Basic Study
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Aug 26, 2022; 14(8): 633-657
Published online Aug 26, 2022. doi: 10.4252/wjsc.v14.i8.633
Combination of mesenchymal stem cells and three-dimensional collagen scaffold preserves ventricular remodeling in rat myocardial infarction model
Rida-e-Maria Qazi, Irfan Khan, Kanwal Haneef, Tuba Shakil Malick, Nadia Naeem, Waqas Ahmad, Asmat Salim, Sadia Mohsin
Rida-e-Maria Qazi, Irfan Khan, Tuba Shakil Malick, Waqas Ahmad, Asmat Salim, Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi 75270, Sindh, Pakistan
Kanwal Haneef, Dr.Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Sindh, Pakistan
Nadia Naeem, Dow Research Institute of Biotechnology and Biomedical Sciences (DRIBBS), Dow University of Health and Sciences, Ojha Campus, Karachi 74200, Sindh, Pakistan
Sadia Mohsin, Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States
Author contributions: Qazi REM performed all experiments, analyzed the data and wrote the first draft of the manuscript; Khan I co-supervised the research study, provided technical guidance for in vivo experiments; Haneef K provided technical guidance for in vitro 3D culture and wrote part of the manuscript; Malick TS performed some of the experiments; Naeem N and Ahmad W helped in echocardiographic analysis; Salim A conceived the idea, designed and supervised the research study and wrote the final manuscript; Mohsin S co-supervised the research study and provided overall technical guidance; all authors approved the final version of the manuscript.
Institutional review board statement: Human subjects have not been used in this study.
Institutional animal care and use committee statement: Ethical approval was obtained from the Institutional Animal Care and Use Committee (IACUC), Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi. Experimental procedures were carried out according to the international guidelines for care and use of laboratory animals under protocol number 2020-006.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: There is no additional data.
ARRIVE guidelines statement: The authors have read the ARRIVE guidelines, and the manuscript was prepared and revised according to the ARRIVE guidelines.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Asmat Salim, PhD, Professor, Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Main University Road, Karachi 75270, Sindh, Pakistan. asmat.salim@iccs.edu
Received: March 18, 2022
Peer-review started: March 18, 2022
First decision: April 25, 2022
Revised: May 9, 2022
Accepted: July 8, 2022
Article in press: July 8, 2022
Published online: August 26, 2022
ARTICLE HIGHLIGHTS
Research background

Cardiovascular diseases are the leading cause of death globally. Adult heart tissue possesses impaired self-renewal capability and thus shows inadequate capability of restoring its structure and function after injury. Stem cell based therapy to treat cardiac injuries has achieved moderate success due to some limitations. Cardiac tissue engineering constructs the cardiac patch or scaffold to restore cardiac function following injury. Mesenchymal stem cells (MSCs) have great potential to be used for myocardial regeneration due to their multilineage differentiation potential. Controlled fate of grafted cells can be achieved by inducing in vitro cardiac differentiation by demethylating agent such as zebularine.

Research motivation

MSCs are potential candidates for the regeneration of damaged cardiac tissue but their insufficient survival and engraftment at the injured tissue is a major hurdle. This can be overcome by pre-differentiation of MSCs using a demethylating agent and providing three-dimensional (3D) microenvironment through biological scaffold. In vivo transplantation of pre-differentiated cell seeded scaffold can provide mechanical support and enhance cell survival, engraftment and regeneration of cardiac tissue and pave the way to develop an improved cardiovascular therapeutic strategy.

Research objectives

The study was aimed to enhance the differentiation of MSCs by treating them with demethylating agent, zebularine, in a 3D microenvironment provided by collagen scaffold and subsequent enhancement of cell engraftment, survival and myocardial regeneration upon in vivo transplantation in the rat myocardial infarction (MI) model.

Research methods

MSCs were isolated from rat bone marrow and characterized on the basis of specific cell surface markers and trilineage differentiation potential. MSCs were seeded in collagen scaffold and treated with zebularine to induce cardiac differentiation. MSC-seeded scaffolds were transplanted in the rat MI model. Cardiac function assessment was done by echocardiographic analysis and ventricular regeneration by histological analysis. Neovascularization was analyzed by immunohistochemistry with α-smooth muscle actin staining. DiI labeled cell seeded scaffolds were transplanted to track the cells and their in vivo cardiac differentiation was analyzed by immunohistochemistry.

Research results

In vitro results showed significantly enhanced cardiac differentiation of MSCs after zebularine treatment in 3D culture. Transplantation of pre-differentiated MSC-seeded collagen scaffold in the rat MI model improved cardiac function more efficiently than the untreated MSC-seeded collagen scaffold group. Histological analysis also showed improvement in myocardial regeneration, ventricular wall thickness and reduction in fibrotic tissue. Immunohistochemical analysis showed significantly enhanced vasculature and in vivo cardiac differentiation of transplanted MSCs in zebularine treated MSC-seeded collagen scaffold group.

Research conclusions

Pre-differentiation of MSC-seeded collagen scaffold transplantation improves cardiac function, preserves ventricular remodeling and enhances myocardial regeneration after acute MI. This strategy provided the 3D microenvironment to the transplanted cells, enhanced their survival and engraftment at the injured tissue, as well as increased blood supply by forming a new vascular system.

Research perspectives

The combination approach using pre-differentiated MSCs and the 3D collagen scaffold can open a new insight to repair the damage caused by ischemic cardiovascular injuries.