ijms-logo

Journal Browser

Journal Browser

Cell Programming for Cardiovascular Disease Modeling and Therapy

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (31 March 2021) | Viewed by 35185

Special Issue Editor


E-Mail Website
Guest Editor
1. University Medical Center Rostock, Clinic for Cardiac Surgery, University of Rostock, Rostock, Germany
2. Dept. Life, Light & Matter, Interdisc. Faculty, University of Rostock, Rostock, Germany
Interests: cardiovascular diseases; iPSCs (induced pluripotent stem cells); ESCs adult stem cells; autologous cell therapy; cardiovascular organoids; cardiovascular disease modeling; drug discovery; direct reprogramming; forward programming; cardiovascular repair; cell targeting
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Cardiovascular diseases still represent a leading cause of mortality in the developed countries with highly limited therapy options. A main reason is the very limited regeneration potential of collapsed cardiomyocytes – therefore novel approaches toward personalized regenerative therapy and drug development are of major importance. In recent years, forward programming of iPSCs as well as Direct Reprogramming of somatic cells and adult stem cells have introduced entirely novel options to circumvent obstacles commonly encountered in regenerative medicine by utilizing autologous cells as the source of treatment. This has greatly benefitted from efforts to identify and optimize master regulator combinations to redefine cellular fates. Multiple research groups have shown direct somatic cell conversion towards cardiovascular cells, thereby avoiding a pluripotent intermediate state. Moreover, in vitro test systems based on organoid cultures derived from patient specific programmed cardiovascular cells are being developed which will enable personalized drug testing in precision medicine. In this Special Issue, we are aiming to broadly adress topics from understanding the basic science of somatic and stem cell reprogramming to their applications in cardiovascular regeneration and disease treatment as well as in vitro disease modeling approaches.

The current Special Issue will accept original studies, reviews and technical reports in the field of cardiovascular cell programming, disease modeling and cell based therapy, written by scientists active in the field.

Dr. David Robert
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Cardiovascular diseases
  • iPSCs (induced pluripotent stem cells)
  • ESCs
  • Adult Stem Cells
  • Autologous cell therapy
  • Cardiovascular organoids
  • Cardiovascular disease modeling
  • Drug discovery
  • Direct Reprogramming
  • Forward Programming
  • Cardiovascular Repair
  • Cell Targeting

Published Papers (10 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

4 pages, 177 KiB  
Editorial
Cell Programming for Cardiovascular Disease Modeling and Therapy
by Robert David
Int. J. Mol. Sci. 2023, 24(9), 7877; https://doi.org/10.3390/ijms24097877 - 26 Apr 2023
Viewed by 790
Abstract
Cardiovascular diseases have a high mortality due to a very limited regenerative potential of lost cardiomyocytes and therefore are one of the leading causes of death in developed countries [...] Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)

Research

Jump to: Editorial, Review

20 pages, 6385 KiB  
Article
Dual Function of iPSC-Derived Pericyte-Like Cells in Vascularization and Fibrosis-Related Cardiac Tissue Remodeling In Vitro
by Monika Szepes, Anna Melchert, Julia Dahlmann, Jan Hegermann, Christopher Werlein, Danny Jonigk, Axel Haverich, Ulrich Martin, Ruth Olmer and Ina Gruh
Int. J. Mol. Sci. 2020, 21(23), 8947; https://doi.org/10.3390/ijms21238947 - 25 Nov 2020
Cited by 15 | Viewed by 3393
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, [...] Read more.
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. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Figure 1

20 pages, 4491 KiB  
Article
Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid Improves Energetic Status and Cardiomyogenic Differentiation of Human Dilated Myocardium-Derived Primary Mesenchymal Cells
by Rokas Miksiunas, Kestutis Rucinskas, Vilius Janusauskas, Siegfried Labeit and Daiva Bironaite
Int. J. Mol. Sci. 2020, 21(14), 4845; https://doi.org/10.3390/ijms21144845 - 8 Jul 2020
Cited by 5 | Viewed by 2226
Abstract
Background. In this study the effect of histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) on the energetic status and cardiomyogenic differentiation of human healthy and dilated myocardium-derived mesenchymal stromal cells (hmMSC) have been investigated. Methods. The hmMSC were isolated from the healthy [...] Read more.
Background. In this study the effect of histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) on the energetic status and cardiomyogenic differentiation of human healthy and dilated myocardium-derived mesenchymal stromal cells (hmMSC) have been investigated. Methods. The hmMSC were isolated from the healthy and dilated post-operation heart biopsies by explant outgrowth method. Cell proliferation, HDAC activity, mitochondrial membrane potential, and level of adenosine triphosphate (ATP) were evaluated. The effect of SAHA on mitochondrial parameters has been investigated also by Seahorse XF analyzer and cardiomyogenic differentiation was confirmed by the expression of transcription factor NK2 Homeobox 5 (Nkx2.5), cardiac troponin T and alpha cardiac actin at gene and protein levels. Results. Dilated myocardium-derived hmMSC had almost 1.5 folds higher HDAC activity compared to the healthy cells and significantly lower mitochondrial membrane potential and ATP level. HDAC class I and II inhibitor SAHA improved energetic status of mitochondria in dilated myocardium-isolated hmMSC and increased expression of cardiac specific proteins during 14 days of exposure of cells to SAHA. Conclusions. HDAC inhibitor SAHA can be a promising therapeutic for dilated cardiomyopathy (DCM). Dilated hmMSC exposed to SAHA improved energetic status and, subsequently, cardiomyogenic differentiation. Data suggest that human dilated myocardium-derived MSC still have cardio tissue regenerative potential, which might be stimulated by HDAC inhibitors. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Figure 1

9 pages, 5138 KiB  
Article
18F-FDG PET-Based Imaging of Myocardial Inflammation Following Acute Myocardial Infarction in a Mouse Model
by Praveen Vasudevan, Ralf Gäbel, Jan Stenzel, Joanna Förster, Jens Kurth, Brigitte Vollmar, Bernd Joachim Krause, Hüseyin Ince, Robert David and Cajetan Immanuel Lang
Int. J. Mol. Sci. 2020, 21(9), 3340; https://doi.org/10.3390/ijms21093340 - 8 May 2020
Cited by 8 | Viewed by 2912
Abstract
Cellular inflammation is an integral part of the healing process following acute myocardial infarction and has been under intense investigation for both therapeutic and prognostic approaches. Monocytes and macrophages are metabolically highly active and show increased uptake rates of glucose and its analog, [...] Read more.
Cellular inflammation is an integral part of the healing process following acute myocardial infarction and has been under intense investigation for both therapeutic and prognostic approaches. Monocytes and macrophages are metabolically highly active and show increased uptake rates of glucose and its analog, 18F-FDG. Yet, the specific allocation of the radioactivity to the inflammatory cells via positron emission tomography (PET) imaging requires the suppression of glucose metabolism in viable myocardium. In mice, the most important model organism in basic research, this can be achieved by the application of ketamine/xylazine (KX) for anesthesia instead of isoflurane. Yet, while the consensus exists that glucose metabolism is effectively suppressed, a strategy for reproducible image analysis is grossly lacking and causes uncertainty concerning data interpretation. We introduce a simple strategy for systematic image analysis, which is a prerequisite to evaluate therapies targeting myocardial inflammation. Mice underwent permanent occlusion of the left anterior descending artery (LAD), inducing an acute myocardial infarction (MI). Five days after MI induction, 10MBq 18F-FDG was injected intravenously and a static PET/CT scan under ketamine/xylazine anesthesia was performed. For image reconstruction, we used an algorithm based on three-dimensional ordered subsets expectation maximization (3D-OSEM) followed by three-dimensional ordinary Poisson maximum a priori (MAP) reconstruction. Using this approach, high focal tracer uptake was typically located in the border zone of the infarct by visual inspection. To precisely demarcate the border zone for reproducible volume of interest (VOI) positioning, our protocol relies on positioning VOIs around the whole left ventricle, the inferobasal wall and the anterolateral wall guided by anatomical landmarks. This strategy enables comparable data in mouse studies, which is an important prerequisite for using a PET-based assessment of myocardial inflammation as a prognostic tool in therapeutic applications. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Figure 1

10 pages, 1530 KiB  
Article
Quantitative Evaluation of the Sarcomere Network of Human hiPSC-Derived Cardiomyocytes Using Single-Molecule Localization Microscopy
by Heiko Lemcke, Anna Skorska, Cajetan Immanuel Lang, Lisa Johann and Robert David
Int. J. Mol. Sci. 2020, 21(8), 2819; https://doi.org/10.3390/ijms21082819 - 17 Apr 2020
Cited by 10 | Viewed by 3340
Abstract
The maturation of iPSC-derived cardiomyocytes is still a critical point for their application in cardiovascular research as well as for their clinical use. Although multiple differentiation protocols have been established, researchers failed to generate fully mature cardiomyocytes in vitro possessing identical phenotype-related and [...] Read more.
The maturation of iPSC-derived cardiomyocytes is still a critical point for their application in cardiovascular research as well as for their clinical use. Although multiple differentiation protocols have been established, researchers failed to generate fully mature cardiomyocytes in vitro possessing identical phenotype-related and functional properties as their native adult counterparts. Besides electrophysiological and metabolic changes, the establishment of a well structured sarcomere network is important for the development of a mature cardiac phenotype. Here, we present a super resolution-based approach to quantitatively evaluate the structural maturation of iPSC-derived cardiomyocytes. Fluorescence labelling of the α-actinin cytoskeleton and subsequent visualization by photoactivated localization microscopy allows the acquisition of highly resolved images for measuring sarcomere length and z-disc thickness. Our image analysis revealed that iPSC and neonatal cardiomyocyte share high similarity with respect to their sarcomere organization, however, contraction capacity was inferior in iPSC-derived cardiac cells, indicating an early maturation level. Moreover, we demonstrate that this imaging approach can be used as a tool to monitor cardiomyocyte integrity, helping to optimize iPSC differentiation as well as somatic cell direct-reprogramming strategies. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

22 pages, 761 KiB  
Review
Xenogeneic and Stem Cell-Based Therapy for Cardiovascular Diseases: Genetic Engineering of Porcine Cells and Their Applications in Heart Regeneration
by Anne-Marie Galow, Tom Goldammer and Andreas Hoeflich
Int. J. Mol. Sci. 2020, 21(24), 9686; https://doi.org/10.3390/ijms21249686 - 18 Dec 2020
Cited by 6 | Viewed by 4257
Abstract
Cardiovascular diseases represent a major health concern worldwide with few therapy options for ischemic injuries due to the limited regeneration potential of affected cardiomyocytes. Innovative cell replacement approaches could facilitate efficient regenerative therapy. However, despite extensive attempts to expand primary human cells in [...] Read more.
Cardiovascular diseases represent a major health concern worldwide with few therapy options for ischemic injuries due to the limited regeneration potential of affected cardiomyocytes. Innovative cell replacement approaches could facilitate efficient regenerative therapy. However, despite extensive attempts to expand primary human cells in vitro, present technological limitations and the lack of human donors have so far prevented their broad clinical use. Cell xenotransplantation might provide an ethically acceptable unlimited source for cell replacement therapies and bridge the gap between waiting recipients and available donors. Pigs are considered the most suitable candidates as a source for xenogeneic cells and tissues due to their anatomical and physiological similarities with humans. The potential of porcine cells in the field of stem cell-based therapy and regenerative medicine is under intensive investigation. This review outlines the current progress and highlights the most promising approaches in xenogeneic cell therapy with a focus on the cardiovascular system. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Graphical abstract

30 pages, 1712 KiB  
Review
The Future of Direct Cardiac Reprogramming: Any GMT Cocktail Variety?
by Leyre López-Muneta, Josu Miranda-Arrubla and Xonia Carvajal-Vergara
Int. J. Mol. Sci. 2020, 21(21), 7950; https://doi.org/10.3390/ijms21217950 - 26 Oct 2020
Cited by 11 | Viewed by 4465
Abstract
Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which [...] Read more.
Direct cardiac reprogramming has emerged as a novel therapeutic approach to treat and regenerate injured hearts through the direct conversion of fibroblasts into cardiac cells. Most studies have focused on the reprogramming of fibroblasts into induced cardiomyocytes (iCMs). The first study in which this technology was described, showed that at least a combination of three transcription factors, GATA4, MEF2C and TBX5 (GMT cocktail), was required for the reprogramming into iCMs in vitro using mouse cells. However, this was later demonstrated to be insufficient for the reprogramming of human cells and additional factors were required. Thereafter, most studies have focused on implementing reprogramming efficiency and obtaining fully reprogrammed and functional iCMs, by the incorporation of other transcription factors, microRNAs or small molecules to the original GMT cocktail. In this respect, great advances have been made in recent years. However, there is still no consensus on which of these GMT-based varieties is best, and robust and highly reproducible protocols are still urgently required, especially in the case of human cells. On the other hand, apart from CMs, other cells such as endothelial and smooth muscle cells to form new blood vessels will be fundamental for the correct reconstruction of damaged cardiac tissue. With this aim, several studies have centered on the direct reprogramming of fibroblasts into induced cardiac progenitor cells (iCPCs) able to give rise to all myocardial cell lineages. Especially interesting are reports in which multipotent and highly expandable mouse iCPCs have been obtained, suggesting that clinically relevant amounts of these cells could be created. However, as of yet, this has not been achieved with human iCPCs, and exactly what stage of maturity is appropriate for a cell therapy product remains an open question. Nonetheless, the major concern in regenerative medicine is the poor retention, survival, and engraftment of transplanted cells in the cardiac tissue. To circumvent this issue, several cell pre-conditioning approaches are currently being explored. As an alternative to cell injection, in vivo reprogramming may face fewer barriers for its translation to the clinic. This approach has achieved better results in terms of efficiency and iCMs maturity in mouse models, indicating that the heart environment can favor this process. In this context, in recent years some studies have focused on the development of safer delivery systems such as Sendai virus, Adenovirus, chemical cocktails or nanoparticles. This article provides an in-depth review of the in vitro and in vivo cardiac reprograming technology used in mouse and human cells to obtain iCMs and iCPCs, and discusses what challenges still lie ahead and what hurdles are to be overcome before results from this field can be transferred to the clinical settings. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Figure 1

18 pages, 1684 KiB  
Review
Strategies and Challenges to Improve Cellular Programming-Based Approaches for Heart Regeneration Therapy
by Lin Jiang, Jialiang Liang, Wei Huang, Zhichao Wu, Christian Paul and Yigang Wang
Int. J. Mol. Sci. 2020, 21(20), 7662; https://doi.org/10.3390/ijms21207662 - 16 Oct 2020
Cited by 4 | Viewed by 3423
Abstract
Limited adult cardiac cell proliferation after cardiovascular disease, such as heart failure, hampers regeneration, resulting in a major loss of cardiomyocytes (CMs) at the site of injury. Recent studies in cellular reprogramming approaches have provided the opportunity to improve upon previous techniques used [...] Read more.
Limited adult cardiac cell proliferation after cardiovascular disease, such as heart failure, hampers regeneration, resulting in a major loss of cardiomyocytes (CMs) at the site of injury. Recent studies in cellular reprogramming approaches have provided the opportunity to improve upon previous techniques used to regenerate damaged heart. Using these approaches, new CMs can be regenerated from differentiation of iPSCs (similar to embryonic stem cells), the direct reprogramming of fibroblasts [induced cardiomyocytes (iCMs)], or induced cardiac progenitors. Although these CMs have been shown to functionally repair infarcted heart, advancements in technology are still in the early stages of development in research laboratories. In this review, reprogramming-based approaches for generating CMs are briefly introduced and reviewed, and the challenges (including low efficiency, functional maturity, and safety issues) that hinder further translation of these approaches into a clinical setting are discussed. The creative and combined optimal methods to address these challenges are also summarized, with optimism that further investigation into tissue engineering, cardiac development signaling, and epigenetic mechanisms will help to establish methods that improve cell-reprogramming approaches for heart regeneration. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Graphical abstract

18 pages, 962 KiB  
Review
Preconditioned and Genetically Modified Stem Cells for Myocardial Infarction Treatment
by Kamila Raziyeva, Aiganym Smagulova, Yevgeniy Kim, Saltanat Smagul, Ayan Nurkesh and Arman Saparov
Int. J. Mol. Sci. 2020, 21(19), 7301; https://doi.org/10.3390/ijms21197301 - 2 Oct 2020
Cited by 33 | Viewed by 4315
Abstract
Ischemic heart disease and myocardial infarction remain leading causes of mortality worldwide. Existing myocardial infarction treatments are incapable of fully repairing and regenerating the infarcted myocardium. Stem cell transplantation therapy has demonstrated promising results in improving heart function following myocardial infarction. However, poor [...] Read more.
Ischemic heart disease and myocardial infarction remain leading causes of mortality worldwide. Existing myocardial infarction treatments are incapable of fully repairing and regenerating the infarcted myocardium. Stem cell transplantation therapy has demonstrated promising results in improving heart function following myocardial infarction. However, poor cell survival and low engraftment at the harsh and hostile environment at the site of infarction limit the regeneration potential of stem cells. Preconditioning with various physical and chemical factors, as well as genetic modification and cellular reprogramming, are strategies that could potentially optimize stem cell transplantation therapy for clinical application. In this review, we discuss the most up-to-date findings related to utilizing preconditioned stem cells for myocardial infarction treatment, focusing mainly on preconditioning with hypoxia, growth factors, drugs, and biological agents. Furthermore, genetic manipulations on stem cells, such as the overexpression of specific proteins, regulation of microRNAs, and cellular reprogramming to improve their efficiency in myocardial infarction treatment, are discussed as well. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Graphical abstract

27 pages, 960 KiB  
Review
Human Cell Modeling for Cardiovascular Diseases
by Melania Lippi, Ilaria Stadiotti, Giulio Pompilio and Elena Sommariva
Int. J. Mol. Sci. 2020, 21(17), 6388; https://doi.org/10.3390/ijms21176388 - 2 Sep 2020
Cited by 13 | Viewed by 5280
Abstract
The availability of appropriate and reliable in vitro cell models recapitulating human cardiovascular diseases has been the aim of numerous researchers, in order to retrace pathologic phenotypes, elucidate molecular mechanisms, and discover therapies using simple and reproducible techniques. In the past years, several [...] Read more.
The availability of appropriate and reliable in vitro cell models recapitulating human cardiovascular diseases has been the aim of numerous researchers, in order to retrace pathologic phenotypes, elucidate molecular mechanisms, and discover therapies using simple and reproducible techniques. In the past years, several human cell types have been utilized for these goals, including heterologous systems, cardiovascular and non-cardiovascular primary cells, and embryonic stem cells. The introduction of induced pluripotent stem cells and their differentiation potential brought new prospects for large-scale cardiovascular experiments, bypassing ethical concerns of embryonic stem cells and providing an advanced tool for disease modeling, diagnosis, and therapy. Each model has its advantages and disadvantages in terms of accessibility, maintenance, throughput, physiological relevance, recapitulation of the disease. A higher level of complexity in diseases modeling has been achieved with multicellular co-cultures. Furthermore, the important progresses reached by bioengineering during the last years, together with the opportunities given by pluripotent stem cells, have allowed the generation of increasingly advanced in vitro three-dimensional tissue-like constructs mimicking in vivo physiology. This review provides an overview of the main cell models used in cardiovascular research, highlighting the pros and cons of each, and describing examples of practical applications in disease modeling. Full article
(This article belongs to the Special Issue Cell Programming for Cardiovascular Disease Modeling and Therapy)
Show Figures

Figure 1

Back to TopTop