GET THE APP

A Review on 3D Organoid Models Over Stem Cell Based Cardiovascular Regrowth
..

Molecular and Genetic Medicine

ISSN: 1747-0862

Open Access

Mini Review - (2023) Volume 17, Issue 2

A Review on 3D Organoid Models Over Stem Cell Based Cardiovascular Regrowth

Lauren Brunelli*
*Correspondence: Lauren Brunelli, Department of Science, University of Toronto, Canada, Email:
Department of Science, University of Toronto, Canada

Received: 01-Feb-2023, Manuscript No. jmgm-23-94454; Editor assigned: 03-Feb-2023, Pre QC No. P-94454; Reviewed: 15-Feb-2023, QC No. Q-94454; Revised: 21-Feb-2023, Manuscript No. R-94454; Published: 28-Feb-2023 , DOI: 10.37421/1747-0862.2023.17.598
Citation: Brunelli, Lauren. “A Review on 3D Organoid Models Over Stem Cell Based Cardiovascular Regrowth.” J Mol Genet Med 17 (2023): 598.
Copyright: © 2023 Brunelli L. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Over the past few decades, stem cell-based cardiac regeneration has emerged as a promising therapy for patients with heart failure, a condition that affects millions of people worldwide. Stem cells have the ability to differentiate into various types of cells, including heart cells, and can potentially be used to replace damaged or dead heart tissue. One of the challenges in using stem cells for cardiac regeneration is creating a suitable environment for their growth and differentiation. Traditional two-dimensional (2D) culture methods have limitations in mimicking the complex three-dimensional (3D) environment of the heart. This is where 3D organoid models come into play. Organoids are 3D structures that can be grown from stem cells, which can self-organize and differentiate into specific cell types, mimicking the structure and function of organs. In the context of cardiac regeneration, 3D organoids can be used to model heart tissue, providing a more accurate representation of the complex 3D architecture of the heart. Recent advances in 3D organoid models for stem cell-based cardiac regeneration have shown promising results. For example, researchers have been able to create 3D heart organoids from human induced pluripotent stem cells (iPSCs), which can be used for drug screening, disease modeling, and potentially for transplantation.

Keywords

Heart organoids • Electrophysiology • Mesenchymal stem cells

Introduction

In addition, researchers have been able to create 3D organoids that mimic specific regions of the heart, such as the ventricles, and have demonstrated that these organoids can contract and pump blood, suggesting their potential for cardiac regeneration. Furthermore, advances in bioengineering techniques have enabled the creation of 3D organoids that can be vascularized, allowing for the development of blood vessels within the organoid. This is crucial for the survival and function of transplanted organoids, as they require a blood supply to survive. Overall, the use of 3D organoid models for stem cell-based cardiac regeneration holds great promise for the future of this field. While there are still challenges to overcome, such as ensuring the scalability and reproducibility of these models, the potential benefits of using 3D organoids for cardiac regeneration are significant, and further research in this area is warranted.

Despite the advances in cardiac perfection drug, clinical restatement of technologies exercising stem cells remains fugitive. Crucial questions regarding the maturity, stability, and essential benefit of iCMs implanted in vivo remain. First, the electrophysiology and generally mononucleated iCMs act a fetal or immature CM in vitro. Although, maturity of iCMs can be kindly increased with extended culture times, mechanical and electrical stimulation, and towel engineering strategies, as considerably reviewed away. Likewise, the salutary goods of direct injection of iCMs have been proven in acute ischemic injury models, though habitual studies don't show similar restorative goods. The clinical benefit of cell- grounded curatives is presently limited by the minimum retention of scattered cells in the diseased or ischemic towel. Either scattered or fitted cells don't reach the target towel and/ or die during or shortly after operation.

Literature Review

Interestingly, recent advances have been made by theco-transplantation of non-myocyte and iCMs in a rat myocardial infarction model. Co-transplantation enhanced the graft size, vascular viscosity, and development in comparison to iCMs alone, which indicates that paracrine goods between cardiac cell types may be crucial in generating estimable curatives. still, a crucial tailback that influences all implantation approaches is the allogeneic vulnerable rejection by the philanthropist. Autologous transplantation is presently expensive and time consuming. Thus, banking for mortal leukocyte antigen(HLA) matching has been established, but needs farther evaluation. An indispensable strategy may be to develop lower immunogenic PSC lines, by suppressing HLA class I and II genes while stimulating HLA-E [1-3]. The successful development of safe and effective immunocompatible strategies is essential to grease the clinical development of implantation PSC- grounded curatives. Stem cell-based cardiac regeneration is a promising field of research that aims to repair or replace damaged or diseased heart tissue using stem cells. The goal of this approach is to regenerate functional heart tissue and restore the heart's normal function.

Discussion

There are several types of stem cells that can be used for cardiac regeneration, including embryonic stem cells, induced pluripotent stem cells, and adult stem cells. Each type has its advantages and disadvantages. Embryonic stem cells are pluripotent cells that can differentiate into any cell type in the body, including heart cells. However, the use of embryonic stem cells is controversial due to ethical concerns surrounding the destruction of embryos. Induced pluripotent stem cells (iPSCs) are generated by reprogramming adult cells, such as skin cells, back to an embryonic-like state. iPSCs have the potential to differentiate into heart cells and can be generated from a patient's own cells, which reduces the risk of rejection. Adult stem cells, such as mesenchymal stem cells, are found in various tissues throughout the body, including the bone marrow, adipose tissue, and heart tissue itself.

These cells have the ability to differentiate into various cell types, including heart cells. Stem cell-based cardiac regeneration involves implanting stem cells into the damaged heart tissue, where they differentiate into heart cells and replace the damaged cells. This approach has shown promising results in preclinical studies and early-phase clinical trials [4,5]. However, there are still many challenges that need to be addressed before stem cell-based cardiac regeneration can be widely used in clinical practice. These include improving the survival and differentiation of implanted stem cells, optimizing the timing and delivery of stem cell therapy, and addressing safety concerns such as the risk of tumor formation. Presently, the sub-type-specific generation of PSC- deduced atrial and ventricle CMs is possible in 2D and 3D. Still, there are limited studies showing chamber particularity or functional crosstalk between chambers in cardiac organoids. Lately, investigators linked a robust and scalable approach for generating cardiac organoids that resembles the heart field conformation and atrioventricular specification of fetal hearts [6,7].

Conclusion

As these styles further develop, the stopgap is to potentially abstract the adult mortal heart in vitro. Moment, there are still cell types missing from cardiac organoids similar as the cardiac conduction system and vulnerable cell populations. It's known that cardiac organoids cannot show diastolic function because of the lack of the sinoatrial knot, atrioventricular knot, and Purkenje filaments, which make up the cardiac conduction system. Likewise, we also can not overlook the vulnerable cell population as signals from these cells are tightly regulated with cardiac development and response to towel damage. Therefore, beast models remain necessary in preclinical studies due to the limitation of organoids to abstract multifactorial pathologies involving the vulnerable system and multi-organ communication.

Acknowledgement

None

Conflict of Interest

Authors declare no conflict of interest.

References

  1. Poss, Kenneth D., Lindsay G. Wilson and Mark T. Keating. "Heart regeneration in zebrafish."Science298 (2002): 2188-2190.
  2. Google Scholar, Crossref, Indexed at

  3. Jopling, Chris, Eduard Sleep, Marina Raya and Mercè Martí, et al. "Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation." Nature 464 (2010): 606-609.
  4. Google Scholar, Crossref, Indexed at

  5. Archer, Caroline R., Rebecca Sargeant, Jayati Basak and James Pilling, et al. "Characterization and validation of a human 3D cardiac microtissue for the assessment of changes in cardiac pathology." Sci Rep 8 (2018): 10160.
  6. Google Scholar, Crossref, Indexed at

  7. Mills, Richard J., Benjamin L. Parker, Gregory A. Quaife-Ryan and Holly K. Voges, et al. "Drug screening in human PSC-cardiac organoids identifies pro-proliferative compounds acting via the mevalonate pathway." Cell stem cell 24 (2019): 895-907.
  8. Google Scholar, Crossref, Indexed at

  9. Forsythe, Steven D., Mahesh Devarasetty, Thomas Shupe and Colin Bishop, et al. "Environmental toxin screening using human-derived 3D bioengineered liver and cardiac organoids."Front Public Health 6 (2018): 103.
  10. Google Scholar, Crossref, Indexed at

  11. Murakami, Masahiro and Michael Simons. "Fibroblast growth factor regulation of neovascularization." Curr Opin Hematol 15 (2008): 215.
  12. Google Scholar, Crossref, Indexed at

  13. Vasquez, Carolina, Najate Benamer and Gregory E. Morley. "The cardiac fibroblast: Functional and electrophysiological considerations in healthy and diseased hearts." J Cardiovasc Pharmacol 57(2011): 380.
  14. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3919

Molecular and Genetic Medicine received 3919 citations as per Google Scholar report

Molecular and Genetic Medicine peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward