Review Papers in (Re)programming Cells for Cardiac Repair

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cells of the Cardiovascular System".

Deadline for manuscript submissions: 15 June 2024 | Viewed by 10469

Special Issue Editors


E-Mail Website
Guest Editor
College of Medicine, University of Cincinnati, Cincinnati, OH, USA
Interests: cardiac repair; stem cell therapy; cell reprogramming

E-Mail
Guest Editor
Botnar Research Centre, Nuffield Department of Orthopaedics, University of Oxford, Rheumatology and Musculoskeletal Sciences Old Road, Headington, Oxford OX3 7LD, UK
Interests: 3D genomics; epigenetics; cardiac repair; stem cell therapy; exosomes

Special Issue Information

Dear Colleagues,

Cardiac disease is the leading cause of death worldwide due to the failure of the adult human heart to replenish the considerable loss of cardiomyocytes caused by various insults (e.g., ischemia). Regenerative medicine is an innovative approach to repair and replace damaged heart cells and is undergoing a major revolution due to the unprecedented need for therapeutics to treat cardiac patients around the world.

Induced pluripotent stem cells (iPSCs) have been widely studied in the field of cardiac regeneration and are considered one of the most promising candidate therapeutics. However, several issues remain to be resolved prior to clinical application, such as immaturity of iPSC-derived cardiomyocytes, poor survival of transplanted cells in the injured heart, and how we can best employ cardiac tissue engineering technologies to optimize repair. This Special Issue aims to publish high-quality review articles that provide novel insights and conceptual advancements in the field of cardiac (re)programming for cardiac repair, in particular in terms of the signaling pathways and transcriptional/epigenetic regulation for cardiac lineage commitment. We kindly encourage all research groups covering relevant areas within the issue’s scope to contribute up-to-date, full-length comprehensive reviews, highlighting the latest developments in their research field, or to invite relevant experts and colleagues to do so.

Prof. Dr. Yigang Wang
Dr. Yuliang Feng
Guest Editors

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. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). 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

  • cardiac programming
  • cardiogenesis
  • transcriptional regulation
  • epigenetics
  • cardiac tissue engineering

Published Papers (4 papers)

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

Review

21 pages, 1121 KiB  
Review
Cardiomyocyte Ploidy, Metabolic Reprogramming and Heart Repair
by Andrea Elia, Sadia Mohsin and Mohsin Khan
Cells 2023, 12(12), 1571; https://doi.org/10.3390/cells12121571 - 7 Jun 2023
Cited by 2 | Viewed by 1797
Abstract
The adult heart is made up of cardiomyocytes (CMs) that maintain pump function but are unable to divide and form new myocytes in response to myocardial injury. In contrast, the developmental cardiac tissue is made up of proliferative CMs that regenerate injured myocardium. [...] Read more.
The adult heart is made up of cardiomyocytes (CMs) that maintain pump function but are unable to divide and form new myocytes in response to myocardial injury. In contrast, the developmental cardiac tissue is made up of proliferative CMs that regenerate injured myocardium. In mammals, CMs during development are diploid and mononucleated. In response to cardiac maturation, CMs undergo polyploidization and binucleation associated with CM functional changes. The transition from mononucleation to binucleation coincides with unique metabolic changes and shift in energy generation. Recent studies provide evidence that metabolic reprogramming promotes CM cell cycle reentry and changes in ploidy and nucleation state in the heart that together enhances cardiac structure and function after injury. This review summarizes current literature regarding changes in CM ploidy and nucleation during development, maturation and in response to cardiac injury. Importantly, how metabolism affects CM fate transition between mononucleation and binucleation and its impact on cell cycle progression, proliferation and ability to regenerate the heart will be discussed. Full article
(This article belongs to the Special Issue Review Papers in (Re)programming Cells for Cardiac Repair)
Show Figures

Figure 1

21 pages, 1461 KiB  
Review
Direct Reprogramming of Resident Non-Myocyte Cells and Its Potential for In Vivo Cardiac Regeneration
by Sadia Perveen, Roberto Vanni, Marco Lo Iacono, Raffaella Rastaldo and Claudia Giachino
Cells 2023, 12(8), 1166; https://doi.org/10.3390/cells12081166 - 15 Apr 2023
Cited by 2 | Viewed by 2001
Abstract
Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been [...] Read more.
Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been paid to regenerative medicine to overcome this issue. Direct reprogramming is a promising therapeutic approach in regenerative cardiac medicine that has the potential to provide in situ cardiac regeneration. It consists of direct cell fate conversion of one cell type into another, avoiding transition through an intermediary pluripotent state. In injured cardiac tissue, this strategy directs transdifferentiation of resident non-myocyte cells (NMCs) into mature functional cardiac cells that help to restore the native tissue. Over the years, developments in reprogramming methods have suggested that regulation of several intrinsic factors in NMCs can help to achieve in situ direct cardiac reprogramming. Among NMCs, endogenous cardiac fibroblasts have been studied for their potential to be directly reprogrammed into both induced cardiomyocytes and induced cardiac progenitor cells, while pericytes can transdifferentiate towards endothelial cells and smooth muscle cells. This strategy has been indicated to improve heart function and reduce fibrosis after cardiac injury in preclinical models. This review summarizes the recent updates and progress in direct cardiac reprogramming of resident NMCs for in situ cardiac regeneration. Full article
(This article belongs to the Special Issue Review Papers in (Re)programming Cells for Cardiac Repair)
Show Figures

Figure 1

20 pages, 1687 KiB  
Review
Advances in Cellular Reprogramming-Based Approaches for Heart Regenerative Repair
by Xingyu He, Jialiang Liang, Christian Paul, Wei Huang, Suchandrima Dutta and Yigang Wang
Cells 2022, 11(23), 3914; https://doi.org/10.3390/cells11233914 - 3 Dec 2022
Cited by 3 | Viewed by 2568
Abstract
Continuous loss of cardiomyocytes (CMs) is one of the fundamental characteristics of many heart diseases, which eventually can lead to heart failure. Due to the limited proliferation ability of human adult CMs, treatment efficacy has been limited in terms of fully repairing damaged [...] Read more.
Continuous loss of cardiomyocytes (CMs) is one of the fundamental characteristics of many heart diseases, which eventually can lead to heart failure. Due to the limited proliferation ability of human adult CMs, treatment efficacy has been limited in terms of fully repairing damaged hearts. It has been shown that cell lineage conversion can be achieved by using cell reprogramming approaches, including human induced pluripotent stem cells (hiPSCs), providing a promising therapeutic for regenerative heart medicine. Recent studies using advanced cellular reprogramming-based techniques have also contributed some new strategies for regenerative heart repair. In this review, hiPSC-derived cell therapeutic methods are introduced, and the clinical setting challenges (maturation, engraftment, immune response, scalability, and tumorigenicity), with potential solutions, are discussed. Inspired by the iPSC reprogramming, the approaches of direct cell lineage conversion are merging, such as induced cardiomyocyte-like cells (iCMs) and induced cardiac progenitor cells (iCPCs) derived from fibroblasts, without induction of pluripotency. The studies of cellular and molecular pathways also reveal that epigenetic resetting is the essential mechanism of reprogramming and lineage conversion. Therefore, CRISPR techniques that can be repurposed for genomic or epigenetic editing become attractive approaches for cellular reprogramming. In addition, viral and non-viral delivery strategies that are utilized to achieve CM reprogramming will be introduced, and the therapeutic effects of iCMs or iCPCs on myocardial infarction will be compared. After the improvement of reprogramming efficiency by developing new techniques, reprogrammed iCPCs or iCMs will provide an alternative to hiPSC-based approaches for regenerative heart therapies, heart disease modeling, and new drug screening. Full article
(This article belongs to the Special Issue Review Papers in (Re)programming Cells for Cardiac Repair)
Show Figures

Figure 1

14 pages, 1365 KiB  
Review
Liquid–Liquid Phase Separation in Cardiovascular Diseases
by Yuanxi Mo, Yuliang Feng, Wei Huang, Ning Tan, Xinyi Li, Minwen Jie, Tong Feng, Hao Jiang and Lei Jiang
Cells 2022, 11(19), 3040; https://doi.org/10.3390/cells11193040 - 28 Sep 2022
Cited by 9 | Viewed by 3451
Abstract
Liquid–liquid phase separation (LLPS) is a biochemical process in cells that can drive proteins, RNA, and other molecules to concentrate into droplets. These droplets do not have a lipid membrane but rather exist as distinct organelles relative to the surrounding environment, and act [...] Read more.
Liquid–liquid phase separation (LLPS) is a biochemical process in cells that can drive proteins, RNA, and other molecules to concentrate into droplets. These droplets do not have a lipid membrane but rather exist as distinct organelles relative to the surrounding environment, and act as biochemical reaction chambers. In recent years, significant progress has been made in the study of LLPS, especially in the neurodegenerative disease, cancer, and virology fields, but little is known about LLPS in cardiovascular disease (CVD). In this review, we discuss the current understanding of the mechanism and biological functions of LLPS, particularly its roles in regulating CVD. Full article
(This article belongs to the Special Issue Review Papers in (Re)programming Cells for Cardiac Repair)
Show Figures

Figure 1

Back to TopTop