43 results on '"I. Gruh"'
Search Results
2. 3rd EACTS Meeting on Cardiac and Pulmonary Regeneration Berlin-Brandenburgische Akademie, Berlin, Germany, 14–15 December 2012
- Author
-
A. Bader, A. Brodarac, R. Hetzer, A. Kurtz, C. Stamm, H. Baraki, G. Kensah, S. Asch, S. Rojas, A. Martens, I. Gruh, A. Haverich, I. Kutschka, L. Cortes-Dericks, L. Froment, G. Kocher, R. A. Schmid, E. Delyagina, A. Schade, D. Scharfenberg, A. Skorska, C. Lux, W. Li, G. Steinhoff, F. Drey, V. Lepperhof, K. Neef, A. Fatima, T. Wittwer, T. Wahlers, T. Saric, Y.- H. Choi, D. Fehrenbach, A. Lehner, F. Herrmann, T. Hollweck, S. Pfeifer, E. Wintermantel, R. Kozlik-Feldmann, C. Hagl, B. Akra, M. Gyongyosi, M. Zimmermann, N. Pavo, M. Mildner, M. Lichtenauer, G. Maurer, J. Ankersmit, S. Hacker, R. Mittermayr, T. Haider, S. Nickl, L. Beer, D. Lebherz-Eichinger, T. Schweiger, A. Mitterbauer, C. Keibl, G. Werba, M. Frey, H. J. Ankersmit, S. Herrmann, C. A. Lux, J. Holfeld, C. Tepekoylu, F.- S. Wang, R. Kozaryn, W. Schaden, M. Grimm, C.- J. Wang, A. Urbschat, K. Zacharowski, P. Paulus, M. J. Avaca, H. Kempf, D. Malan, P. Sasse, B. Fleischmann, J. Palecek, G. Drager, A. Kirschning, R. Zweigerdt, U. Martin, K. Katsirntaki, R. Haller, S. Ulrich, M. Sgodda, V. Puppe, J. Duerr, A. Schmiedl, M. Ochs, T. Cantz, M. Mall, C. Mauritz, A. R. Lara, J. Dahlmann, K. Schwanke, J. Hegermann, D. Skvorc, A. Gawol, A. Azizian, S. Wagner, A. Krause, C. Klopsch, R. Gaebel, A. Kaminski, B. Chichkov, S. Jockenhoevel, K. Klose, R. Roy, K.- S. Kang, K. Bieback, B. Nasseri, O. Polchynska, K. Kruttwig, C. Bruggemann, G. Xu, A. Baumgartner, M. Hasun, B. K. Podesser, M. Ludwig, A. Tolk, T. Noack, R. Margaryan, N. Assanta, A. Menciassi, S. Burchielli, M. Matteucci, V. Lionetti, C. Luchi, E. Cariati, F. Coceani, B. Murzi, S. V. Rojas, A. Rotarmel, B. A. Nasseri, W. Ebell, M. Dandel, M. Kukucka, R. Gebker, H. Mutlak, P. Ockelmann, S. Tacke, B. Scheller, A. Pereszlenyi, M. Meier, N. Schecker, C. Rathert, P. M. Becher, N. Drori-Carmi, N. Bercovich, E. Zahavi-Goldstein, M. Jack, N. Netzer, L. Pinzur, A. Chajut, C. Tschope, U. Ruch, B.- E. Strauer, G. Tiedemann, F. Schlegel, S. Dhein, O. Akhavuz, F. W. Mohr, P. M. Dohmen, A. Salameh, K. Oelmann, P. Kiefer, S. Merkert, C. Templin, M. Jara-Avaca, S. Muller, S. von Haehling, S. Slavic, C. Curato, W. Altarche-Xifro, T. Unger, J. Li, Y. Zhang, W. Z. Li, L. Ou, N. Ma, A. Haase, and R. Alt
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Abstracts ,business.industry ,Regeneration (biology) ,General surgery ,Physiology ,Medicine ,Surgery ,Cardiology and Cardiovascular Medicine ,business - Published
- 2013
3. Author Correction: A micro-LED array based platform for spatio-temporal optogenetic control of various cardiac models.
- Author
-
Junge S, Ricci Signorini ME, Al Masri M, Gülink J, Brüning H, Kasperek L, Szepes M, Bakar M, Gruh I, Heisterkamp A, and Torres-Mapa ML
- Published
- 2024
- Full Text
- View/download PDF
4. Protein-free media for cardiac differentiation of hPSCs in 2000 mL suspension culture.
- Author
-
Kriedemann N, Manstein F, Hernandez-Bautista CA, Ullmann K, Triebert W, Franke A, Mertens M, Stein ICAP, Leffler A, Witte M, Askurava T, Fricke V, Gruh I, Piep B, Kowalski K, Kraft T, and Zweigerdt R
- Subjects
- Humans, Cell Culture Techniques methods, Pluripotent Stem Cells cytology, Pluripotent Stem Cells metabolism, Pluripotent Stem Cells drug effects, Cells, Cultured, Cell Differentiation drug effects, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Myocytes, Cardiac drug effects, Culture Media chemistry, Culture Media pharmacology
- Abstract
Background: Commonly used media for the differentiation of human pluripotent stem cells into cardiomyocytes (hPSC-CMs) contain high concentrations of proteins, in particular albumin, which is prone to quality variations and presents a substantial cost factor, hampering the clinical translation of in vitro-generated cardiomyocytes for heart repair. To overcome these limitations, we have developed chemically defined, entirely protein-free media based on RPMI, supplemented with L-ascorbic acid 2-phosphate (AA-2P) and either the non-ionic surfactant Pluronic F-68 or a specific polyvinyl alcohol (PVA)., Methods and Results: Both media compositions enable the efficient, directed differentiation of embryonic and induced hPSCs, matching the cell yields and cardiomyocyte purity ranging from 85 to 99% achieved with the widely used protein-based CDM3 medium. The protein-free differentiation approach was readily up-scaled to a 2000 mL process scale in a fully controlled stirred tank bioreactor in suspension culture, producing > 1.3 × 10
9 cardiomyocytes in a single process run. Transcriptome analysis, flow cytometry, electrophysiology, and contractile force measurements revealed that the mass-produced cardiomyocytes differentiated in protein-free medium exhibit the expected ventricular-like properties equivalent to the well-established characteristics of CDM3-control cells., Conclusions: This study promotes the robustness and upscaling of the cardiomyogenic differentiation process, substantially reduces media costs, and provides an important step toward the clinical translation of hPSC-CMs for heart regeneration., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
5. Fabrication of heart tubes from iPSC derived cardiomyocytes and human fibrinogen by rotating mold technology.
- Author
-
Andrée B, Voß N, Kriedemann N, Triebert W, Teske J, Mertens M, Witte M, Szádocka S, Hilfiker A, Aper T, Gruh I, and Zweigerdt R
- Subjects
- Humans, Fibroblasts metabolism, Cell Differentiation, Cells, Cultured, Bioreactors, Fibrin metabolism, Fibrin chemistry, Tissue Scaffolds chemistry, Myocytes, Cardiac metabolism, Myocytes, Cardiac cytology, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Fibrinogen metabolism, Fibrinogen chemistry, Tissue Engineering methods
- Abstract
Due to its structural and functional complexity the heart imposes immense physical, physiological and electromechanical challenges on the engineering of a biological replacement. Therefore, to come closer to clinical translation, the development of a simpler biological assist device is requested. Here, we demonstrate the fabrication of tubular cardiac constructs with substantial dimensions of 6 cm in length and 11 mm in diameter by combining human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and human foreskin fibroblast (hFFs) in human fibrin employing a rotating mold technology. By centrifugal forces employed in the process a cell-dense layer was generated enabling a timely functional coupling of iPSC-CMs demonstrated by a transgenic calcium sensor, rhythmic tissue contractions, and responsiveness to electrical pacing. Adjusting the degree of remodeling as a function of hFF-content and inhibition of fibrinolysis resulted in stable tissue integrity for up to 5 weeks. The rotating mold device developed in frame of this work enabled the production of tubes with clinically relevant dimensions of up to 10 cm in length and 22 mm in diameter which-in combination with advanced bioreactor technology for controlled production of functional iPSC-derivatives-paves the way towards the clinical translation of a biological cardiac assist device., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. A micro-LED array based platform for spatio-temporal optogenetic control of various cardiac models.
- Author
-
Junge S, Ricci Signorini ME, Al Masri M, Gülink J, Brüning H, Kasperek L, Szepes M, Bakar M, Gruh I, Heisterkamp A, and Torres-Mapa ML
- Subjects
- Humans, Channelrhodopsins genetics, Myocytes, Cardiac physiology, Optogenetics methods, Induced Pluripotent Stem Cells
- Abstract
Optogenetics relies on dynamic spatial and temporal control of light to address emerging fundamental and therapeutic questions in cardiac research. In this work, a compact micro-LED array, consisting of 16 × 16 pixels, is incorporated in a widefield fluorescence microscope for controlled light stimulation. We describe the optical design of the system that allows the micro-LED array to fully cover the field of view regardless of the imaging objective used. Various multicellular cardiac models are used in the experiments such as channelrhodopsin-2 expressing aggregates of cardiomyocytes, termed cardiac bodies, and bioartificial cardiac tissues derived from human induced pluripotent stem cells. The pacing efficiencies of the cardiac bodies and bioartificial cardiac tissues were characterized as a function of illumination time, number of switched-on pixels and frequency of stimulation. To demonstrate dynamic stimulation, steering of calcium waves in HL-1 cell monolayer expressing channelrhodopsin-2 was performed by applying different configurations of patterned light. This work shows that micro-LED arrays are powerful light sources for optogenetic control of contraction and calcium waves in cardiac monolayers, multicellular bodies as well as three-dimensional artificial cardiac tissues., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
7. Performance and feasibility of three different approaches for computer based semi-automated analysis of ventricular arrhythmias in telemetric long-term ECG in cynomolgus monkeys.
- Author
-
Eiringhaus J, de Vries AL, Hohmann S, Böthig D, Müller-Leisse J, Hillmann HAK, Martens A, Zweigerdt R, Schrod A, Martin U, Duncker D, Gruh I, and Veltmann C
- Subjects
- Animals, Male, Macaca fascicularis, Feasibility Studies, Computers, Arrhythmias, Cardiac diagnosis, Electrocardiography
- Abstract
Computer-based analysis of long-term electrocardiogram (ECG) monitoring in animal models represents a cost and time-consuming process as manual supervision is often performed to ensure accuracy in arrhythmia detection. Here, we investigate the performance and feasibility of three ECG interval analysis approaches A) attribute-based, B) attribute- and pattern recognition-based and C) combined approach with additional manual beat-to-beat analysis (gold standard) with regard to subsequent detection of ventricular arrhythmias (VA) and time consumption. ECG analysis was performed on ECG raw data of 5 male cynomolgus monkeys (1000 h total, 2 × 100 h per animal). Both approaches A and B overestimated the total number of arrhythmias compared to gold standard (+8.92% vs. +6.47%). With regard to correct classification of detected VA event numbers (accelerated idioventricular rhythms [AIVR], ventricular tachycardia [VT]) approach B revealed higher accuracy compared to approach A. Importantly, VA burden (% of time) was precisely depicted when using approach B (-1.13%), whereas approach A resulted in relevant undersensing of ventricular arrhythmias (-11.76%). Of note, approach A and B could be performed with significant less working time (-95% and - 91% working time) compared to gold standard. In sum, we show that a combination of attribute-based and pattern recognition analysis (approach B) can reproduce VA burden with acceptable accuracy without using manual supervision. Since this approach allowed analyses to be performed with distinct time saving it represents a valuable approach for cost and time efficient analysis of large preclinical ECG datasets., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
8. Generation of human induced pluripotent stem cell line encoding for a genetically encoded voltage indicator Arclight A242.
- Author
-
Vivekanandan R, Szepes M, Ricci Signorini ME, Kravchenko D, Kiefer J, Berger S, Fricke V, Göhring G, and Gruh I
- Subjects
- Humans, Action Potentials, Myocytes, Cardiac metabolism, Cell Differentiation physiology, Electrophysiological Phenomena, Induced Pluripotent Stem Cells metabolism
- Abstract
Genetically encoded voltage indicators (GEVIs) allow for monitoring membrane potential changes in neurons and cardiomyocytes (CMs) as an alternative to patch-clamp techniques. GEVIs facilitate non-invasive, high throughput screening of electrophysiological properties of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). A dual transgenic hiPSC line with Arclight A242 (GEVI) and an antibiotic resistance cardiac selection cassette was successfully generated from an earlier established hiPSC line MHHi001-A. After cardiac differentiation and selection, purified populations of CMs with constitutive GEVI expression can be utilized for studying cardiac development, disease modeling, and drug testing., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
9. Generation of human induced pluripotent stem cell lines encoding for genetically encoded calcium indicators RCaMP1h and GCaMP6f.
- Author
-
Ricci Signorini ME, Szepes M, Melchert A, Bakar M, Merkert S, Haase A, Göhring G, Martin U, and Gruh I
- Subjects
- Action Potentials, Calcium metabolism, Cell Differentiation, Humans, Myocytes, Cardiac metabolism, Induced Pluripotent Stem Cells metabolism
- Abstract
Calcium plays a key role in cardiomyocytes (CMs) for the translation of the electrical impulse of an action potential into contraction forces. A rapid, not-invasive fluorescence imaging technology allows for the monitoring of calcium transients in human induced pluripotent stem cell derived-cardiomyocytes (hiPSC-CMs) to investigate the cardiac electrophysiology in vitro and after cell transplantation in vivo. The genetically encoded calcium indicators (GECIs) GCaMP6f or RCaMP1h were successfully transfected in the previously established hiPSC line MHHi001-A, together with a cardiac specific antibiotic selection cassette facilitating the monitoring of the calcium handling in highly pure populations of hiPSC-CMs., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
10. iPSC culture expansion selects against putatively actionable mutations in the mitochondrial genome.
- Author
-
Kosanke M, Davenport C, Szepes M, Wiehlmann L, Kohrn T, Dorda M, Gruber J, Menge K, Sievert M, Melchert A, Gruh I, Göhring G, and Martin U
- Subjects
- Cell Culture Techniques, Genome, Mitochondrial, Genomic Instability, Humans, Cell Differentiation, Cellular Reprogramming, DNA, Mitochondrial genetics, Induced Pluripotent Stem Cells physiology, Mitochondria genetics, Mutation, Selection, Genetic
- Abstract
Therapeutic application of induced pluripotent stem cell (iPSC) derivatives requires comprehensive assessment of the integrity of their nuclear and mitochondrial DNA (mtDNA) to exclude oncogenic potential and functional deficits. It is unknown, to which extent mtDNA variants originate from their parental cells or from de novo mutagenesis, and whether dynamics in heteroplasmy levels are caused by inter- and intracellular selection or genetic drift. Sequencing of mtDNA of 26 iPSC clones did not reveal evidence for de novo mutagenesis, or for any selection processes during reprogramming or differentiation. Culture expansion, however, selected against putatively actionable mtDNA mutations. Altogether, our findings point toward a scenario in which intracellular selection of mtDNA variants during culture expansion shapes the mutational landscape of the mitochondrial genome. Our results suggest that intercellular selection and genetic drift exert minor impact and that the bottleneck effect in context of the mtDNA genetic pool might have been overestimated., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. Dextran-based scaffolds for in-situ hydrogelation: Use for next generation of bioartificial cardiac tissues.
- Author
-
Banerjee S, Szepes M, Dibbert N, Rios-Camacho JC, Kirschning A, Gruh I, and Dräger G
- Subjects
- Collagen chemistry, Cross-Linking Reagents chemistry, Fibroblasts metabolism, Humans, Hydrazones chemistry, Induced Pluripotent Stem Cells metabolism, Myocardium metabolism, Tissue Engineering methods, Dextrans chemistry, Hydrogels chemistry, Myocytes, Cardiac metabolism, Tissue Scaffolds chemistry
- Abstract
In pursuit of a chemically-defined matrix for in vitro cardiac tissue generation, we present dextran (Dex)-derived hydrogels as matrices suitable for bioartificial cardiac tissues (BCT). The dextran hydrogels were generated in situ by using hydrazone formation as the crosslinking reaction. Material properties were flexibly adjusted, by varying the degrees of derivatization and the molecular weight of dextran used. Furthermore, to modulate dextran's bioactivity, cyclic pentapeptide RGD was coupled to its backbone. BCTs were generated by using a blend of modified dextran and human collagen (hColI) in combination with induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and fibroblasts. These hColI + Dex blends with or without RGD supported tissue formation and functional maturation of CMs. Contraction forces (hColI + Dex-RGD: 0.27 ± 0.02 mN; hColI + Dex: 0.26 ± 0.01 mN) and frequencies were comparable to published constructs. Thus, we could demonstrate that, independent of the presence of RGD, our covalently linked dextran hydrogels are a promising matrix for building cardiac grafts., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
12. Establishment of MHHi001-A-5, a GCaMP6f and RedStar dual reporter human iPSC line for in vitro and in vivo characterization and in situ tracing of iPSC derivatives.
- Author
-
Haase A, Kohrn T, Fricke V, Ricci Signorini ME, Witte M, Göhring G, Gruh I, and Martin U
- Subjects
- Animals, Calcium, Cell Differentiation, Genes, Reporter, Genetic Engineering, Humans, Myocytes, Cardiac, Induced Pluripotent Stem Cells
- Abstract
Transgenic hiPSC lines carrying reporter genes represent valuable tools for functional characterization of iPSC derivatives, disease modelling and clinical evaluation of cell therapies. Here, the hiPSC line 'Phoenix' (Haase et al., 2017) was genetically engineered using TALEN-based integration of the calcium sensor GCaMP6f and RedStar
nuc reporter into the AAVS1 site. Characterization of undifferentiated cells and functional investigation of hiPSC-derived cardiomyocytes-containing BCTs showed a strong intracellular calcium transient-dependent GCaMP6f and eminent RedStarnuc signal. Therefore, our dual reporter line provides an excellent tool to facilitate monitoring of engraftment, calcium fluctuations and coupling of iPSC derivatives such as cardiomyocytes in vitro and in vivo in animal models., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
13. Dual Function of iPSC-Derived Pericyte-Like Cells in Vascularization and Fibrosis-Related Cardiac Tissue Remodeling In Vitro.
- Author
-
Szepes M, Melchert A, Dahlmann J, Hegermann J, Werlein C, Jonigk D, Haverich A, Martin U, Olmer R, and Gruh I
- Subjects
- Bioartificial Organs, Endothelial Cells cytology, Extracellular Matrix metabolism, Fibroblasts cytology, Fibroblasts metabolism, Fibrosis genetics, Fibrosis pathology, Humans, Induced Pluripotent Stem Cells metabolism, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac cytology, Myocytes, Cardiac pathology, Neovascularization, Pathologic genetics, Neovascularization, Pathologic pathology, Pericytes cytology, Pericytes metabolism, Sarcomeres genetics, Sarcomeres metabolism, Ventricular Remodeling genetics, Cell Differentiation genetics, Fibrosis therapy, Induced Pluripotent Stem Cells cytology, Myocytes, Cardiac metabolism, Neovascularization, Pathologic therapy
- Abstract
Myocardial interstitial fibrosis (MIF) is characterized by excessive extracellular matrix (ECM) deposition, increased myocardial stiffness, functional weakening, and compensatory cardiomyocyte (CM) hypertrophy. Fibroblasts (Fbs) are considered the principal source of ECM, but the contribution of perivascular cells, including pericytes (PCs), has gained attention, since MIF develops primarily around small vessels. The pathogenesis of MIF is difficult to study in humans because of the pleiotropy of mutually influencing pathomechanisms, unpredictable side effects, and the lack of available patient samples. Human pluripotent stem cells (hPSCs) offer the unique opportunity for the de novo formation of bioartificial cardiac tissue (BCT) using a variety of different cardiovascular cell types to model aspects of MIF pathogenesis in vitro. Here, we have optimized a protocol for the derivation of hPSC-derived PC-like cells (iPSC-PCs) and present a BCT in vitro model of MIF that shows their central influence on interstitial collagen deposition and myocardial tissue stiffening. This model was used to study the interplay of different cell types-i.e., hPSC-derived CMs, endothelial cells (ECs), and iPSC-PCs or primary Fbs, respectively. While iPSC-PCs improved the sarcomere structure and supported vascularization in a PC-like fashion, the functional and histological parameters of BCTs revealed EC- and PC-mediated effects on fibrosis-related cardiac tissue remodeling.
- Published
- 2020
- Full Text
- View/download PDF
14. The Long Non-coding RNA Cyrano Is Dispensable for Pluripotency of Murine and Human Pluripotent Stem Cells.
- Author
-
Hunkler HJ, Hoepfner J, Huang CK, Chatterjee S, Jara-Avaca M, Gruh I, Bolesani E, Zweigerdt R, Thum T, and Bär C
- Subjects
- Animals, Base Sequence, CRISPR-Cas Systems genetics, Cell Differentiation genetics, Cell Self Renewal genetics, Gene Silencing, Human Embryonic Stem Cells metabolism, Humans, Mice, Knockout, RNA, Long Noncoding genetics, Transcriptome genetics, Induced Pluripotent Stem Cells metabolism, RNA, Long Noncoding metabolism
- Abstract
Pluripotency is tightly regulated and is crucial for stem cells and their implementation for regenerative medicine. Non-coding RNAs, especially long non-coding RNAs (lncRNAs) emerged as orchestrators of versatile (patho)-physiological processes on the transcriptional and post-transcriptional level. Cyrano, a well-conserved lncRNA, is highly expressed in stem cells suggesting an important role in pluripotency, which we aimed to investigate in loss-off-function (LOF) experiments. Cyrano was described previously to be essential for the maintenance of mouse embryonic stem cell (ESC) pluripotency. In contrast, using different genetic models, we here found Cyrano to be dispensable in murine and human iPSCs and in human ESCs. RNA sequencing revealed only a moderate influence of Cyrano on the global transcriptome. In line, Cyrano-depleted iPSCs retained the potential to differentiate into the three germ layers. In conclusion, different methods were applied for LOF studies to rule out potential off-target effects. These approaches revealed that Cyrano does not impact pluripotency., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
15. Continuous WNT Control Enables Advanced hPSC Cardiac Processing and Prognostic Surface Marker Identification in Chemically Defined Suspension Culture.
- Author
-
Halloin C, Schwanke K, Löbel W, Franke A, Szepes M, Biswanath S, Wunderlich S, Merkert S, Weber N, Osten F, de la Roche J, Polten F, Christoph Wollert K, Kraft T, Fischer M, Martin U, Gruh I, Kempf H, and Zweigerdt R
- Published
- 2019
- Full Text
- View/download PDF
16. A gene therapeutic approach to inhibit calcium and integrin binding protein 1 ameliorates maladaptive remodelling in pressure overload.
- Author
-
Grund A, Szaroszyk M, Döppner JK, Malek Mohammadi M, Kattih B, Korf-Klingebiel M, Gigina A, Scherr M, Kensah G, Jara-Avaca M, Gruh I, Martin U, Wollert KC, Gohla A, Katus HA, Müller OJ, Bauersachs J, and Heineke J
- Subjects
- Animals, Calcineurin metabolism, Calcium-Binding Proteins genetics, Cells, Cultured, Disease Models, Animal, Fibrosis, Heart Failure genetics, Heart Failure metabolism, Heart Failure physiopathology, Hypertrophy, Left Ventricular genetics, Hypertrophy, Left Ventricular metabolism, Hypertrophy, Left Ventricular physiopathology, Male, Mice, Inbred C57BL, Myocytes, Cardiac pathology, NFATC Transcription Factors metabolism, Neovascularization, Physiologic, RNA, Small Interfering genetics, Rats, Sprague-Dawley, Signal Transduction, Ventricular Dysfunction, Left genetics, Ventricular Dysfunction, Left metabolism, Ventricular Dysfunction, Left physiopathology, Calcium-Binding Proteins metabolism, Heart Failure therapy, Hypertrophy, Left Ventricular therapy, Myocytes, Cardiac metabolism, RNA, Small Interfering metabolism, RNAi Therapeutics, Ventricular Dysfunction, Left therapy, Ventricular Function, Left, Ventricular Remodeling
- Abstract
Aims: Chronic heart failure is becoming increasingly prevalent and is still associated with a high mortality rate. Myocardial hypertrophy and fibrosis drive cardiac remodelling and heart failure, but they are not sufficiently inhibited by current treatment strategies. Furthermore, despite increasing knowledge on cardiomyocyte intracellular signalling proteins inducing pathological hypertrophy, therapeutic approaches to target these molecules are currently unavailable. In this study, we aimed to establish and test a therapeutic tool to counteract the 22 kDa calcium and integrin binding protein (CIB) 1, which we have previously identified as nodal regulator of pathological cardiac hypertrophy and as activator of the maladaptive calcineurin/NFAT axis., Methods and Results: Among three different sequences, we selected a shRNA construct (shCIB1) to specifically down-regulate CIB1 by 50% upon adenoviral overexpression in neonatal rat cardiomyocytes (NRCM), and upon overexpression by an adeno-associated-virus (AAV) 9 vector in mouse hearts. Overexpression of shCIB1 in NRCM markedly reduced cellular growth, improved contractility of bioartificial cardiac tissue and reduced calcineurin/NFAT activation in response to hypertrophic stimulation. In mice, administration of AAV-shCIB1 strongly ameliorated eccentric cardiac hypertrophy and cardiac dysfunction during 2 weeks of pressure overload by transverse aortic constriction (TAC). Ultrastructural and molecular analyses revealed markedly reduced myocardial fibrosis, inhibition of hypertrophy associated gene expression and calcineurin/NFAT as well as ERK MAP kinase activation after TAC in AAV-shCIB1 vs. AAV-shControl treated mice. During long-term exposure to pressure overload for 10 weeks, AAV-shCIB1 treatment maintained its anti-hypertrophic and anti-fibrotic effects, but cardiac function was no longer improved vs. AAV-shControl treatment, most likely resulting from a reduction in myocardial angiogenesis upon downregulation of CIB1., Conclusions: Inhibition of CIB1 by a shRNA-mediated gene therapy potently inhibits pathological cardiac hypertrophy and fibrosis during pressure overload. While cardiac function is initially improved by shCIB1, this cannot be kept up during persisting overload.
- Published
- 2019
- Full Text
- View/download PDF
17. Corrigendum to "Anti-androgenic therapy with finasteride improves cardiac function, attenuates remodeling and reverts pathologic gene-expression after myocardial infarction in mice" Journal of Molecular and Cellular Cardiology 122 (2018) 114-124.
- Author
-
Froese N, Wang H, Zwadlo C, Wang Y, Grund A, Gigina A, Hofmann M, Kilian K, Scharf G, Korf-Klingebiel M, Melchert A, Signorini MER, Halloin C, Zweigerdt R, Martin U, Gruh I, Wollert KC, Geffers R, Bauersachs J, and Heineke J
- Published
- 2018
- Full Text
- View/download PDF
18. Anti-androgenic therapy with finasteride improves cardiac function, attenuates remodeling and reverts pathologic gene-expression after myocardial infarction in mice.
- Author
-
Froese N, Wang H, Zwadlo C, Wang Y, Grund A, Gigina A, Hofmann M, Kilian K, Scharf G, Korf-Klingebiel M, Melchert A, Signorini MER, Halloin C, Zweigerdt R, Martin U, Gruh I, Wollert KC, Geffers R, Bauersachs J, and Heineke J
- Subjects
- Analysis of Variance, Animals, Cardiomegaly drug therapy, Cell Line, Dihydrotestosterone metabolism, Endothelial Cells drug effects, Fibroblasts drug effects, Fibrosis, Human Umbilical Vein Endothelial Cells, Humans, Induced Pluripotent Stem Cells, Male, Mice, Muscle Contraction drug effects, Myocardium pathology, Neovascularization, Physiologic drug effects, Rats, Rats, Sprague-Dawley, Androgen Antagonists therapeutic use, Finasteride therapeutic use, Gene Expression drug effects, Myocardial Infarction drug therapy, Ventricular Function, Left drug effects, Ventricular Remodeling drug effects
- Abstract
Maladaptive cardiac remodeling after myocardial infarction (MI) is increasingly contributing to the prevalence of chronic heart failure. Women show less severe remodeling, a reduced mortality and a better systolic function after MI compared to men. Although sex hormones are being made responsible for these differences, it remains currently unknown how this could be translated into therapeutic strategies. Because we had recently demonstrated that inhibition of the conversion of testosterone to its highly active metabolite dihydrotestosterone (DHT) by finasteride effectively reduces cardiac hypertrophy and improves heart function during pressure overload, we asked here whether this strategy could be applied to post-MI remodeling. We found increased abundance of DHT and increased expression of androgen responsive genes in the mouse myocardium after experimental MI. Treatment of mice with finasteride for 21 days (starting 7 days after surgery), reduced myocardial DHT levels and markedly attenuated cardiac dysfunction as well as hypertrophic remodeling after MI. Histological and molecular analyses showed reduced MI triggered interstitial fibrosis, reduced cardiomyocyte hypertrophy and increased capillary density in the myocardium of finasteride treated mice. Mechanistically, this was associated with decreased activation of myocardial growth-signaling pathways, a comprehensive normalization of pathological myocardial gene-expression as revealed by RNA deep-sequencing and with direct effects of finasteride on cardiac fibroblasts and endothelial cells. In conclusion, we demonstrated a beneficial role of anti-androgenic treatment with finasteride in post-MI remodeling of mice. As finasteride is already approved for the treatment of benign prostate disease, it could potentially be evaluated as therapeutic strategy for heart failure after MI., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
19. Bioengineered Cardiac Tissue Based on Human Stem Cells for Clinical Application.
- Author
-
Jara Avaca M and Gruh I
- Subjects
- Animals, Humans, Myocardium cytology, Myocardium metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Pluripotent Stem Cells cytology, Pluripotent Stem Cells metabolism, Tissue Engineering methods
- Abstract
Engineered cardiac tissue might enable novel therapeutic strategies for the human heart in a number of acquired and congenital diseases. With recent advances in stem cell technologies, namely the availability of pluripotent stem cells, the generation of potentially autologous tissue grafts has become a realistic option. Nevertheless, a number of limitations still have to be addressed before clinical application of engineered cardiac tissue based on human stem cells can be realized. We summarize current progress and pending challenges regarding the optimal cell source, cardiomyogenic lineage specification, purification, safety of genetic cell engineering, and genomic stability. Cardiac cells should be combined with clinical grade scaffold materials for generation of functional myocardial tissue in vitro. Scale-up to clinically relevant dimensions is mandatory, and tissue vascularization is most probably required both for preclinical in vivo testing in suitable large animal models and for clinical application. Graphical Abstract.
- Published
- 2018
- Full Text
- View/download PDF
20. EMC10 (Endoplasmic Reticulum Membrane Protein Complex Subunit 10) Is a Bone Marrow-Derived Angiogenic Growth Factor Promoting Tissue Repair After Myocardial Infarction.
- Author
-
Reboll MR, Korf-Klingebiel M, Klede S, Polten F, Brinkmann E, Reimann I, Schönfeld HJ, Bobadilla M, Faix J, Kensah G, Gruh I, Klintschar M, Gaestel M, Niessen HW, Pich A, Bauersachs J, Gogos JA, Wang Y, and Wollert KC
- Subjects
- Angiogenic Proteins administration & dosage, Angiogenic Proteins deficiency, Angiogenic Proteins genetics, Animals, Bone Marrow Transplantation, Cells, Cultured, Disease Models, Animal, Endothelial Cells metabolism, Genotype, Intracellular Signaling Peptides and Proteins metabolism, Macrophages metabolism, Membrane Proteins administration & dosage, Membrane Proteins deficiency, Membrane Proteins genetics, Mice, Inbred C57BL, Mice, Knockout, Monocytes metabolism, Monomeric GTP-Binding Proteins metabolism, Myocardial Infarction drug therapy, Myocardial Infarction genetics, Myocardial Infarction pathology, Myocardium pathology, Phenotype, Protein Serine-Threonine Kinases metabolism, Signal Transduction, Time Factors, p21-Activated Kinases metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Angiogenic Proteins metabolism, Bone Marrow Cells metabolism, Membrane Proteins metabolism, Myocardial Infarction metabolism, Myocardium metabolism, Neovascularization, Physiologic drug effects, Wound Healing drug effects
- Abstract
Background: Clinical trials of bone marrow cell-based therapies after acute myocardial infarction (MI) have produced mostly neutral results. Treatment with specific bone marrow cell-derived secreted proteins may provide an alternative biological approach to improving tissue repair and heart function after MI. We recently performed a bioinformatic secretome analysis in bone marrow cells from patients with acute MI and discovered a poorly characterized secreted protein, EMC10 (endoplasmic reticulum membrane protein complex subunit 10), showing activity in an angiogenic screen., Methods: We investigated the angiogenic potential of EMC10 and its mouse homolog (Emc10) in cultured endothelial cells and infarcted heart explants. We defined the cellular sources and function of Emc10 after MI using wild-type, Emc10 -deficient, and Emc10 bone marrow-chimeric mice subjected to transient coronary artery ligation. Furthermore, we explored the therapeutic potential of recombinant Emc10 delivered by osmotic minipumps after MI in heart failure-prone FVB/N mice., Results: Emc10 signaled through small GTPases, p21-activated kinase, and the p38 mitogen-activated protein kinase (MAPK)-MAPK-activated protein kinase 2 (MK2) pathway to promote actin polymerization and endothelial cell migration. Confirming the importance of these signaling events in the context of acute MI, Emc10 stimulated endothelial cell outgrowth from infarcted mouse heart explants via p38 MAPK-MK2. Emc10 protein abundance was increased in the infarcted region of the left ventricle and in the circulation of wild-type mice after MI. Emc10 expression was also increased in left ventricular tissue samples from patients with acute MI. Bone marrow-derived monocytes and macrophages were the predominant sources of Emc10 in the infarcted murine heart. Emc10 KO mice showed no cardiovascular phenotype at baseline. After MI, however, capillarization of the infarct border zone was impaired in KO mice, and the animals developed larger infarct scars and more pronounced left ventricular remodeling compared with wild-type mice. Transplanting KO mice with wild-type bone marrow cells rescued the angiogenic defect and ameliorated left ventricular remodeling. Treating FVB/N mice with recombinant Emc10 enhanced infarct border-zone capillarization and exerted a sustained beneficial effect on left ventricular remodeling., Conclusions: We have identified Emc10 as a previously unknown angiogenic growth factor that is produced by bone marrow-derived monocytes and macrophages as part of an endogenous adaptive response that can be enhanced therapeutically to repair the heart after MI., (© 2017 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
21. Transplantation of purified iPSC-derived cardiomyocytes in myocardial infarction.
- Author
-
Rojas SV, Kensah G, Rotaermel A, Baraki H, Kutschka I, Zweigerdt R, Martin U, Haverich A, Gruh I, and Martens A
- Subjects
- Animals, Cell Differentiation genetics, Disease Models, Animal, Humans, Magnetic Resonance Imaging, Mice, Myocardial Contraction genetics, Myocardial Infarction diagnostic imaging, Myocardial Infarction physiopathology, Myocardium pathology, Heart Transplantation, Induced Pluripotent Stem Cells transplantation, Myocardial Infarction therapy, Myocytes, Cardiac transplantation, Stem Cell Transplantation
- Abstract
Background: Induced pluripotent stem cells (iPSC) can be differentiated into cardiomyocytes and represent a possible autologous cell source for myocardial repair. We analyzed the engraftment and functional effects of murine iPSC-derived cardiomyocytes (iPSC-CMs) in a murine model of myocardial infarction., Methods and Results: To maximize cardiomyocyte yield and purity a genetic purification protocol was applied. Murine iPSCs were genetically modified to express a Zeocin™ resistance gene under control of the cardiac-specific α-myosin heavy chain (α-MHC, MYH6) promoter. Thus, CM selection was performed during in vitro differentiation. iPSC-CM aggregates ("cardiac bodies", CBs) were transplanted on day 14 after LAD ligation into the hearts of previously LAD-ligated mice (800 CBs/animal; 2-3x106 CMs). Animals were treated with placebo (PBS, n = 14) or iPSC-CMs (n = 35). Myocardial remodeling and function were evaluated by magnetic resonance imaging (MRI), conductance catheter (CC) analysis and histological morphometry. In vitro and in vivo differentiation was investigated. Follow up was 28 days (including histological assessment and functional analysis). iPSC-CM purity was >99%. Transplanted iPSC-CMs formed mature grafts within the myocardium, expressed cardiac markers and exhibited sarcomeric structures. Intramyocardial transplantation of iPSC-CMs significantly improved myocardial remodeling and left ventricular function 28 days after LAD-ligation., Conclusions: We conclude that iPSCs can effectively be differentiated into cardiomyocytes and genetically enriched to high purity. iPSC derived cardiomyocytes engraft within the myocardium of LAD-ligated mice and contribute to improve left ventricular function.
- Published
- 2017
- Full Text
- View/download PDF
22. A Synthetic Toolbox for the In Situ Formation of Functionalized Homo- and Heteropolysaccharide-Based Hydrogel Libraries.
- Author
-
Dibbert N, Krause A, Rios-Camacho JC, Gruh I, Kirschning A, and Dräger G
- Subjects
- Glucuronic Acid chemistry, Hexuronic Acids chemistry, Hydrogels chemistry, Polysaccharides chemistry, Tissue Engineering, Alginates chemistry, Dextrans chemistry, Fibroblasts chemistry, Glucans chemistry, Hyaluronic Acid chemistry, Hydrogels chemical synthesis, Polysaccharides chemical synthesis
- Abstract
A synthetic toolbox for the introduction of aldehydo and hydrazido groups into the polysaccharides hyaluronic acid, alginate, dextran, pullulan, glycogen, and carboxymethyl cellulose and their use for hydrogel formation is reported. Upon mixing differently functionalized polysaccharides derived from the same natural precursor, hydrazone cross-linking takes place, which results in formation of a hydrogel composed of one type of polysaccharide backbone. Likewise, hydrogels based on two different polysaccharide strands can be formed after mixing the corresponding aldehydo- and hydrazido-modified polysaccharides. A second line of these studies paves the way to introduce a biomedically relevant ligand, namely, the adhesion factor cyclic RGD pentapeptide, by using an orthogonal click reaction. This set of modified polysaccharides served to create a library of hydrogels that differ in the combination of polysaccharide strands and the degree of cross-linking. The different hydrogels were evaluated with respect to their rheological properties, their ability to absorb water, and their cytotoxicity towards human fibroblast cell cultures. None of the hydrogels studied were cytotoxic, and, hence, they are in principal biocompatible for applications in tissue engineering., (© 2016 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2016
- Full Text
- View/download PDF
23. Tightly regulated 'all-in-one' lentiviral vectors for protection of human hematopoietic cells from anticancer chemotherapy.
- Author
-
Lachmann N, Brennig S, Hillje R, Schermeier H, Phaltane R, Dahlmann J, Gruh I, Heinz N, Schiedlmeier B, Baum C, and Moritz T
- Subjects
- Antimetabolites, Antineoplastic toxicity, Cytarabine toxicity, Cytidine Deaminase biosynthesis, Cytidine Deaminase genetics, Doxycycline pharmacology, Genetic Vectors administration & dosage, Genetic Vectors genetics, Hematopoietic Stem Cells virology, Humans, K562 Cells, Primary Cell Culture, Promoter Regions, Genetic, Transgenes, Genetic Therapy methods, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells physiology, Lentivirus genetics
- Abstract
Successful application of gene therapy strategies may require stringently regulated transgene expression. Along this line, we describe a doxycycline (Dox)-inducible 'all-in-one' lentiviral vector design using the pTET-T11 (TII) minimal-promoter and a reverse transactivator protein (rtTA2S-M2) driven by the phosphoglycerate kinase promoter allowing for tight regulation of transgene expression (Lv.TII vectors). Vector design was evaluated in human hematopoietic cells in the context of cytidine deaminase (hCDD)-based myeloprotective gene therapy. Upon Dox administration, a rapid (16-24 h) and dose-dependent (>0.04 μg ml(-1) Dox) onset of transgene expression was detected in Lv.TII.CDD gene-modified K562 cells as well as in primary human CD34(+) hematopoietic cells. Importantly, in both cell models low background transgene expression was observed in the absence of Dox. Functionality of Dox-inducible hCDD expression was demonstrated by >10-fold increase in cytosine arabinoside (1-β-d-arabinofuranosylcytosine, Ara-C) resistance of Lv.TII.CDD-transduced K562 cells. In addition, Lv.TII.CDD-transduced CD34(+)-derived myeloid cells were protected from up to 300 nm Ara-C (control affected from 50 nm onwards). These data clearly demonstrate the suitability of our self-inactivating lentiviral vector to induce robust, tightly regulated transgene expression in human hematopoietic cells with minimal background activity and highlight the potential of our construct in myeloprotective gene therapy strategies.
- Published
- 2015
- Full Text
- View/download PDF
24. Controlling expansion and cardiomyogenic differentiation of human pluripotent stem cells in scalable suspension culture.
- Author
-
Kempf H, Olmer R, Kropp C, Rückert M, Jara-Avaca M, Robles-Diaz D, Franke A, Elliott DA, Wojciechowski D, Fischer M, Roa Lara A, Kensah G, Gruh I, Haverich A, Martin U, and Zweigerdt R
- Subjects
- Bioreactors, Culture Media, Culture Media, Conditioned, Humans, Membrane Potentials, Myocytes, Cardiac metabolism, Pluripotent Stem Cells metabolism, Batch Cell Culture Techniques, Cell Culture Techniques, Cell Differentiation, Myocytes, Cardiac cytology, Pluripotent Stem Cells cytology
- Abstract
To harness the potential of human pluripotent stem cells (hPSCs), an abundant supply of their progenies is required. Here, hPSC expansion as matrix-independent aggregates in suspension culture was combined with cardiomyogenic differentiation using chemical Wnt pathway modulators. A multiwell screen was scaled up to stirred Erlenmeyer flasks and subsequently to tank bioreactors, applying controlled feeding strategies (batch and cyclic perfusion). Cardiomyogenesis was sensitive to the GSK3 inhibitor CHIR99021 concentration, whereas the aggregate size was no prevailing factor across culture platforms. However, in bioreactors, the pattern of aggregate formation in the expansion phase dominated subsequent differentiation. Global profiling revealed a culture-dependent expression of BMP agonists/antagonists, suggesting their decisive role in cell-fate determination. Furthermore, metallothionein was discovered as a potentially stress-related marker in hPSCs. In 100 ml bioreactors, the production of 40 million predominantly ventricular-like cardiomyocytes (up to 85% purity) was enabled that were directly applicable to bioartificial cardiac tissue formation., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
25. Your heart on a chip: iPSC-based modeling of Barth-syndrome-associated cardiomyopathy.
- Author
-
Zweigerdt R, Gruh I, and Martin U
- Subjects
- Humans, Barth Syndrome physiopathology, Cardiomyopathy, Dilated physiopathology, Induced Pluripotent Stem Cells physiology, Mitochondrial Diseases physiopathology, Models, Biological, Tissue Engineering methods, Transcription Factors genetics
- Abstract
Disease-specific induced pluripotent stem cells (iPSCs) are invaluable tools for studying genetic disorders in a dish. A recent paper by Wang et al. (2014) powerfully combines analysis of human iPSCs with genome editing and tissue engineering, in conjunction with biochemical and physiological assays, to provide insights into Barth-syndrome-associated cardiomyopathy., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
26. Higher frequencies of BCRP+ cardiac resident cells in ischaemic human myocardium.
- Author
-
Emmert MY, Emmert LS, Martens A, Ismail I, Schmidt-Richter I, Gawol A, Seifert B, Haverich A, Martin U, and Gruh I
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2, Adolescent, Adult, Aged, Aged, 80 and over, Biopsy, Cell Differentiation physiology, Female, Heart Atria metabolism, Heart Atria pathology, Heart Ventricles metabolism, Heart Ventricles pathology, Humans, Male, Middle Aged, Myocardial Ischemia metabolism, Myocytes, Cardiac classification, Myocytes, Cardiac pathology, Proto-Oncogene Proteins c-kit metabolism, Stem Cells classification, Stem Cells pathology, Young Adult, ATP-Binding Cassette Transporters metabolism, Myocardial Ischemia pathology, Myocytes, Cardiac metabolism, Neoplasm Proteins metabolism, Stem Cells metabolism
- Abstract
Aims: Several cardiac resident progenitor cell types have been reported for the adult mammalian heart. Here we characterize their frequencies and distribution pattern in non-ischaemic human myocardial tissue and after ischaemic events., Methods and Results: We obtained 55 biopsy samples from human atria and ventricles and used immunohistological analysis to investigate two cardiac cell types, characterized by the expression of breast cancer resistance protein (BCRP)/ABCG2 [for side population (SP) cells] or c-kit. Highest frequencies of BCRP+ cells were detected in the ischaemic right atria with a median of 5.40% (range: 2.48-11.1%) vs. 4.40% (1.79-7.75%) in the non-ischaemic right atria (P = 0.47). Significantly higher amounts were identified in ischaemic compared with non-ischaemic ventricles, viz. 5.44% (3.24-9.30%) vs. 0.74% (0-5.23%) (P = 0.016). Few numbers of BCRP+ cells co-expressed the cardiac markers titin, sarcomeric α-actinin, or Nkx2.5; no co-expression of BCRP and progenitor cell marker Sca-1 or pluripotency markers Oct-3/4, SSEA-3, and SSEA-4 was detected. C-kit+ cells displayed higher frequencies in ischaemic (ratio: 1:25 000 ± 2500 of cell counts) vs. non-ischaemic myocardium (1:105 000 ± 43 000). Breast cancer resistance protein+/c-kit+ cells were not identified. Following in vitro differentiation, BCRP+ cells isolated from human heart biopsy samples (n = 6) showed expression of cardiac troponin T and α-myosin heavy-chain, but no full differentiation into functional beating cardiomyocytes was observed., Conclusion: We were able to demonstrate that BCRP+/CD31- cells are more abundant in the heart than their c-kit+ counterparts. In the non-ischaemic hearts, they are preferentially located in the atria. Following ischaemia, their numbers are elevated significantly. Our data might provide a valuable snapshot at potential progenitor cells after acute ischaemia in vivo, and mapping of these easily accessible cells may influence future cell therapeutic strategies.
- Published
- 2013
- Full Text
- View/download PDF
27. Murine and human pluripotent stem cell-derived cardiac bodies form contractile myocardial tissue in vitro.
- Author
-
Kensah G, Roa Lara A, Dahlmann J, Zweigerdt R, Schwanke K, Hegermann J, Skvorc D, Gawol A, Azizian A, Wagner S, Maier LS, Krause A, Dräger G, Ochs M, Haverich A, Gruh I, and Martin U
- Subjects
- Animals, Ascorbic Acid pharmacology, Cell Culture Techniques methods, Cell Enlargement, Cell Line, Gene Expression, Humans, Induced Pluripotent Stem Cells physiology, Mice, Myocytes, Cardiac physiology, Sarcomeres physiology, Vitamins pharmacology, Bioprosthesis, Induced Pluripotent Stem Cells cytology, Myocardial Contraction physiology, Myocardium cytology, Myocytes, Cardiac cytology, Tissue Engineering methods
- Abstract
Aims: We explored the use of highly purified murine and human pluripotent stem cell (PSC)-derived cardiomyocytes (CMs) to generate functional bioartificial cardiac tissue (BCT) and investigated the role of fibroblasts, ascorbic acid (AA), and mechanical stimuli on tissue formation, maturation, and functionality., Methods and Results: Murine and human embryonic/induced PSC-derived CMs were genetically enriched to generate three-dimensional CM aggregates, termed cardiac bodies (CBs). Addressing the critical limitation of major CM loss after single-cell dissociation, non-dissociated CBs were used for BCT generation, which resulted in a structurally and functionally homogenous syncytium. Continuous in situ characterization of BCTs, for 21 days, revealed that three critical factors cooperatively improve BCT formation and function: both (i) addition of fibroblasts and (ii) ascorbic acid supplementation support extracellular matrix remodelling and CB fusion, and (iii) increasing static stretch supports sarcomere alignment and CM coupling. All factors together considerably enhanced the contractility of murine and human BCTs, leading to a so far unparalleled active tension of 4.4 mN/mm(2) in human BCTs using optimized conditions. Finally, advanced protocols were implemented for the generation of human PSC-derived cardiac tissue using a defined animal-free matrix composition., Conclusion: BCT with contractile forces comparable with native myocardium can be generated from enriched, PSC-derived CMs, based on a novel concept of tissue formation from non-dissociated cardiac cell aggregates. In combination with the successful generation of tissue using a defined animal-free matrix, this represents a major step towards clinical applicability of stem cell-based heart tissue for myocardial repair.
- Published
- 2013
- Full Text
- View/download PDF
28. The use of agarose microwells for scalable embryoid body formation and cardiac differentiation of human and murine pluripotent stem cells.
- Author
-
Dahlmann J, Kensah G, Kempf H, Skvorc D, Gawol A, Elliott DA, Dräger G, Zweigerdt R, Martin U, and Gruh I
- Subjects
- Animals, Cell Differentiation physiology, Humans, Mice, Tissue Engineering, Embryoid Bodies cytology, Embryonic Stem Cells cytology, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Sepharose chemistry
- Abstract
In most pluripotent stem cell differentiation protocols the formation of embryoid bodies (EBs) is an important step. Here we describe a rapid, straightforward soft lithography approach for the preparation of hydrophilic silicon masters from different templates and the subsequent production of patterned agarose-DMEM microwell surfaces for scalable well standardized stem cell aggregation and EB formation. The non-adhesive agarose microwell plates represent an accurate replication of the templates' topography and were used for aggregation of murine induced pluripotent stem cells (iPSCs) and human embryonic stem cells (ESCs). Direct microscopic assessment by time-lapse analysis demonstrated rapid formation of uniformly shaped EBs from murine iPSCs with similar or even more consistent results concerning size distribution and harvesting efficiency compared to the commonly used but time-consuming hanging drop technique. For human ESCs, homogenous aggregates were obtained after single cell inoculation on agarose microwells with efficient differentiation into the cardiac lineage using state-of-the-art protocols for directed differentiation via small molecules. With this soft lithography-based strategy, sufficient and reproducible numbers of stem cell-derived cardiomyocytes necessary for tissue engineering purposes can be realized in a highly controllable manner. Moreover, it might be useful for different cell types in any application that requires scalable and highly standardized aggregation., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
29. Efficient in vivo regulation of cytidine deaminase expression in the haematopoietic system using a doxycycline-inducible lentiviral vector system.
- Author
-
Lachmann N, Brennig S, Pfaff N, Schermeier H, Dahlmann J, Phaltane R, Gruh I, Modlich U, Schambach A, Baum C, and Moritz T
- Subjects
- Animals, Blotting, Western, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Bone Marrow Transplantation, Cell Line, Cells, Cultured, Cytarabine pharmacology, Cytidine Deaminase metabolism, Dose-Response Relationship, Drug, Female, Genetic Vectors genetics, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Hematopoietic System cytology, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microscopy, Fluorescence, Spleen cytology, Spleen drug effects, Spleen metabolism, Thymus Gland cytology, Thymus Gland drug effects, Thymus Gland metabolism, Time-Lapse Imaging methods, Transgenes genetics, Cytidine Deaminase genetics, Doxycycline pharmacology, Gene Expression Regulation, Enzymologic drug effects, Hematopoietic System metabolism, Lentivirus genetics
- Abstract
Regulated transgene expression may reduce transgene-specific and genotoxic risks associated with gene therapy. To prove this concept, we have investigated the suitability of doxycycline (Dox)-inducible human cytidine deaminase (hCDD) overexpression from lentiviral vectors to mediate effective myeloprotection while circumventing the lymphotoxicity observed with constitutive CDD activity. Rapid Dox-mediated transgene induction associated with a 6-17-fold increase in drug resistance was observed in 32D and primary murine bone marrow (BM) cells. Moreover, robust Dox-regulated transgene expression in the entire haematopoietic system was demonstrated for primary and secondary recipients of hCDD-transduced R26-M2rtTA transgenic BM cells. Furthermore, mice were significantly protected from myelosuppressive chemotherapy as evidenced by accelerated recovery of granulocytes (1.9±0.6 vs 1.3±0.3, P=0.034) and platelets (883±194 vs 584±160 10(3) per μl, P=0.011). Minimal transgene expression in the non-induced state and no overt cellular toxicities including lymphotoxicity were detected. Thus, using a relevant murine transplant model our data provide conclusive evidence that drug-resistance transgenes can be expressed in a regulated fashion in the lymphohaematopoietic system, and that Dox-inducible systems may be used to reduce myelotoxic side effect of anticancer chemotherapy or to avoid side effects of high constitutive transgene expression.
- Published
- 2013
- Full Text
- View/download PDF
30. Fully defined in situ cross-linkable alginate and hyaluronic acid hydrogels for myocardial tissue engineering.
- Author
-
Dahlmann J, Krause A, Möller L, Kensah G, Möwes M, Diekmann A, Martin U, Kirschning A, Gruh I, and Dräger G
- Subjects
- Animals, Animals, Newborn, Biocompatible Materials chemistry, Cells, Cultured, Cross-Linking Reagents chemistry, Equipment Design, Glucuronic Acid chemistry, Hexuronic Acids chemistry, Hydrogels chemistry, Materials Testing, Myocardium cytology, Rats, Rats, Sprague-Dawley, Tissue Scaffolds, Alginates chemistry, Heart growth & development, Hyaluronic Acid chemistry, Myocardial Contraction physiology, Myocytes, Cardiac cytology, Myocytes, Cardiac physiology, Tissue Engineering instrumentation
- Abstract
Despite recent major advances including reprogramming and directed cardiac differentiation of human cells, therapeutic application of in vitro engineered myocardial tissue is still not feasible due to the inability to construct functional large vascularized contractile tissue patches based on clinically applicable and fully defined matrix components. Typical matrices with preformed porous 3D structure cannot be applied due to the obvious lack of migratory capacity of cardiomyocytes (CM). We have therefore developed a fully defined in situ hydrogelation system based on alginate (Alg) and hyaluronic acid (HyA), in which their aldehyde and hydrazide-derivatives enable covalent hydrazone cross-linking of polysaccharides in the presence of viable myocytes. By varying degrees of derivatization, concentrations and composition of blends in a modular system, mechanophysical properties of the resulting hydrogels are easily adjustable. The hydrogel allowed for the generation of contractile bioartificial cardiac tissue from CM-enriched neonatal rat heart cells, which resembles native myocardium. A combination of HyA and highly purified human collagen I led to significantly increased active contraction force compared to collagen, only. Therefore, our in situ cross-linking hydrogels represent a valuable toolbox for the fine-tuning of engineered cardiac tissue's mechanical properties and improved functionality, facilitating clinical translation toward therapeutic heart muscle reconstruction., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
31. Two-photon induced collagen cross-linking in bioartificial cardiac tissue.
- Author
-
Kuetemeyer K, Kensah G, Heidrich M, Meyer H, Martin U, Gruh I, and Heisterkamp A
- Subjects
- Animals, Embryo, Mammalian cytology, Fibroblasts drug effects, Fibroblasts metabolism, Indoles metabolism, Lasers, Mice, Microscopy, Fluorescence, Microscopy, Phase-Contrast, Rats, Riboflavin pharmacology, Tomography, Optical, Bioartificial Organs, Collagen pharmacology, Cross-Linking Reagents pharmacology, Heart drug effects, Photons
- Abstract
Cardiac tissue engineering is a promising strategy for regenerative therapies to overcome the shortage of donor organs for transplantation. Besides contractile function, the stiffness of tissue engineered constructs is crucial to generate transplantable tissue surrogates with sufficient mechanical stability to withstand the high pressure present in the heart. Although several collagen cross-linking techniques have proven to be efficient in stabilizing biomaterials, they cannot be applied to cardiac tissue engineering, as cell death occurs in the treated area. Here, we present a novel method using femtosecond (fs) laser pulses to increase the stiffness of collagen-based tissue constructs without impairing cell viability. Raster scanning of the fs laser beam over riboflavin-treated tissue induced collagen cross-linking by two-photon photosensitized singlet oxygen production. One day post-irradiation, stress-strain measurements revealed increased tissue stiffness by around 40% being dependent on the fibroblast content in the tissue. At the same time, cells remained viable and fully functional as demonstrated by fluorescence imaging of cardiomyocyte mitochondrial activity and preservation of active contraction force. Our results indicate that two-photon induced collagen cross-linking has great potential for studying and improving artificially engineered tissue for regenerative therapies., (© 2011 Optical Society of America)
- Published
- 2011
- Full Text
- View/download PDF
32. Rhesus monkey cardiosphere-derived cells for myocardial restoration.
- Author
-
Martens A, Gruh I, Dimitroulis D, Rojas SV, Schmidt-Richter I, Rathert C, Khaladj N, Gawol A, Chikobava MG, Martin U, Haverich A, and Kutschka I
- Subjects
- Animals, Cell Differentiation, Coronary Vessels surgery, Disease Models, Animal, Female, Humans, Macaca mulatta, Mice, Mice, SCID, Myocardial Infarction mortality, Myocardial Infarction pathology, Myocardial Infarction therapy, Myocardium cytology, Stem Cell Transplantation methods
- Abstract
Background Aims: Cardiosphere-derived cells (CDC) have been proposed as a promising myocardial stem cell source for cardiac repair. They have been isolated from human, porcine and rodent cardiac biopsies. However, their usefulness for myocardial restoration remains controversial. We aimed to determine the survival, differentiation and functional effects of Rhesus monkey CDC (RhCDC) in a mouse model of myocardial infarction., Methods: RhCDC were isolated and characterized by flow cytometry and reverse transcriptase (RT)-polymerase chain reaction (PCR) and compared with human CDC. They were injected intramyocardially into severe combined immune deficiency (SCID) beige mice after ligature of the left anterior descending artery (LAD). Phosphate-buffered saline (PBS) served as placebo. Medium treatment alone was used to distinguish between cellular and non-cellular effects. Animals were divided into a non-infarcted control group (n = 7), infarct control groups (n = 24), medium-treated infarct groups (n = 35) and RhCDC-treated infarct groups (n = 33). Follow-up was either 1 or 4 weeks. LV function was assessed by pressure-volume loop analysis. Differentiation was analyzed by immunhistochemical profiling and RT-PCR., Results: Proliferating RhCDC grafts were detected after transplantation in an acute infarct model. RhCDC as well as medium treatment protected myocardium within the infarct area and improved LV function. RhCDC had a superior regenerative effect than medium alone., Conclusions: For the first time, RhCDC have been used for the restoration of infarcted myocardium. RhCDC proliferated in vivo and positively influenced myocardial remodeling. This effect could be mimicked by treatment with unconditioned medium alone, emphasizing a non-cellular paracrine therapeutic mechanism. However, as a robust cardiac stem cell source, CDC might be useful to evoke prolonged paracrine actions in cardiac stem cell therapy.
- Published
- 2011
- Full Text
- View/download PDF
33. A novel miniaturized multimodal bioreactor for continuous in situ assessment of bioartificial cardiac tissue during stimulation and maturation.
- Author
-
Kensah G, Gruh I, Viering J, Schumann H, Dahlmann J, Meyer H, Skvorc D, Bär A, Akhyari P, Heisterkamp A, Haverich A, and Martin U
- Subjects
- Animals, Biomarkers metabolism, Cell Survival, Gene Expression Regulation, Mechanical Phenomena, Microscopy, Fluorescence, Myocardial Contraction, Myocytes, Cardiac cytology, Organ Specificity genetics, Rats, Rats, Sprague-Dawley, Artificial Organs, Bioreactors, Heart physiology, Miniaturization instrumentation, Tissue Engineering instrumentation, Tissue Engineering methods
- Abstract
Stem cell-based cardiac tissue engineering is a promising approach for regenerative therapy of the injured heart. At present, the small number of stem cell-derived cardiomyocytes that can be obtained using current culture and enrichment techniques represents one of the key limitations for the development of functional bioartificial cardiac tissue (BCT). We have addressed this problem by construction of a novel bioreactor with functional features of larger systems that enables the generation and in situ monitoring of miniaturized BCTs. BCTs were generated from rat cardiomyocytes to demonstrate advantages and usefulness of the bioreactor. Tissues showed spontaneous, synchronized contractions with cell orientation along the axis of strain. Cyclic stretch induced cardiomyocyte hypertrophy, demonstrated by a shift of myosin heavy chain expression from the alpha to beta isoform, together with elevated levels of atrial natriuretic factor. Stretch led to a moderate increase in systolic force (1.42 ± 0.09 mN vs. 0.96 ± 0.09 mN in controls), with significantly higher forces observed after β-adrenergic stimulation with noradrenalin (2.54 ± 0.11 mN). Combined mechanical and β-adrenergic stimulation had no synergistic effect. This study demonstrates for the first time that mechanical stimulation and direct real-time contraction force measurement can be combined into a single multimodal bioreactor system, including electrical stimulation of excitable tissue, perfusion of the culture chamber, and the possibility of (fluorescence) microscopic assessment during continuous cultivation. Thus, this bioreactor represents a valuable tool for monitoring tissue development and, ultimately, the optimization of stem cell-based tissue replacement strategies in regenerative medicine.
- Published
- 2011
- Full Text
- View/download PDF
34. Preparation and evaluation of hydrogel-composites from methacrylated hyaluronic acid, alginate, and gelatin for tissue engineering.
- Author
-
Möller L, Krause A, Dahlmann J, Gruh I, Kirschning A, and Dräger G
- Subjects
- Alginates radiation effects, Animals, Animals, Newborn, Cell Adhesion, Cell Survival, Cells, Cultured, Gelatin radiation effects, Hyaluronic Acid analogs & derivatives, Hyaluronic Acid radiation effects, Magnetic Resonance Spectroscopy, Methacrylates radiation effects, Molecular Structure, Myocytes, Cardiac physiology, Rats, Rats, Sprague-Dawley, Rheology, Time Factors, Ultraviolet Rays, Alginates chemical synthesis, Gelatin chemical synthesis, Hyaluronic Acid chemical synthesis, Hydrogels, Methacrylates chemical synthesis, Tissue Engineering methods, Tissue Scaffolds
- Abstract
Hydrogels are three-dimensional water-insoluble hydrophilic natural or synthetic polymer networks made up of crosslinked water-soluble polymers. The purpose of this study was to develop and directly compare photo crosslinked hydrogels on the basis of pure gelatin, alginate and hyaluronic acid as well as their blends. The functionalization of starting materials with methacrylate moieties was evaluated by 1H-NMR spectroscopy. Hydrogels were prepared from methacrylates by photo cross-linking using UV light. The effect of changing the hydrogel composition was quantified through examination of hydrogel swelling behavior and rheological properties. In addition, the viability and adhesion of neonatal rat cardiomyocytes (NRCM) seeded onto the hydrogels was examined by in vivo imaging of NRCM-mediated scaffold contraction as well as by histological evaluation after immunostaining. Biological testing showed good biocompatibility and cell survival in the presence of all materials discussed. Adhesion of cells could only be observed in the presence of gelatin. Blends of gelatin, alginate and hyaluronic acid are promising candidates for the generation of non-toxic, biocompatible hydrogel scaffolds for tissue engineering. Variation of individual compound ratios in the blends can be used for a precise control of mechanical properties and may allow wide-ranging uses in various tissue engineering applications with different mechanical requirements.
- Published
- 2011
- Full Text
- View/download PDF
35. Generation of induced pluripotent stem cells from human cord blood.
- Author
-
Haase A, Olmer R, Schwanke K, Wunderlich S, Merkert S, Hess C, Zweigerdt R, Gruh I, Meyer J, Wagner S, Maier LS, Han DW, Glage S, Miller K, Fischer P, Schöler HR, and Martin U
- Subjects
- Cell Differentiation, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Endothelial Cells cytology, Endothelial Cells metabolism, Humans, Induced Pluripotent Stem Cells metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Patch-Clamp Techniques, Reverse Transcriptase Polymerase Chain Reaction, Fetal Blood cytology, Induced Pluripotent Stem Cells cytology
- Abstract
Induced pluripotent stem cells (iPSCs) may represent an ideal cell source for future regenerative therapies. A critical issue concerning the clinical use of patient-specific iPSCs is the accumulation of mutations in somatic (stem) cells over an organism's lifetime. Acquired somatic mutations are passed onto iPSCs during reprogramming and may be associated with loss of cellular functions and cancer formation. Here we report the generation of human iPSCs from cord blood (CB) as a juvenescent cell source. CBiPSCs show characteristics typical of embryonic stem cells and can be differentiated into derivatives of all three germ layers, including functional cardiomyocytes. For future therapeutic production of autologous and allogeneic iPSC derivatives, CB could be routinely harvested for public and commercial CB banks without any donor risk. CB could readily become available for pediatric patients and, in particular, for newborns with genetic diseases or congenital malformations.
- Published
- 2009
- Full Text
- View/download PDF
36. Transdifferentiation of stem cells: a critical view.
- Author
-
Gruh I and Martin U
- Subjects
- Adult, Adult Stem Cells physiology, Cell Differentiation, Cell Tracking, Hematopoietic Stem Cells physiology, Hepatocytes cytology, Hepatocytes physiology, Humans, Mesenchymal Stem Cells physiology, Molecular Imaging, Muscle Cells cytology, Muscle Cells physiology, Neurons cytology, Neurons physiology, Adult Stem Cells cytology, Cell Transdifferentiation, Hematopoietic Stem Cells cytology, Mesenchymal Stem Cells cytology
- Abstract
Recently a large amount of new data on the plasticity of stem cells of various lineages have emerged, providing new perspectives especially for the therapeutic application of adult stem cells. Previously unknown possibilities of cell differentiation beyond the known commitment of a given stem cell have been described using keywords such as "blood to liver," or "bone to brain." Controversies on the likelihood, as well as the biological significance, of these conversions almost immediately arose within this young field of stem cell biology. This chapter will concentrate on these controversies and focus on selected examples demonstrating the technical aspects of stem cell transdifferentiation and the evaluation of the tools used to analyze these events.
- Published
- 2009
- Full Text
- View/download PDF
37. Serum-free differentiation of murine embryonic stem cells into alveolar type II epithelial cells.
- Author
-
Winkler ME, Mauritz C, Groos S, Kispert A, Menke S, Hoffmann A, Gruh I, Schwanke K, Haverich A, and Martin U
- Subjects
- Algorithms, Animals, Cell Culture Techniques, Cell Differentiation genetics, Cells, Cultured, Culture Media, Serum-Free pharmacology, Embryonic Stem Cells metabolism, Embryonic Stem Cells physiology, Embryonic Stem Cells ultrastructure, Epithelial Cells metabolism, Epithelial Cells physiology, Epithelial Cells ultrastructure, Gene Expression Profiling, Gene Expression Regulation drug effects, Mice, Pulmonary Alveoli metabolism, Pulmonary Alveoli physiology, Pulmonary Alveoli ultrastructure, Pulmonary Surfactant-Associated Protein B metabolism, Pulmonary Surfactant-Associated Protein C metabolism, Respiratory Mucosa drug effects, Respiratory Mucosa physiology, Serum physiology, Time Factors, Cell Differentiation drug effects, Embryonic Stem Cells drug effects, Epithelial Cells drug effects, Pulmonary Alveoli drug effects
- Abstract
Alveolar type II (AT2) epithelial cells have important functions including the production of surfactant and regeneration of lost alveolar type I epithelial cells. The ability of in vitro production of AT2 cells would offer new therapeutic options in treating pulmonary injuries and disorders including genetically based surfactant deficiencies. Aiming at the generation of AT2-like cells, the differentiation of murine embryonic stem cells (mESCs) toward mesendodermal progenitors (MEPs) was optimized using a "Brachyury-eGFP-knock in" mESC line. eGFP expression demonstrated generation of up to 65% MEPs at day 4 after formation of embryoid bodies (EBs) under serum-free conditions. Plated EBs were further differentiated into AT2-like cells for a total of 25 days in serum-free media resulting in the expression of endodermal marker genes (FoxA2, Sox17, TTR, TTF-1) and of markers for distal lung epithelium (surfactant proteins (SP-) A, B, C, and D, CCSP, aquaporin 5). Notably, expression of SP-C as the only known AT2 cell specific marker could be detected after serum-induction as well as under serum-free conditions. Cytoplasmic localization of SP-C was demonstrated by confocal microscopy. The presence of AT2-like cells was confirmed by electron microscopy providing evidence for polarized cells with apical microvilli and lamellar body-like structures. Our results demonstrate the differentiation of AT2-like cells from mESCs after serum-induction and under serum-free conditions. The established serum-free differentiation protocol will facilitate the identification of key differentiation factors leading to a more specific and effective generation of AT2-like cells from ESCs.
- Published
- 2008
- Full Text
- View/download PDF
38. Type II pneumocyte-restricted green fluorescent protein expression after lentiviral transduction of lung epithelial cells.
- Author
-
Wunderlich S, Gruh I, Winkler ME, Beier J, Radtke K, Schmiedl A, Groos S, Haverich A, and Martin U
- Subjects
- Animals, Cells, Cultured, Epithelial Cells metabolism, Green Fluorescent Proteins genetics, Humans, Mice, Phenotype, Promoter Regions, Genetic, Rats, Transduction, Genetic, Transgenes, Genetic Vectors, Green Fluorescent Proteins metabolism, Lentivirus genetics, Pulmonary Alveoli cytology, Pulmonary Alveoli metabolism, Pulmonary Surfactant-Associated Protein C genetics
- Abstract
Type II alveolar epithelial (AT2) cell-specific reporter expression has been highly useful in the study of embryology and alveolar regeneration in transgenic mice. Technologies enabling efficient gene transfer and cell type-restricted transgene expression in AT2 cells would allow for correction of AT2 cell-based diseases such as genetic surfactant deficiencies. Moreover, such approaches are urgently required to investigate differentiation of AT2 cells from adult and embryonic stem cells of other species than mouse. Using a human surfactant protein C (SP-C) promoter fragment, we have constructed lentiviral vectors enabling AT2-restricted transgene expression and identification of stem cell-derived AT2 cells. Lung epithelial cell lines M3E3/C3, H441, RLE-6TN, A549, MLE-12, and MLE-15 were characterized at the molecular and ultrastructural levels to identify cell lines useful to assess the cell type specificity of our vector constructs. After transduction, no green fluorescent protein (GFP) expression was observed in nontarget cells including bronchial H441 cells, pulmonary A549 cells, fibroblasts, smooth muscle cells, and endothelial cells. In contrast, and in correlation with endogenous SP-C expression, lentiviral transduction resulted in stable GFP expression in MLE-12 and MLE-15 AT2 cells. In conclusion, we have constructed a lentiviral vector mediating SP-C promoter-dependent GFP expression. Transgene expression strictly corresponds with an AT2 phenotype of the transduced cells. In particular, the generated vector should facilitate local alveolar gene therapy and investigation of alveolar regeneration and stem cell differentiation.
- Published
- 2008
- Full Text
- View/download PDF
39. Human CMV immediate-early enhancer: a useful tool to enhance cell-type-specific expression from lentiviral vectors.
- Author
-
Gruh I, Wunderlich S, Winkler M, Schwanke K, Heinke J, Blömer U, Ruhparwar A, Rohde B, Li RK, Haverich A, and Martin U
- Subjects
- Animals, Atrial Natriuretic Factor genetics, Atrial Natriuretic Factor metabolism, Cells, Cultured, HeLa Cells, Humans, Lentivirus physiology, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, Organ Specificity, Promoter Regions, Genetic genetics, Pulmonary Surfactant-Associated Protein C genetics, Pulmonary Surfactant-Associated Protein C metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Rats, Wistar, Transgenes, Virion genetics, Virus Assembly, Cytomegalovirus genetics, Enhancer Elements, Genetic genetics, Gene Expression Regulation, Genetic Vectors genetics, Lentivirus genetics
- Abstract
Background: Lentiviral vectors are attractive delivery tools for gene therapy, especially in terminally differentiated target cells. While restriction of gene expression to specific cell populations is of particular importance, highly efficient cell-type-specific gene expression after viral gene transfer so far has been hampered by low levels of transgene expression., Methods: Addressing this problem, we have integrated the human cytomegalovirus (CMV) immediate-early enhancer into an 'advanced' generation lentiviral vector. Expression cassettes with the reporter gene green fluorescent protein (GFP), combined with the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) under control of a ubiquitous phosphoglycerate kinase (mouse PGK), cardiomyocyte- (human atrial natriuretic factor (ANF), human ventricular myosin light chain (MLC2v)), or type II alveolar epithelial cell (AT-2)-specific human surfactant protein C (SP-C) promoter, were introduced. As insertion of an enhancing element can interfere with the promoter's specificity, expression levels conferred by our enhancer/promoter constructs were evaluated in target and non-target cells., Results: Transduction of target cells with human CMV enhancer containing lentiviral vectors resulted in a multiple-log increase in GFP expression compared to corresponding vectors lacking the human CMV enhancer. In the case of the ANF, the MLC2v, and the SP-C promoters, tissue-specific reporter gene expression in cardiomyocytes and in lung AT-2 cells was maintained, as expression in non-target cells increased only up to 7-fold., Conclusions: The results of this study indicate that lentiviral vectors with the human CMV enhancer conferring efficient cell-type-specific gene expression may be useful tools for gene therapy purposes or cell tracing, e.g. to analyze stem cell differentiation in transplantation and co-culture settings., ((c) 2007 John Wiley & Sons, Ltd.)
- Published
- 2008
- Full Text
- View/download PDF
40. Enrichment of cardiac pacemaker-like cells: neuregulin-1 and cyclic AMP increase I(f)-current density and connexin 40 mRNA levels in fetal cardiomyocytes.
- Author
-
Ruhparwar A, Er F, Martin U, Radke K, Gruh I, Niehaus M, Karck M, Haverich A, and Hoppe UC
- Subjects
- Animals, Biomarkers analysis, Blotting, Northern methods, Cell Differentiation, Cells, Cultured, Connexin 43 metabolism, Connexins genetics, Flow Cytometry, Gene Expression drug effects, Mice, Mice, Inbred Strains, Patch-Clamp Techniques, RNA, Messenger analysis, RNA, Messenger metabolism, Gap Junction alpha-5 Protein, Connexins metabolism, Cyclic AMP pharmacology, Embryonic Stem Cells metabolism, Myocytes, Cardiac metabolism, Neuregulin-1 pharmacology, Potassium Channels metabolism
- Abstract
Generation of a large number of cells belonging to the cardiac pacemaker system would constitute an important step towards their utilization as a biological cardiac pacemaker system. The aim of the present study was to identify factors, which might induce transformation of a heterogenous population of fetal cardiomyocytes into cells with a pacemaker-like phenotype. Neuregulin-1 (alpha- and beta-isoform) or the cAMP was added to fresh cell cultures of murine embryonic cardiomyocytes. Quantitative northern blot analysis and flowcytometry were performed to detect the expression of connexins 40, 43 and 45. Patch clamp recordings in the whole cell configuration were performed to determine current density of I (f), a characteristic ion current of pacemaker cells. Fetal cardiomyocytes without supplement of neuregulin or cAMP served as control group. Neuregulin and cAMP significantly increased mRNA levels of connexin 40 (Cx-40), a marker of the early differentiating conduction system in mice. On the protein level, flowcytometry revealed no significant differences between treated and untreated groups with regard to the expression of connexins 40, 43 and 45. Treatment with cAMP (11.2 +/- 2.24 pA/pF; P < 0.001) and neuregulin-1-beta (6.23 +/- 1.07 pA/pF; P < 0.001) significantly increased the pacemaker current density compared to control cardiomyocytes (1.76 +/- 0.49 pA/pF). Our results indicate that neuregulin-1 and cAMP possess the capacity to cause significant transformation of a mixed population of fetal cardiomyocytes into cardiac pacemaker-like cells as shown by electrophysiology and increase of Cx-40 mRNA. This method may allow the development of a biological cardiac pacemaker system when applied to adult or embryonic stem cells.
- Published
- 2007
- Full Text
- View/download PDF
41. No evidence of transdifferentiation of human endothelial progenitor cells into cardiomyocytes after coculture with neonatal rat cardiomyocytes.
- Author
-
Gruh I, Beilner J, Blomer U, Schmiedl A, Schmidt-Richter I, Kruse ML, Haverich A, and Martin U
- Subjects
- Animals, Animals, Newborn, Coculture Techniques, Flow Cytometry, Humans, Immunophenotyping, Microscopy, Confocal, Microscopy, Fluorescence, Rats, Cell Differentiation, Endothelial Cells cytology, Myocytes, Cardiac cytology, Stem Cells cytology
- Abstract
Background: Recent studies have suggested the differentiation of human endothelial progenitor cells (huEPCs) isolated from peripheral blood into cardiomyocytes. This study investigates whether, when cocultured, neonatal rat cardiomyocytes (NRCMs) can induce transdifferentiation of huEPCs into cardiomyocytes., Methods and Results: Coculture experiments with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine (DiI)-labeled huEPCs and NRCMs have been performed. Cocultures have been analyzed by means of flow cytometry, 3D confocal laser microscopy, species-specific reverse transcriptase-polymerase chain reaction for the expression of human cardiac marker genes, and electron microscopy. Although fluorescence-activated cell sorting (FACS) analysis and conventional wide-field fluorescence microscopy suggested the existence of DiIpos cardiomyocytes in cocultures, no convincing evidence of cardiac differentiation of huEPCs has been obtained. Apparently, DiIpos cardiomyocytes were identified as necrotic NRCMs or NRCM-derived vesicles with high levels of autofluorescence or, alternatively, as NRCMs lying on top of or below labeled huEPCs or huEPC fragments. Accordingly, no expression of human Nkx2.5, GATA-4, or cardiac troponin I was detected., Conclusions: No convincing evidence of transdifferentiation of huEPCs into cardiomyocytes was obtained. Although we cannot exclude that recent contrary data may be due to slightly different culture protocols, our study has revealed that recently applied standard analysis tools including FACS and wide-field fluorescence microscopy are not sufficient to demonstrate transdifferentiation in coculture settings and can lead to misinterpretation of the data obtained solely with these methods.
- Published
- 2006
- Full Text
- View/download PDF
42. Shuttle system allowing simplified cloning of expression cassettes into advanced generation lentiviral vectors.
- Author
-
Gruh I, Schwanke K, Wunderlich S, Blömer U, Scherr M, Ganser A, Haverich A, and Martin U
- Subjects
- Base Sequence, Cell Line, Cytomegalovirus genetics, Endonucleases chemistry, Escherichia coli genetics, Flow Cytometry, Gene Expression, Genetic Vectors chemistry, Green Fluorescent Proteins metabolism, HIV Long Terminal Repeat genetics, Humans, Luminescent Proteins metabolism, Models, Genetic, Plasmids genetics, Transduction, Genetic, Red Fluorescent Protein, Cloning, Molecular, Genetic Engineering, Genetic Vectors genetics, Lentivirus genetics
- Published
- 2005
- Full Text
- View/download PDF
43. Shuttle of lentiviral vectors via transplanted cells in vivo.
- Author
-
Blömer U, Gruh I, Witschel H, Haverich A, and Martin U
- Subjects
- Animals, Cell Line, Fibroblasts virology, Genetic Therapy methods, Genetic Vectors genetics, Humans, Immunohistochemistry, Myocytes, Cardiac virology, Rats, Rats, Wistar, Transduction, Genetic methods, Cell Transplantation, Genetic Therapy adverse effects, Genetic Vectors administration & dosage, Lentivirus genetics, Lentivirus Infections transmission
- Abstract
Lentiviral vectors have turned out to be an efficient method for stable gene transfer in vitro and in vivo. Not only do fields of application include cell marking and tracing following transplantation in vivo, but also the stable delivery of biological active proteins for gene therapy. A variety of cells, however, need immediate transplantation after preparation, for example, to prevent cell death, differentiation or de-differentiation. Although these cells are usually washed several times following lentiviral transduction, there may be the risk of viral vector shuttle via transplanted cells resulting in undesired in vivo transduction of recipient cells. We investigated whether infectious lentiviral particles are transmitted via ex vivo lentivirally transduced cells. To this end, we explored potential viral shuttle via ex vivo lentivirally transduced cardiomyocytes in vitro and following transplantation into the brain and peripheral muscle. We demonstrate that, even after extensive washing, infectious viral vector particles can be detected in cell suspensions. Those lentiviral vector particles were able to transduce target cells in transwell experiments. Moreover, transmitted vector particles stably transduced resident cells of the recipient central nervous system and muscle in vivo. Our results of lentiviral vector shuttle via transduced cardiomyocytes are significant for both ex vivo gene therapy and for lentiviral cell tracing, in particular for investigation of stem cell differentiation in transplantation models and co-cultivation systems.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.