic strategy for myocardial protection.


Subject(s)
MicroRNAs , Myocardial Reperfusion Injury , Apoptosis/genetics , Humans , Ischemia/metabolism , MicroRNAs/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
20.
Cardiovasc Res ; 118(15): 3071-3084, 2022 12 09.
Article in English | MEDLINE | ID: mdl-34718448

ABSTRACT

Myocardial infarction causes a massive loss of cardiomyocytes (CMs), which can lead to heart failure accompanied by fibrosis, stiffening of the heart, and loss of function. Heart failure causes high mortality rates and is a huge socioeconomic burden, which, based on diets and lifestyle in the developed world, is expected to increase further in the next years. At present, the only curative treatment for heart failure is heart transplantation associated with a number of limitations such as donor organ availability and transplant rejection among others. Thus, the development of cellular reprogramming and defined differentiation protocols provide exciting new possibilities for cell therapy approaches and which opened up a new era in regenerative medicine. Consequently, tremendous research efforts were undertaken to gain a detailed molecular understanding of the reprogramming processes and the in vitro differentiation of pluripotent stem cells into functional CMs for transplantation into the patient's injured heart. In the last decade, non-coding RNAs, particularly microRNAs, long non-coding RNAs, and circular RNAs emerged as critical regulators of gene expression that were shown to fine-tune cellular processes both on the transcriptional and the post-transcriptional level. Unsurprisingly, also cellular reprogramming, pluripotency, and cardiac differentiation and maturation are regulated by non-coding RNAs. In here, we review the current knowledge on non-coding RNAs in these processes and highlight how their modulation may enhance the quality and quantity of stem cells and their derivatives for safe and efficient clinical application in patients with heart failure. In addition, we summarize the clinical cell therapy efforts undertaken thus far.


Subject(s)
Heart Failure , Regenerative Medicine , Humans , Heart Failure/genetics , Heart Failure/therapy , RNA, Long Noncoding
SELECTION OF CITATIONS
SEARCH DETAIL
...