Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Stem Cell Res ; 74: 103261, 2024 02.
Article in English | MEDLINE | ID: mdl-38100916

ABSTRACT

The identification of TBX5-related regulatory sequences in genes essential for heart development is hampered by the absence of antibodies which allow precipitation of TBX5:DNA complexes. Employing CRISPR/Cas9 technology, we have inserted a FLAG-tag sequence at the end of exon 9 of the TBX5 gene prior to the stop codon by homologous recombination. The translated TBX5-FLAG fusion protein of the three iPSC lines can effectively be precipitated by anti-FLAG antibodies and, thus, allow the detection of specific TBX5-binding sites and their associated genes.


Subject(s)
Induced Pluripotent Stem Cells , Induced Pluripotent Stem Cells/metabolism , CRISPR-Cas Systems/genetics , Homologous Recombination , Exons/genetics
2.
Stem Cell Res ; 70: 103126, 2023 08.
Article in English | MEDLINE | ID: mdl-37253295

ABSTRACT

TBX5 is a transcription factor which plays an essential role at different checkpoints during cardiac differentiation. However, regulatory pathways affected by TBX5 still remain ill-defined. We have applied the CRISPR/Cas9 technology using a completely plasmid-free approach to correct a heterozygous causative "loss-of function" TBX5 mutation in an iPSC line (DHMi004-A), that has been established from a patient suffering from Holt-Oram syndrome (HOS). This isogenic iPSC line, DHMi004-A-1, represents a powerful in vitro tool to dissect the regulatory pathways affected by TBX5 in HOS.

3.
J Card Surg ; 37(10): 2999-3005, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35790024

ABSTRACT

BACKGROUND: Isolated tricuspid valve endocarditis (TVE) is a rare disease which is managed medically in most patients. Only in specific cases, surgical intervention becomes necessary. Hence, data about surgical outcomes are sparse. This study reports on the operative experience in patients with isolated TVE over a period of 20 years. METHODS: We retrospectively analyzed 32 patients with isolated TVE who underwent surgery from February 2001 to June 2021 at the German Heart Centre Munich. RESULTS: Thirty-day mortality was 3.1%. Overall survival was 89.9± 5.5% at 1 year and 76.9 ± 8.5% at 5 years. Cumulative incidence for reoperation was 11.1 ± 6.0% at 5 years. Four patients (12.5%) were treated for recurrent endocarditis. Tricuspid valve repair (TVr) was achieved in 16 patients (50%). If the subvalvular apparatus (n = 10) was involved, tricuspid valve replacement was performed more frequently. CONCLUSIONS: Mortality in patients with isolated TVE undergoing cardiac surgery is high. In half of the cases, TVr was achieved but was less likely in patients with affected subvalvular apparatus.


Subject(s)
Endocarditis , Heart Valve Prosthesis Implantation , Tricuspid Valve Insufficiency , Endocarditis/surgery , Humans , Retrospective Studies , Treatment Outcome , Tricuspid Valve/surgery , Tricuspid Valve Insufficiency/surgery
4.
Stem Cell Res ; 60: 102691, 2022 04.
Article in English | MEDLINE | ID: mdl-35121196

ABSTRACT

A number of mutations in the human TBX5 gene have been described which cause Holt-Oram syndrome, a severe congenital disease associated with abnormalities in heart and upper limb development. We have used a prime-editing approach to introduce a patient-specific disease-causing TBX5 mutation (c.920_C > A) into an induced pluripotent stem cell (iPSC) line from a healthy donor. The resulting iPSC line provides a powerful tool to identify and analyze the biological and molecular impact of this specific TBX5 mutation in comparison to the isogenic control iPSC line during cardiac development.


Subject(s)
Induced Pluripotent Stem Cells , T-Box Domain Proteins/genetics , Upper Extremity Deformities, Congenital , CRISPR-Cas Systems/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Point Mutation , T-Box Domain Proteins/metabolism , Upper Extremity Deformities, Congenital/genetics
5.
J Clin Med ; 10(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34768606

ABSTRACT

BACKGROUND: Tricuspid valve (TV) repair is the recommended treatment for severe functional tricuspid regurgitation (fTR) in patients undergoing left-sided surgery. For this purpose, a wide range of annuloplasty devices differing in form and flexibility are available. This study reports the results using a three-dimensional annuloplasty ring (Medtronic, Contour 3D Ring) for TV repair and analysis of risk factors. METHODS: A cohort of 468 patients who underwent TV repair (TVr) with a concomitant cardiac procedure from December 2010 to January 2017 was retrospectively analyzed. RESULTS: At follow-up, 96.1% of patients had no/trivial or mild TR. The 30-day mortality was 4.7%; it significantly differed between electively performed operations (2.7%) and urgent/emergent operations (11.7%). Risk factors for recurrent moderate and severe TR were LVEF < 50%, TAPSE < 16 mm, and moderate mitral valve (MV) regurgitation at follow-up. Preoperatively reduced renal function lead to a higher 30-day and overall mortality. Reoperation of the TV was required in six patients (1.6%). Risk factors for TV related reoperations were preoperative TV annulus over 50 mm and an implanted permanent pacemaker. CONCLUSIONS: TVr with the Contour 3D annuloplasty ring shows low TR recurrence and reoperation rates. Risk-factor analysis for the recurrence of TR revealed the importance of left- and right-ventricular function.

6.
Sci Rep ; 11(1): 3026, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542270

ABSTRACT

Generating cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) has represented a significant advance in our ability to model cardiac disease. Current differentiation protocols, however, have limited use due to their production of heterogenous cell populations, primarily consisting of ventricular-like CMs. Here we describe the creation of two chamber-specific reporter hiPSC lines by site-directed genomic integration using CRISPR-Cas9 technology. In the MYL2-tdTomato reporter, the red fluorescent tdTomato was inserted upstream of the 3' untranslated region of the Myosin Light Chain 2 (MYL2) gene in order faithfully label hiPSC-derived ventricular-like CMs while avoiding disruption of endogenous gene expression. Similarly, in the SLN-CFP reporter, Cyan Fluorescent Protein (CFP) was integrated downstream of the coding region of the atrial-specific gene, Sarcolipin (SLN). Purification of tdTomato+ and CFP+ CMs using flow cytometry coupled with transcriptional and functional characterization validated these genetic tools for their use in the isolation of bona fide ventricular-like and atrial-like CMs, respectively. Finally, we successfully generated a double reporter system allowing for the isolation of both ventricular and atrial CM subtypes within a single hiPSC line. These tools provide a platform for chamber-specific hiPSC-derived CM purification and analysis in the context of atrial- or ventricular-specific disease and therapeutic opportunities.


Subject(s)
Cell Differentiation/genetics , Heart Atria/growth & development , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , CRISPR-Cas Systems/genetics , Cardiac Myosins/genetics , Green Fluorescent Proteins , Heart Atria/cytology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/growth & development , Heart Ventricles/metabolism , Humans , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/cytology , Myosin Light Chains/genetics
7.
J Clin Invest ; 131(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33201861

ABSTRACT

Genetic factors undoubtedly affect the development of congenital heart disease (CHD) but still remain ill defined. We sought to identify genetic risk factors associated with CHD and to accomplish a functional analysis of SNP-carrying genes. We performed a genome-wide association study (GWAS) of 4034 White patients with CHD and 8486 healthy controls. One SNP on chromosome 5q22.2 reached genome-wide significance across all CHD phenotypes and was also indicative for septal defects. One region on chromosome 20p12.1 pointing to the MACROD2 locus identified 4 highly significant SNPs in patients with transposition of the great arteries (TGA). Three highly significant risk variants on chromosome 17q21.32 within the GOSR2 locus were detected in patients with anomalies of thoracic arteries and veins (ATAV). Genetic variants associated with ATAV are suggested to influence the expression of WNT3, and the variant rs870142 related to septal defects is proposed to influence the expression of MSX1. We analyzed the expression of all 4 genes during cardiac differentiation of human and murine induced pluripotent stem cells in vitro and by single-cell RNA-Seq analyses of developing murine and human hearts. Our data show that MACROD2, GOSR2, WNT3, and MSX1 play an essential functional role in heart development at the embryonic and newborn stages.


Subject(s)
Genetic Loci , Heart Defects, Congenital/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Animals , Female , Genome-Wide Association Study , Germany/epidemiology , Heart Defects, Congenital/epidemiology , Humans , Male , Mice , Risk Factors
8.
Stem Cells Int ; 2018: 4136473, 2018.
Article in English | MEDLINE | ID: mdl-29731778

ABSTRACT

Genome editing is a powerful tool to study the function of specific genes and proteins important for development or disease. Recent technologies, especially CRISPR/Cas9 which is characterized by convenient handling and high precision, revolutionized the field of genome editing. Such tools have enormous potential for basic science as well as for regenerative medicine. Nevertheless, there are still several hurdles that have to be overcome, but patient-tailored therapies, termed precision medicine, seem to be within reach. In this review, we focus on the achievements and limitations of genome editing in the cardiovascular field. We explore different areas of cardiac research and highlight the most important developments: (1) the potential of genome editing in human pluripotent stem cells in basic research for disease modelling, drug screening, or reprogramming approaches and (2) the potential and remaining challenges of genome editing for regenerative therapies. Finally, we discuss social and ethical implications of these new technologies.

9.
Sci Rep ; 8(1): 6618, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29700394

ABSTRACT

Bioactive lipids such as sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) regulate diverse processes including cell proliferation, differentiation, and migration. However, their roles in cardiac differentiation and cardiomyocyte proliferation have not been explored. Using a 96-well differentiation platform for generating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) we found that S1P and LPA can independently enhance cardiomyocyte generation when administered at an early stage of differentiation. We showed that the combined S1P and LPA treatment of undifferentiated hiPSCs resulted in increased nuclear accumulation of ß-catenin, the canonical Wnt signaling pathway mediator, and synergized with CHIR99021, a glycogen synthase kinase 3 beta inhibitor, to enhance mesodermal induction and subsequent cardiac differentiation. At later stages of cardiac differentiation, the addition of S1P and LPA resulted in cell cycle initiation in hiPSC-CMs, an effect mediated through increased ERK signaling. Although the addition of S1P and LPA alone was insufficient to induce cell division, it was able to enhance ß-catenin-mediated hiPSC-CM proliferation. In summary, we demonstrated a developmental stage-specific effect of bioactive lipids to enhance hiPSC-CM differentiation and proliferation via modulating the effect of canonical Wnt/ß-catenin and ERK signaling. These findings may improve hiPSC-CM generation for cardiac disease modeling, precision medicine, and regenerative therapies.


Subject(s)
Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Lipids/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Biomarkers , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/metabolism , MAP Kinase Signaling System/drug effects , Mesoderm/cytology , Mesoderm/drug effects , Models, Biological , Myocytes, Cardiac/metabolism , Wnt Signaling Pathway/drug effects
10.
Cardiovasc Res ; 114(8): 1098-1114, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29579159

ABSTRACT

Aims: The contribution of resident stem or progenitor cells to cardiomyocyte renewal after injury in adult mammalian hearts remains a matter of considerable debate. We evaluated a cell population in the adult mouse heart induced by myocardial infarction (MI) and characterized by an activated Nkx2.5 enhancer element that is specific for multipotent cardiac progenitor cells (CPCs) during embryonic development. We hypothesized that these MI-induced cells (MICs) harbour cardiomyogenic properties similar to their embryonic counterparts. Methods and results: MICs reside in the heart and mainly localize to the infarction area and border zone. Interestingly, gene expression profiling of purified MICs 1 week after infarction revealed increased expression of stem cell markers and embryonic cardiac transcription factors (TFs) in these cells as compared to the non-mycoyte cell fraction of adult hearts. A subsequent global transcriptome comparison with embryonic CPCs and fibroblasts and in vitro culture of MICs unveiled that (myo-)fibroblastic features predominated and that cardiac TFs were only expressed at background levels. Conclusions: Adult injury-induced reactivation of a cardiac-specific Nkx2.5 enhancer element known to specifically mark myocardial progenitor cells during embryonic development does not reflect hypothesized embryonic cardiomyogenic properties. Our data suggest a decreasing plasticity of cardiac progenitor (-like) cell populations with increasing age. A re-expression of embryonic, stem or progenitor cell features in the adult heart must be interpreted very carefully with respect to the definition of cardiac resident progenitor cells. Albeit, the abundance of scar formation after cardiac injury suggests a potential to target predestinated activated profibrotic cells to push them towards cardiomyogenic differentiation to improve regeneration.


Subject(s)
Homeobox Protein Nkx-2.5/metabolism , Muscle Development , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Regeneration , Stem Cells/metabolism , Ventricular Remodeling , Animals , Cell Differentiation , Cell Plasticity , Cells, Cultured , Chromatin Assembly and Disassembly , Disease Models, Animal , Enhancer Elements, Genetic , Epigenesis, Genetic , Homeobox Protein Nkx-2.5/deficiency , Homeobox Protein Nkx-2.5/genetics , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Phenotype , Signal Transduction , Stem Cells/pathology , Time Factors , Transcriptome
11.
J Hypertens ; 36(4): 847-856, 2018 04.
Article in English | MEDLINE | ID: mdl-29283973

ABSTRACT

AIMS: Left ventricular (LV) hypertrophy is characterized by cardiomyocyte hypertrophy and interstitial fibrosis ultimately leading to increased myocardial stiffness and reduced contractility. There is substantial evidence that the altered expression of matrix metalloproteinases (MMP) and Tenascin-C (TN-C) are associated with the progression of adverse LV remodeling. However, the role of TN-C in the development of LV hypertrophy because of chronic pressure overload as well as the regulatory role of TN-C on MMPs remains unknown. METHODS AND RESULTS: In a knockout mouse model of TN-C, we investigated the effect of 10 weeks of pressure overload using transverse aortic constriction (TAC). Cardiac function was determined by magnetic resonance imaging. The expression of MMP-2 and MMP-9, CD147 as well as myocardial fibrosis were assessed by immunohistochemistry. The expression of TN-C was assessed by RT-qPCR and ELISA. TN-C knockout mice showed marked reduction in fibrosis (P < 0.001) and individual cardiomyocytes size (P < 0.01), in expression of MMP-2 (P < 0.05) and MMP-9 (P < 0.001) as well as preserved cardiac function (P < 0.01) in comparison with wild-type mice after 10 weeks of TAC. In addition, CD147 expression was markedly increased under pressure overload (P < 0.01), irrespectively of genotype. TN-C significantly increased the expression of the markers of hypertrophy such as ANP and BNP as well as MMP-2 in H9c2 cells (P < 0.05, respectively). CONCLUSION: Our results are pointed toward a novel signaling mechanism that contributes to LV remodeling via MMPs upregulation, cardiomyocyte hypertrophy as well as myocardial fibrosis by TN-C under chronic pressure overload.


Subject(s)
Hypertension/complications , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Myocardium/pathology , Tenascin/genetics , Tenascin/metabolism , Ventricular Remodeling/genetics , Animals , Basigin/genetics , Basigin/metabolism , Cardiac Output , Cell Line , Fibrosis , Genotype , Hypertrophy, Left Ventricular/physiopathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Signal Transduction , Ventricular Remodeling/physiology
12.
J Thorac Dis ; 9(Suppl 1): S9-S16, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28446964

ABSTRACT

As the basic unit of living organisms, each single cell has unique molecular signatures and functions. Our ability to uncover the transcriptional and epigenetic signature of single cells has been hampered by the lack of tools to explore this area of research. The advent of microfluidic single cell technology along with single cell genome-wide DNA amplification methods had greatly improved our understanding of the expression variation in single cells. Transcriptional expression profile by multiplex qPCR or genome-wide RNA sequencing has enabled us to examine genes expression in single cells in different tissues. With the new tools, the identification of new cellular heterogeneity, novel marker genes, unique subpopulations, and spatial locations of each single cell can be acquired successfully. Epigenetic modifications for each single cell can also be obtained via similar methods. Based on single cell genome sequencing, single cell epigenetic information including histone modifications, DNA methylation, and chromatin accessibility have been explored and provided valuable insights regarding gene regulation and disease prognosis. In this article, we review the development of strategies to obtain single cell transcriptional and epigenetic data. Furthermore, we discuss ways in which single cell studies may help to provide greater understanding of the mechanisms of basic cardiovascular biology that will eventually lead to improvement in our ability to diagnose disease and develop new therapies.

14.
ESC Heart Fail ; 2(3): 171-177, 2015 Sep.
Article in English | MEDLINE | ID: mdl-28834679

ABSTRACT

AIMS: The interest in cardiac remodelling (REM) has steadily increased during recent years. The aim of this study was to functionally characterize REM following myocardial infarction (MI) in mice using high-end in vivo and ex vivo methods. METHODS AND RESULTS: Myocardial infarction or sham operation was induced in A/J mice. Six weeks later, mice underwent cardiac magnetic resonance imaging and were subsequently sacrificed for ex vivo measurements on the isolated heart. Thereafter, hearts were trichrome stained for infarction size calculation. Magnetic resonance imaging showed significantly reduced ejection fraction (P < 0.01) as well as increased end-systolic and end-diastolic volumes (P < 0.01) after MI. The mean infarct size was 48.8 ± 6.9% of left ventricle. In the isolated working heart coronary flow (time point 20': 6.6 ± 0.9 vs. 13.9 ± 1.6 mL/min, P < 0.01), cardiac output (time point 20': 17.5 ± 2.6 vs. 36.1 ± 4.3 mL/min, P < 0.01) and pump function (80 mmHg: 2.15 ± 0.88 vs. 4.83 ± 0.76, P < 0.05) were significantly attenuated in MI hearts during all measurements. Systolic and diastolic wall stress were significantly elevated in MI animals. CONCLUSION: This two-step approach is reasonable, since data quality increases while animals are not exposed to major additional interventions. Both the working heart and magnetic resonance imaging offer a reliable characterization of the functional changes that go along with the development of post-MI REM. By combining these two techniques, additional information such as wall stress can be evaluated.

15.
Interact Cardiovasc Thorac Surg ; 20(3): 387-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25468794

ABSTRACT

OBJECTIVES: Currently available cardioplegic solutions provide excellent protection in patients with normal surgical risk; in high-risk patients, however, such as in emergency coronary artery bypass surgery, there is still room for improvement. As most of the cardioplegic solutions primarily protect myocytes, the addition of substances for protection of the endothelium might improve their protective potential. The nitric oxide donor, S-nitroso human serum albumin (S-NO-HSA), which has been shown to prevent endothelial nitric oxide synthase uncoupling, was added to the newly developed histidine-tryptophan-ketoglutarat (HTK-N) cardioplegia in an isolated heart perfusion system after subjecting rats to acute myocardial infarction (MI) and reperfusion. METHODS: In male Sprague-Dawley rats, acute MI was induced by ligation for 1 h of the anterior descending coronary artery. After 2 h of in vivo reperfusion hearts were evaluated on an isolated erythrocyte-perfused working heart model. Cold ischaemia (4°C) for 60 min was followed by 45 min of reperfusion. Cardiac arrest was induced either with HTK (n = 10), HTK-N (n = 10) or HTK-N + S-NO-HSA (n = 10). In one group (HTK-N + S-NO-HSA plus in vivo S-NO-HSA; n = 9) an additional in vivo infusion of S-NO-HSA was performed. RESULTS: Post-ischaemic recovery of cardiac output (HTK: 77 ± 4%, HTK-N: 86 ± 7%, HTK-N + S-NO-HSA: 101 ± 5%, in vivo S-NO-HSA: 93 ± 8%), external heart work (HTK: 79 ± 5%, HTK-N: 83 ± 3%, HTK-N + S-NO-HSA: 101 ± 8%, in vivo S-NO-HSA: 109 ± 13%), coronary flow (HTK: 77 ± 4%, HTK-N: 94 ± 6%, HTK-N + S-NO-HSA: 118 ± 15%, in vivo S-NO-HSA: 113 ± 3.17%) [HTK-N + S-NO-HSA vs HTK P < 0.001; HTK-N + S-NO-HSA vs HTK-N P < 0.05] and left atrial diastolic pressure (HTK: 122 ± 31%, HTK-N: 159 ± 43%, HTK-N + S-NO-HSA: 88 ± 30, in vivo S-NO-HSA: 62 ± 10%) [HTK-N + S-NO-HSA vs HTK P < 0.05; in vivo S-NO-HSA vs HTK-N P < 0.05] were significantly improved in both S-NO-HSA-treated groups compared with HTK and HTK-N, respectively. This was accompanied by better preservation of high-energy phosphates (adenosine triphosphate; energy charge) and ultrastructural integrity on transmission electron microscopy. However, no additional benefit of in vivo S-NO-HSA infusion was observed. CONCLUSIONS: Addition of the NO donor, S-NO-HSA refines the concept of HTK-N cardioplegia in improving post-ischaemic myocardial perfusion. HTK-N with S-NO-HSA is a possible therapeutic option for patients who have to be operated on for acute MI.


Subject(s)
Heart Arrest, Induced/methods , Heart Arrest/prevention & control , Myocardial Infarction/therapy , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Serum Albumin/pharmacology , Animals , Cardiac Output/drug effects , Cardiac Output/physiology , Cardioplegic Solutions/pharmacology , Disease Models, Animal , Glucose/pharmacology , Heart Arrest/etiology , Male , Mannitol/pharmacology , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Potassium Chloride/pharmacology , Procaine/pharmacology , Rats , Rats, Sprague-Dawley , Serum Albumin, Human , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL