Next Article in Journal
The Diagnostic Algorithm in Pre-Invasive Cervical Lesions
Previous Article in Journal
The Molecular Mechanisms Linking Metabolic Syndrome to Endometrial and Breast Cancers
 
 
Journal of Mind and Medical Sciences is published by MDPI from Volume 12 Issue 1 (2025). Previous articles were published by another publisher in Open Access under a CC-BY (or CC-BY-NC-ND) licence, and they are hosted by MDPI on mdpi.com as a courtesy and upon agreement with Valparaiso University (ValpoScholar).
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Efficacy of Mesenchymal Stem Cells for Cardiovascular Diseases

by
Dragana Radoje Miloradovic
1,
Dragica Radoje Pavlovic
1,
Miodrag Bozidar Stojkovic
1,2,3,4,
Sanja Bratislav Bojic
5,
Vladislav Bogdan Volarevic
6,
Marina Milosav Gazdic Jankovic
1 and
Biljana Tomislav Ljujic
1,*
1
Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Serbia
2
Spebo Medical Fertility Hospital, Leskovac, Serbia
3
Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA
4
Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
5
School of Computing, Newcastle University, Newcastle Upon Tyne, UK
6
Center For Molecular Medicine and Stem Cell Research, Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Serbia
*
Author to whom correspondence should be addressed.
J. Mind Med. Sci. 2021, 8(2), 179-190; https://doi.org/10.22543/7674.82.P179190
Submission received: 17 June 2021 / Revised: 18 June 2021 / Accepted: 10 August 2021 / Published: 10 October 2021

Abstract

:
Despite the improvements in pharmacological and surgical treatments, cardiovascular diseases (CVDs) are the number one cause of death worldwide. During the last two decades, the search for new therapies has been revolutionized with the growing knowledge of stem cell biology. Due to their huge differentiation capacity and paracrine effects, mesenchymal stem cells (MSCs) are a promising tool for the treatment of CVDs. The encouraging outcomes of preclinical studies using MSCs as a treatment for diseased myocardium have set the scene for worldwide clinical trials. In this review, we overview either complete or ongoing clinical trials using MSCs for the therapy of CVDs. In particular, we analyze the biological properties of MSCs, elucidate recent clinical findings and clinical trial phases of investigation, highlight clinical therapeutic effects of MSCs, and discuss challenges towards the clinical use of these cells in the therapy of CVDs.

Introduction

The leading cause of mortality and disability worldwide are cardiovascular diseases (CVDs), systemic circulatory diseases which arise in the field of abnormal heart and blood vessels. CVDs accounted for approximately 31% of all global deaths, causing 17.8 million deaths in 2017, mostly in low- and middle-income countries [1]. Hypertension, diabetes, hyperlipidemia, tobacco use, obesity, a sedentary lifestyle and the harmful use of alcohol are the pivotal risk factors for CVDs occurrence [2]. In spite of the notable progress in medical and surgical therapies, CVDs continue to exhibit high rates of morbidity and mortality. Drug therapy problems and cardiotoxicity are some of the biggest barriers to pharmacotherapy in the treatment of CVDs [3]. Fever, postoperative bleeding, stroke, atrial fibrillation, renal failure, respiratory distress, cardiogenic shock, and myocardial infarction type 5- represent the ordinary complications that can occur as a consequence of the surgical treatment in patients with CVDs [4,5]. Likewise, these therapeutic problems prolong hospital stays and increase the overall burden of healthcare expenditures. Therefore, a certainty has developed for therapy that it has fewer side effects, lower mortality rates, fewer complications, as well as better and secure recoveries of patients.
With the recent advances in the field of stem cell biology [6], it was suggested that stem cells might be exploited to repair damaged cardiac tissue and blood vessels. In this review, we overview either complete or ongoing clinical trials using MSCs for CVDs conducted worldwide. In particular, we analyze the biological properties of MSCs, elucidate recent clinical findings and clinical trial phases of investigation, highlight the clinical therapeutic effects of MSCs, and discuss the challenges towards the clinical use of these cells in the therapy of CVDs.

Discussions

Mesenchymal stem cells - a new therapeutic strategy for cardiovascular diseases

Mesenchymal stem cells are adult stem cells that can be isolated from nearly every tissue type in the adult or infant human body [7] such as bone marrow [8], adipose tissue [9], peripheral blood [10], synovium, and synovial fluid [11], menstrual blood and endometrium [12], skin [13], the dental pulp [14], umbilical cord blood [15], Wharton’s jelly [16], amniotic fluid and placenta [17], breast milk [18], and urine [19] (Figure 1). Usually, MSCs are plastic-adherent, fibroblast-like cells that are qualified to differentiate to osteoblasts, chondroblasts, and adipocytes in vitro [20]. MSCs are able to transdifferentiate into non-mesodermal cells such as neurons or astrocytes, hepatocytes, and pancreatic cells under specified culture conditions [21]. Due to the absence of an MSC-specific marker, the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy proposed a minimum number of criteria for the phenotypic characterization of human MSCs [22]. MSCs are positive for CD105, CD73, and CD90 and characterized by the lack of expression CD45, CD34, CD14, CD11b, CD79 alpha or CD19, and HLA-DR surface molecules [22].
A tremendous number of animal models that resemble the pathology of CVDs in humans have demonstrated not only the therapeutic efficacy of culture-expanded MSCs, but also their mechanism of action that may be of relevance to the future widespread clinical use of MSCs [23,24,25,26,27,28,29,30]. MSCs represent a cellular source for cardiovascular repair since they are able to differentiate into cardiomyocytes (CMs) [31,32,33], endothelial cells (ECs) [34,35,36], and smooth muscle cells (SMCs) [36,37,38] (Figure 1). However, very few MSCs are found engrafted at the site of injury linked to the degree of functional recovery [7,21]. Thus, the vast majority of recently published studies revealed that the beneficial effects and utilization of MSCs in CVDs will be also based on their ability to produce a wide range of cytokines, chemokines, and growth factors [39] that resolve inflammation [24], inhibit apoptosis [26,27] and fibrosis [26,40], promote angiogenesis [25,29,40,41,42,43,44,45,46,47,48,49,50], and induce endogenous cardiomyocyte regeneration [43,44,45] (Figure 1).
MSCs attenuate cardiac injury through the production of several pro-angiogenic factors such as the placental growth factor (PGF), vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF-1), and basic fibroblast growth factor (bFGF), which facilitate tissue regeneration by inducing the proliferation of ECs and by promoting neovascularization [25,26,29,40,41,42,46]. In cell-to-cell contact and through the production of soluble mediators, MSCs can alter the function of all immune cells that have a key role in the pathogenesis of CVDs [47,48]. MSCs regulate the proliferation, activation, and effector function of T lymphocytes, professional antigen-presenting cells (dendritic cells, macrophages, B lymphocytes), NK cells, and NKT cells [48,49]. Thus, the improvement of a wide range of cardiac functions (increased EF, decreased in scar tissue, reversed remodeling, ameliorated contractility, augmented heart perfusion, and improved blood vessel density) [30,41,50,51,52], indicates the possible use of MSCs in the therapy of CVDs. Over the past few years, MSCs were used in a broad range of clinical trials exploring the therapeutic effects of MSCs in the treatment of coronary artery disease, heart failure, and cardiomyopathy (Table 1), as well as peripheral artery disease and stroke (Table 2) and their findings, were analyzed in this review.

MSC-based therapy of coronary artery disease

One study showed that one month after ST-elevation MI (STEMI), the intracoronary infusion of autologous bone marrow-derived MSCs (BM-MSCs) provided a modest improvement in the left ventricular ejection fraction (LVEF) at six-months follow-up by single-photon emission computed tomography [51]. It is also important to notice that there was no treatment-related toxicity during the administration of MSCs, and no serious cardiovascular complications throughout the follow-up [51]. For the reason that cell therapy is not possible at an early phase of remodeling, growing evidence recommends that the prevention of LV remodeling by MSCs transplantation throughout the late dilation phase is practical [53]. As a proper technique for supplementing primary percutaneous coronary intervention (PCI), several research groups investigated intracoronary transplantation of autologous BM-MSCs [53,54].
In patients with acute anterior wall MI, they found that the administration of autologous BM-MSC at one month after PCI is tolerable and safe with a significant improvement in LVEF one month and four months after the stem cell treatment [54]. Serial cardiac echocardiographic monitoring displayed that the improvement of the cardiac function was kept nearly six months after the procedure [53]. At the 16–36- month follow-up, the implanted cardioverter defibrillators failed to discover sustained or non-sustained ventricular arrhythmia in any patient with previous anteroseptal MI who underwent intracoronary transplantation of autologous BM-MSCs [55]. MSCs may be used as an allogeneic graft for the treatment of CVDs, due to the lack of various major histocompatibility complex and costimulatory cell-surface antigens [56,57].
In a randomized, double-blind, placebo-controlled study, Hare and co. showed that the intravenous administration of allogeneic BM-MSCs (0.5, 1.6, and 5.0×106 MSCs/kg body weight) is well-tolerated, safe, and effective in patients after acute anterior MI [58]. In the same study, they showed that LVEF, forcible expiratory volume and global symptom score were significantly improved, while the incidence of arrhythmia was reduced in MSC-treated compared to placebo-treated post-infarction subjects [58]. In a double-blind, randomized controlled trial of Gao and co., it was elucidated that the intracoronary application of WJMSCs in patients with acute STEMI significantly increased myocardial viability, perfusion within the infarcted territory, as well as the global LVEF [52]. WJMSC infusion averted post-infarct LV adverse remodeling, as evidenced by the modifications in the LV end-systolic volumes (LVESV) and end-diastolic volumes (LVEDV) at 18 months [52] (Table 1).

MSCs as novel therapeutic agents in the treatment of heart failure

It was demonstrated that intra-myocardial injections of autologous BM-MSCs were safe and improved the myocardial function in patients with ischemic HF, compared to placebo in terms of LVEF, stroke volume, myocardial mass, and cardiac output [59,60]. The administration of BM-MSCs, as evidenced by progressive decreases in both LVEDV and LVESV, led to reverse LV remodeling, indicating an improved outcome in patients with HF [59,60].
In patients with ischemic or non-ischemic HF, Perin and co. evaluated the feasibility and safety of different doses (25, 75, or 150 million cells) of immune-selected allogeneic mesenchymal precursors cells (MPCs [61]. They found that the transendocardial route of delivery for MPCs therapy was safe and well-tolerated without clinically symptomatic immune responses [61]. The highest MPCs dose diminished the adverse cardiac events and reversed certain parameters of cardiac remodeling in HF patients [61]. Nevertheless, further studies are needed to explore the relationship between the number of intramyocardial injections, the number of stem cells and the volume administered, and their impact on the observed reverse remodeling outcome. Kastrup and co. formed a centralized production of an allogeneic Cardiology Stem Cell Centre adipose-derived stromal cell (CSCC_ASC) product from healthy donors that delivered directly to the myocardium into the border area of infarcted tissue and showed safety, feasibility, and a tendency toward clinical efficacy in ten patients with ischemic heart disease and ischemic HF [62]. It is important to notice that none of the patients had any clinical symptoms or changes in biochemical parameters or inflammatory signs implying immunization. With the aim of assessing changes in parameters such as LVEDV, LVEF, stroke volume, myocardial mass, cardiac output, and change in clinical symptoms in ischemic HF patients treated with CSCC_ASC, a double-blind, placebo-controlled, multicenter study is currently in phase II [63]. The umbilical cord-derived MSCs (UC-MSCs) are easily attainable stem cells and can differentiate into cardiomyocyte-like and ECs in vitro, and also exert paracrine effects that enhance vascular regeneration and cardiomyocyte protection. Due to concerns about the safety of donor cell entrapment in the pulmonary circulation and whether they will achieve their therapeutic effect in a context of low cardiac engraftment, there is limited experience on intravenous administration of MSCs in patients with CVDs [64]. The intravenous delivery of UC-MSCs seems safe and efficient in patients with chronic stable HF [64]. UC-MSC–treated group exhibited significant improvements in the ventricular systolic function, New York Heart Association functional classification (NYHA Classification), and quality of life indices [64] (Table 1).

MSCs as new agents in cell-based therapy of cardiomyopathy

Progressive disorders most often end with heart transplants and they are known as ischemic cardiomyopathy (ICM) and dilated cardiomyopathy (DCM). Given the fact that MSCs secrete numerous paracrine factors that attenuate these mechanisms and possess immunomodulatory properties, they are a promising therapy for both forms of cardiomyopathy [65].
For patients with chronic ICM and LV dysfunction, it was shown that transendocardial and intramyocardial injections of autologous MSCs were safe [66,67]. MSCs improved the cardiac structure and function through several mechanisms such as reducing fibrosis and promoting neoangiogenesis and neomyogenesis [67]. Increased viable myocardial mass and decreased scar size suggested a true MSC-mediated myocardial regeneration [66,67]. Moreover, an improvement in clinical outcomes such as a 6-minute walk test and Minnesota Living with Heart Failure Questionnaire in cardiac patients receiving MSCs therapy was associated with increased LVEF [66,67].
Hare and co. conducted a randomized comparison on the safety and efficacy of allogeneic versus autologous BM-MSCs in patients with cardiomyopathy [57,68]. In both sorts of cardiomyopathies- ICM and DCM, transendocardial injections of allogeneic and autologous MSCs were equally safe without serious adverse events or immunological reactions, additionally propounding that allogeneic MSCs are characteristically immunomodulatory [57,68]. MSCs therapy improved the functional capacity and the quality of life in both clinical entities [57,68]. Several lines of evidence suggested clinically meaningful efficacy and immunosuppressive capacity of allogeneic MSCs compared to autologous MSCs in DCM patients [68]. Through endocrine or paracrine effects, which might promote neovascularization, inhibit cardiomyocyte apoptosis and enhance cardiac repair it is possible to explain the sustained improvement in LVEF, NYHA class, and myocardial perfusion [69]. Interestingly, ventricular remodeling was reversed by both of MSCs- autologous and allogeneic with inverse dose-response [57]. Specifically, in a group of patients with ICM who received lower-doses of MSCs (20 million cells) compared to the group that received 200 million cells, it was found that the first group of patients had significantly greater LVEF, improvement in LVESV, and a reduction in scar size [57]. Due to discrepant and conflicting results, Florea et al. evaluated the relationship between MSCs dose and clinical benefit in patients with ICM [70]. Although both cell doses (20 or 100 million allogeneic BM-MSCs) reduced the scar size, merely the higher dose of MSCs increased EF [70].

MSC-mediated modulation of peripheral artery disease

Peripheral arterial disease (PAD) is a common complication of atherosclerosis. One of the most severe forms of PAD is critical limb ischemia (CLI) which is characterized by rest pain, ischemic ulceration, or gangrene with or without tissue damage [71]. MSCs may improve the consequences of CLI due to their combining potential for inducing angiogenesis and the immunomodulatory environment in situ (Table 2).
During the 24-week follow-up period, the autologous transplantation of BM-MSCs in diabetic patients with CLI and foot ulcers was well tolerated and effective with respect to a few hemodynamic and clinical parameters [72]. In the same study, it was displayed that magnetic resonance angiography scores, Ankle-Brachial Pressure Index (ABPI), and transcutaneous oxygen pressure before and after the infusion of the cells revealed the increased blood flow, presumable interceded by collateral vessel development [72]. The clinical improvement was verified by ulcer healing rate and painless walking [72]. Posterior to intramuscular administration of allogeneic BM-MSCs (2 million cells/kg) in patients with CLI, significantly improved ankle pressure and ABPI were noticed [73]. There was no utility in the treatment with BM-MSCs for patients with impending amputation [73]. Due to the higher frequency of obtaining the unit of tissue per volume, and larger angiogenic potential compared to BM-MSCs, adipose tissue-derived MSCs (AT-MSCs) have been proposed as feasible approaches for the treatment of disorders related to limb ischemia [74]. A CellDREAM trial displayed that the intramuscular injection of AT-MSCs into the ischemic leg of patients with CLI improved ulcer evolution and wound healing and increased transcutaneous oxygen pressure without any further complications [74]. Besides, a meta-analysis on the usage of stem cell therapy in CLI certified the benefits observed in single studies [75].

Mesenchymal stem cells therapy in stroke

By using the conventional definition of stroke, it can be understood as the abrupt beginning of the loss of focal neurological function due to infarction or hemorrhage in the critical segment of the brain, retina, or spinal cord [76]. It is also important to note that stroke is the leading cause of major disabilities and the second leading cause of death worldwide [77]. Many studies revealed that the application of MSCs in ischemic stroke is secure [78,79,80,81,82], and effective throughout long-term tracking [78,79,80,82] (Table 2). In the first intracerebral stem cell transplantation study, 18 patients with chronic stroke received a single dose of modified BM-MSCs (SB623) (2.5×106, 5.0×106, or 10×106) and demonstrated a significant improvement in the neurological function measured by means of the European Stroke Scale (ESS), the National Institutes of Health Stroke Scale (NIHSS), Fugl-Meyer (F-M) total score, and Fugl-Meyer (F-M) motor function total score at 12 months after the treatment [78].
Lee and co. elucidated that the intravenous application of 5×107 autologous MSCs to patients with ischemic stroke improved the clinical parameters that were correlated with the plasma stromal cell-derived factor-1 alpha level at the time of MSC treatment as well as significantly reduced the mortality rate compared to a control group [82]. Levy and co. showed that the intravenous injection of allogeneic MSCs (the same as the autologous transplantation of MSCs) in patients with chronic stroke did not cause any adverse events [80]. MSCs in each dose (0.5, 1.0, and 1.5 million cells/kg body weight) were safe and related to the functional recovery [80].

Conclusions

Mesenchymal stem cells are considered the new therapeutic agents in the treatment of CVDs, on the grounds of their differentiation abilities and paracrine effects. The results obtained in a large number of clinical trials suggest that the local as well as the systemic application of autologous or allogeneic MSCs is a beneficial therapeutic approach for CVDs. Although these studies are paving the way for the development of MSC-based regenerative therapies for structural and functional disorders of the myocardium and blood vessels, the optimal origin and the number of transplanted MSCs as well as the route of their application should be clearly defined and uniformed in such a way so as to increase the reproducibility and the consistency of the data. Moreover, safety issues regarding MSCs-based therapy are still a matter of debate. The primary concern is the unwanted differentiation of the transplanted MSCs and their potential to suppress the anti-tumor immune response and generate new blood vessels that may promote tumor growth and metastasis [83-86], although some studies displayed opposite results [86]. Therefore, new clinical studies should be focused on the continuous monitoring and the long-term follow-up of MSC-treated patients in order to determine the possible side effects of MSC-based therapy of CVDs.

Funding

This study was supported by the Serbian Ministry of Sciences (project number ON 175103), and by Faculty of Medical Sciences, The University of Kragujevac, Serbia (JP 25/19).

Conflict of interest disclosure

There are no known conflicts of interest in the publication of this article. The manuscript was read and approved by all authors.

Acknowledgments

We would like to thank Andjelka Armus for her help in the preparation and illustration of the figures.

References

  1. GBD 2017 Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018, 392, 1736–1788. [Google Scholar] [CrossRef] [PubMed]
  2. Flora, G.D.; Nayak, M.K. A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes. Curr Pharm Des. 2019, 25, 4063–4084. [Google Scholar] [CrossRef] [PubMed]
  3. Mladěnka, P.; Applová, L.; Patočka, J.; Costa, V.M.; Remiao, F.; Pourová, J.; Mladěnka, A.; Karlíčková, J.; Jahodář, L.; Vopršalová, M.; Varner, K.J.; Štěrba, M.; TOX-OER and CARDIOTOX Hradec Králové Researchers and Collaborators. Comprehensive review of cardiovascular toxicity of drugs and related agents. Med Res Rev. 2018, 38, 1332–1403. [Google Scholar] [CrossRef]
  4. Senst, B.; Kumar, A.; Diaz, R.R. Cardiac Surgery. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2021- forthcoming. https://www.ncbi.nlm.nih. Available online: https://www.ncbi.nlm.nih.gov/books/N BK532935/.
  5. Howitt, S.H.; Herring, M.; Malagon, I.; McCollum, C.N.; Grant, S.W. Incidence and outcomes of sepsis after cardiac surgery as defined by the Sepsis-3 guidelines. Br J Anaesth. 2018, 120, 509–516. [Google Scholar] [CrossRef]
  6. Volarevic, V.; Bojic, S.; Nurkovic, J.; Volarevic, A.; Ljujic, B.; Arsenijevic, N.; Lako, M.; Stojkovic, M. Stem cells as new agents for the treatment of infertility: current and future perspectives and challenges. Biomed Res Int. 2014, 2014, 507234. [Google Scholar] [CrossRef] [PubMed]
  7. Harrell, C.R.; Sadikot, R.; Pascual, J.; Fellabaum, C.; Jankovic, M.G.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Mesenchymal Stem Cell-Based Therapy of Inflammatory Lung Diseases: Current Understanding and Future Perspectives. Stem Cells Int. 2019, 2019, 4236973. [Google Scholar] [CrossRef]
  8. Otsuru, S.; Hofmann, T.J.; Olson, T.S.; Dominici, M.; Horwitz, E.M. Improved isolation and expansion of bone marrow mesenchymal stromal cells using a novel marrow filter device. Cytotherapy. 2013, 15, 146–153. [Google Scholar] [CrossRef]
  9. Guasti, L.; New, S.E.; Hadjidemetriou, I.; Palmiero, M.; Ferretti, P. Plasticity of human adipose-derived stem cells - relevance to tissue repair. Int J Dev Biol. 2018, 62, 431–439. [Google Scholar] [CrossRef]
  10. Tondreau, T.; Meuleman, N.; Delforge, A.; Dejeneffe, M.; Leroy, R.; Massy, M.; Mortier, C.; Bron, D.; Lagneaux, L. Mesenchymal stem cells derived from CD133-positive cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and plasticity. Stem Cells. 2005, 23, 1105–1112. [Google Scholar] [CrossRef]
  11. Tatullo, M.; Codispoti, B.; Pacifici, A.; Palmieri, F.; Marrelli, M.; Pacifici, L.; Paduano, F. Potential Use of Human Periapical Cyst-Mesenchymal Stem Cells (hPCy- MSCs) as a Novel Stem Cell Source for Regenerative Medicine Applications. Front Cell Dev Biol. 2017, 5, 103. [Google Scholar] [CrossRef]
  12. Cagliani, J.; Grande, D.; Molmenti, E.P.; Miller, E.J.; Rilo, H.L.R. Immunomodulation by Mesenchymal Stromal Cells and Their Clinical Applications. J Stem Cell Regen Biol. 2017, 3, 10.15436/2471-0598.17.022. [Google Scholar] [CrossRef]
  13. Niezgoda, A.; Niezgoda, P.; Nowowiejska, L.; Białecka, A.; Męcińska-Jundziłł, K.; Adamska, U.; Czajkowski, R. Properties of skin stem cells and their potential clinical applications in modern dermatology. Eur J Dermatol. 2017, 27, 227–236. [Google Scholar] [CrossRef]
  14. Bojic, S.; Volarevic, V.; Ljujic, B.; Stojkovic, M. Dental stem cells--characteristics and potential. Histol Histopathol. 2014, 29, 699–706. [Google Scholar] [CrossRef]
  15. Majka, M.; Sułkowski, M.; Badyra, B.; Musiałek, P. Concise Review: Mesenchymal Stem Cells in Cardiovascular Regeneration: Emerging Research Directions and Clinical Applications. Stem Cells Transl Med. 2017, 6, 1859–1867. [Google Scholar] [CrossRef]
  16. Alves da Silva, M.L.; Costa-Pinto, A.R.; Martins, A.; Correlo, V.M.; Sol, P.; Bhattacharya, M.; Faria, S.; Reis, R.L.; Neves, N.M. Conditioned medium as a strategy for human stem cells chondrogenic differentiation. J Tissue Eng Regen Med. 2015, 9, 714–723. [Google Scholar] [CrossRef] [PubMed]
  17. Koike, C.; Zhou, K.; Takeda, Y.; Fathy, M.; Okabe, M.; Yoshida, T.; Nakamura, Y.; Kato, Y.; Nikaido, T. Characterization of amniotic stem cells. Cell Reprogram. 2014, 16, 298–305. [Google Scholar] [CrossRef] [PubMed]
  18. Goudarzi, N.; Shabani, R.; Ebrahimi, M.; Baghestani, A.; Dehdashtian, E.; Vahabzadeh, G.; Soleimani, M.; Moradi, F.; Katebi, M. Comparative phenotypic characterization of human colostrum and breast milk-derived stem cells. Hum Cell. 2020, 33, 308–317. [Google Scholar] [CrossRef] [PubMed]
  19. He, W.; Zhu, W.; Cao, Q.; Shen, Y.; Zhou, Q.; Yu, P.; Liu, X.; Ma, J.; Li, Y.; Hong, K. Generation of Mesenchymal-Like Stem Cells From Urine in Pediatric Patients. Transplant Proc. 2016, 48, 2181–2185. [Google Scholar] [CrossRef]
  20. Harrell, C.R.; Gazdic, M.; Fellabaum, C.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Therapeutic Potential of Amniotic Fluid Derived Mesenchymal Stem Cells Based on their Differentiation Capacity and Immunomodulatory Properties. Curr Stem Cell Res Ther. 2019, 14, 327–336. [Google Scholar] [CrossRef]
  21. Gazdic, M.; Arsenijevic, A.; Markovic, B.S.; Volarevic, A.; Dimova, I.; Djonov, V.; Arsenijevic, N.; Stojkovic, M.; Volarevic, V. Mesenchymal Stem Cell-Dependent Modulation of Liver Diseases. Int J Biol Sci. 2017, 13, 1109–1117. [Google Scholar] [CrossRef] [PubMed]
  22. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.j.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006, 8, 315–317. [Google Scholar] [CrossRef]
  23. Toma, C.; Pittenger, M.F.; Cahill, K.S.; Byrne, B.J.; Kessler, P.D. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002, 105, 93–98. [Google Scholar] [CrossRef] [PubMed]
  24. Ohnishi, S.; Yanagawa, B.; Tanaka, K.; Miyahara, Y.; Obata, H.; Kataoka, M.; Kodama, M.; Ishibashi-Ueda, H.; Kangawa, K.; Kitamura, S.; Nagaya, N. Transplantation of mesenchymal stem cells attenuates myocardial injury and dysfunction in a rat model of acute myocarditis. J Mol Cell Cardiol. 2007, 42, 88–97. [Google Scholar] [CrossRef] [PubMed]
  25. Musiał-Wysocka, A.; Kot, M.; Sułkowski, M.; Majka, M. Regenerative Potential of the Product "CardioCell" Derived from the Wharton's Jelly Mesenchymal Stem Cells for Treating Hindlimb Ischemia. Int J Mol Sci. 2019, 20, 4632. [Google Scholar] [CrossRef] [PubMed]
  26. Gong, X.; Wang, P.; Wu, Q.; Wang, S.; Yu, L.; Wang, G. Human umbilical cord blood derived mesenchymal stem cells improve cardiac function in cTnT(R141W) transgenic mouse of dilated cardiomyopathy. Eur J Cell Biol. 2016, 95, 57–67. [Google Scholar] [CrossRef]
  27. Zhang, B.; Zhang, J.; Zhu, D.; Kong, Y. Mesenchymal stem cells rejuvenate cardiac muscle after ischemic injury. Aging (Albany NY). 2019, 11, 63–72. [Google Scholar] [CrossRef]
  28. Cheng, Z.; Wang, L.; Qu, M.; Liang, H.; Li, W.; Li, Y.; Deng, L.; Zhang, Z.; Yang, G.Y. Mesenchymal stem cells attenuate blood-brain barrier leakage after cerebral ischemia in mice. J Neuroinflammation. 2018, 15, 135. [Google Scholar] [CrossRef]
  29. Yao, Y.; Huang, J.; Geng, Y.; Qian, H.; Wang, F.; Liu, X.; Shang, M.; Nie, S.; Liu, N.; Du, X.; Dong, J.; Ma, C. Paracrine action of mesenchymal stem cells revealed by single cell gene profiling in infarcted murine hearts. PLoS One. 2015, 10, e0129164. [Google Scholar] [CrossRef] [PubMed]
  30. Luger, D.; Lipinski, M.J.; Westman, P.C.; Glover, D.K.; Dimastromatteo, J.; Frias, J.C.; Albelda, M.T.; Sikora, S.; Kharazi, A.; Vertelov, G.; Waksman, R.; Epstein, S.E. Intravenously Delivered Mesenchymal Stem Cells: Systemic Anti-Inflammatory Effects Improve Left Ventricular Dysfunction in Acute Myocardial Infarction and Ischemic Cardiomyopathy. Circ Res. 2017, 120, 1598–1613. [Google Scholar] [CrossRef]
  31. Huang, Y.S.; Li, I.H.; Chueh, S.H.; Hueng, D.Y.; Tai, M.C.; Liang, C.M.; Lien, S.B.; Sytwu, H.K.; Ma, K.H. Mesenchymal stem cells from rat olfactory bulbs can differentiate into cells with cardiomyocyte characteristics. J Tissue Eng Regen Med. 2015, 9, E191–E201. [Google Scholar] [CrossRef]
  32. Soltani, L.; Rahmani, H.R.; Daliri Joupari, M.; Ghaneialvar, H.; Mahdavi, A.H.; Shamsara, M. Ovine fetal mesenchymal stem cell differentiation to cardiomyocytes, effects of co-culture, role of small molecules; reversine and 5-azacytidine. Cell Biochem Funct. 2016, 34, 250–261. [Google Scholar] [CrossRef]
  33. Hafez, P.; Jose, S.; Chowdhury, S.R.; Ng, M.H.; Ruszymah, B.H.; Abdul Rahman Mohd, R. Cardiomyogenic differentiation of human sternal bone marrow mesenchymal stem cells using a combination of basic fibroblast growth factor and hydrocortisone. Cell Biol Int. 2016, 40, 55–64. [Google Scholar] [CrossRef]
  34. Joddar, B.; Kumar, S.A.; Kumar, A. A Contact-Based Method for Differentiation of Human Mesenchymal Stem Cells into an Endothelial Cell-Phenotype. Cell Biochem Biophys. 2018, 76, 187–195. [Google Scholar] [CrossRef] [PubMed]
  35. Gu, W.; Hong, X.; Le Bras, A.; Nowak, W.N.; Issa Bhaloo, S.; Deng, J.; Xie, Y.; Hu, Y.; Ruan, X.Z.; Xu, Q. Smooth muscle cells differentiated from mesenchymal stem cells are regulated by microRNAs and suitable for vascular tissue grafts. J Biol Chem. 2018, 293, 8089–8102. [Google Scholar] [CrossRef]
  36. Zhang, X.; Bendeck, M.P.; Simmons, C.A.; Santerre, J.P. Deriving vascular smooth muscle cells from mesenchymal stromal cells: Evolving differentiation strategies and current understanding of their mechanisms. Biomaterials. 2017, 145, 9–22. [Google Scholar] [CrossRef]
  37. Gu, W.; Hong, X.; Le Bras, A.; Nowak, W.N.; Issa Bhaloo, S.; Deng, J.; Xie, Y.; Hu, Y.; Ruan, X.Z.; Xu, Q. Smooth muscle cells differentiated from mesenchymal stem cells are regulated by microRNAs and suitable for vascular tissue grafts. J Biol Chem. 2018, 293, 8089–8102. [Google Scholar] [CrossRef]
  38. White, I.A.; Sanina, C.; Balkan, W.; Hare, J.M. Mesenchymal Stem Cells in Cardiology. Methods Mol Biol. 2016, 1416, 55–87. [Google Scholar] [CrossRef] [PubMed]
  39. Volarevic, V.; Gazdic, M.; Simovic Markovic, B.; Jovicic, N.; Djonov, V.; Arsenijevic, N. Mesenchymal stem cell- derived factors: Immuno-modulatory effects and therapeutic potential. Biofactors. 2017, 43, 633–644. [Google Scholar] [CrossRef]
  40. Zhang, J.; Wu, Y.; Chen, A.; Zhao, Q. Mesenchymal stem cells promote cardiac muscle repair via enhanced neovascularization. Cell Physiol Biochem. 2015, 35, 1219–1229. [Google Scholar] [CrossRef]
  41. Ohnishi, S.; Yanagawa, B.; Tanaka, K.; Miyahara, Y.; Obata, H.; Kataoka, M.; Kodama, M.; Ishibashi-Ueda, H.; Kangawa, K.; Kitamura, S.; Nagaya, N. Transplantation of mesenchymal stem cells attenuates myocardial injury and dysfunction in a rat model of acute myocarditis. J Mol Cell Cardiol. 2007, 42, 88–97. [Google Scholar] [CrossRef]
  42. Markel, T.A.; Wang, Y.; Herrmann, J.L.; Crisostomo, P.R.; Wang, M.; Novotny, N.M.; Herring, C.M.; Tan, J.; Lahm, T.; Meldrum, D.R. VEGF is critical for stem cell-mediated cardioprotection and a crucial paracrine factor for defining the age threshold in adult and neonatal stem cell function. Am J Physiol Heart Circ Physiol. 2008, 295, H2308–H2314. [Google Scholar] [CrossRef] [PubMed]
  43. Hatzistergos, K.E.; Quevedo, H.; Oskouei, B.N.; Hu, Q.; Feigenbaum, G.S.; Margitich, I.S.; Mazhari, R.; Boyle, A.J.; Zambrano, J.P.; Rodriguez, J.E.; Dulce, R.; Pattany, P.M.; Valdes, D.; Revilla, C.; Heldman, A.W.; McNiece, I.; Hare, J.M. Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circ Res. 2010, 107, 913–922. [Google Scholar] [CrossRef] [PubMed]
  44. Hatzistergos, K.E.; Saur, D.; Seidler, B.; Balkan, W.; Breton, M.; Valasaki, K.; Takeuchi, L.M.; Landin, A.M.; Khan, A.; Hare, J.M. Stimulatory Effects of Mesenchymal Stem Cells on cKit+ Cardiac Stem Cells Are Mediated by SDF1/CXCR4 and SCF/cKit Signaling Pathways. Circ Res. 2016, 119, 921–930. [Google Scholar] [CrossRef]
  45. Beigi, F.; Schmeckpeper, J.; Pow-Anpongkul, P.; Payne, J.A.; Zhang, L.; Zhang, Z.; Huang, J.; Mirotsou, M.; Dzau, V.J. C3orf58, a novel paracrine protein, stimulates cardiomyocyte cell-cycle progression through the PI3K-AKT-CDK7 pathway. Circ Res. 2013, 113, 372–380. [Google Scholar] [CrossRef]
  46. Liu, X.; Chen, H.; Zhu, W.; Chen, H.; Hu, X.; Jiang, Z.; Xu, Y.; Zhou, Y.; Wang, K.; Wang, L.; Chen, P.; Hu, H.; Wang, C.; Zhang, N.; Ma, Q.; Huang, M.; Hu, D.; Zhang, L.; Wu, R.; Wang, Y.; Xu, Q.; Yu, H.; Wang, J. Transplantation of SIRT1-engineered aged mesenchymal stem cells improves cardiac function in a rat myocardial infarction model. J Heart Lung Transplant. 2014, 33, 1083–1092. [Google Scholar] [CrossRef]
  47. Harrell, C.R.; Jankovic, M.G.; Fellabaum, C.; Volarevic, A.; Djonov, V.; Arsenijevic, A.; Volarevic, V. Molecular Mechanisms Responsible for Anti-inflammatory and Immunosuppressive Effects of Mesenchymal Stem Cell-Derived Factors. Adv Exp Med Biol. 2019, 1084, 187–206. [Google Scholar] [CrossRef]
  48. Gazdic, M.; Volarevic, V.; Arsenijevic, N.; Stojkovic, M. Mesenchymal stem cells: a friend or foe in immune- mediated diseases. Stem Cell Rev Rep. 2015, 11, 280–287. [Google Scholar] [CrossRef]
  49. Markovic, B.S.; Kanjevac, T.; Harrell, C.R.; Gazdic, M.; Fellabaum, C.; Arsenijevic, N.; Volarevic, V. Molecular and Cellular Mechanisms Involved in Mesenchymal Stem Cell-Based Therapy of Inflammatory Bowel Diseases. Stem Cell Rev Rep. 2018, 14, 153–165. [Google Scholar] [CrossRef]
  50. Molina, E.J.; Palma, J.; Gupta, D.; Torres, D.; Gaughan, J.P.; Houser, S.; Macha, M. Reverse remodeling is associated with changes in extracellular matrix proteases and tissue inhibitors after mesenchymal stem cell (MSC) treatment of pressure overload hypertrophy. J Tissue Eng Regen Med. 2009, 3, 85–91. [Google Scholar] [CrossRef]
  51. Lee, J.W.; Lee, S.H.; Youn, Y.J.; Ahn, M.S.; Kim, J.Y.; Yoo, B.S.; Yoon, J.; Kwon, W.; Hong, I.S.; Lee, K.; Kwan, J.; Park, K.S.; Choi, D.; Jang, Y.S.; Hong, M.K. A randomized, open- label, multicenter trial for the safety and efficacy of adult mesenchymal stem cells after acute myocardial infarction. J Korean Med Sci. 2014, 29, 23–31. [Google Scholar] [CrossRef]
  52. Gao, L.R.; Chen, Y.; Zhang, N.K.; Yang, X.L.; Liu, H.L.; Wang, Z.G.; Yan, X.Y.; Wang, Y.; Zhu, Z.M.; Li, T.C.; Wang, L.H.; Chen, H.Y.; Chen, Y.D.; Huang, C.L.; Qu, P.; Yao, C.; Wang, B.; Chen, G.H.; Wang, Z.M.; Xu, Z.Y.; Bai, J.; Lu, D.; Shen, Y.H.; Guo, F.; Liu, M.Y.; Yang, Y.; Ding, Y.C.; Yang, Y.; Tian, H.T.; Ding, Q.A.; Li, L.N.; Yang, X.C.; Hu, X. Intracoronary infusion of Wharton's jelly-derived mesenchymal stem cells in acute myocardial infarction: double-blind, randomized controlled trial. BMC Med. 2015, 13, 162. [Google Scholar] [CrossRef] [PubMed]
  53. Chen, S.L.; Fang, W.W.; Ye, F.; Liu, Y.H.; Qian, J.; Shan, S.J.; Zhang, J.J.; Chunhua, R.Z.; Liao, L.M.; Lin, S.; Sun, J.P. Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. Am J Cardiol. 2004, 94, 92–95. [Google Scholar] [CrossRef]
  54. Kim, S.H.; Cho, J.H.; Lee, Y.H.; Lee, J.H.; Kim, S.S.; Kim, M.Y.; Lee, M.G.; Kang, W.Y.; Lee, K.S.; Ahn, Y.K.; Jeong, M.H.; Kim, H.S. Improvement in Left Ventricular Function with Intracoronary Mesenchymal Stem Cell Therapy in a Patient with Anterior Wall ST-Segment Elevation Myocardial Infarction. Cardiovasc Drugs Ther. 2018, 32, 329–338. [Google Scholar] [CrossRef]
  55. Katritsis, D.G.; Sotiropoulou, P.; Giazitzoglou, E.; Karvouni, E.; Papamichail, M. Electrophysiological effects of intracoronary transplantation of autologous mesenchymal and endothelial progenitor cells. Europace. 2007, 9, 167–171. [Google Scholar] [CrossRef] [PubMed]
  56. Golpanian, S.; Wolf, A.; Hatzistergos, K.E.; Hare, J.M. Rebuilding the Damaged Heart: Mesenchymal Stem Cells, Cell-Based Therapy, and Engineered Heart Tissue. Physiol Rev. 2016, 96, 1127–1168. [Google Scholar] [CrossRef]
  57. Hare, J.M.; DiFede, D.L.; Rieger, A.C.; Florea, V.; Landin, A.M.; El-Khorazaty, J.; Khan, A.; Mushtaq, M.; Lowery, M.H.; Byrnes, J.J.; Hendel, R.C.; Cohen, M.G.; Alfonso, C.E.; Valasaki, K.; Pujol, M.V.; Golpanian, S.; Ghersin, E.; Fishman, J.E.; Pattany, P.; Gomes, S.A.; Delgado, C.; Miki, R.; Abuzeid, F.; Vidro-Casiano, M.; Premer, C.; Medina, A.; Porras, V.; Hatzistergos, K.E.; Anderson, E.; Mendizabal, A.; Mitrani, R.; Heldman, A.W. Randomized Comparison of Allogeneic Versus Autologous Mesenchymal Stem Cells for Nonischemic Dilated Cardiomyopathy: POSEIDON-DCM Trial. J Am Coll Cardiol. 2017, 69, 526–537. [Google Scholar] [CrossRef]
  58. Hare, J.M.; Traverse, J.H.; Henry, T.D.; Dib, N.; Strumpf, R.K.; Schulman, S.P.; Gerstenblith, G.; DeMaria, A.N.; Denktas, A.E.; Gammon, R.S.; Hermiller, J.B., Jr.; Reisman, M.A.; Schaer, G.L.; Sherman, W. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009, 54, 2277–2286. [Google Scholar] [CrossRef] [PubMed]
  59. Mathiasen, A.B.; Qayyum, A.A.; Jørgensen, E.; Helqvist, S.; Fischer-Nielsen, A.; Kofoed, K.F.; Haack-Sørensen, M.; Ekblond, A.; Kastrup, J. Bone marrow-derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure: a randomized placebo- controlled trial (MSC-HF trial). Eur Heart J. 2015, 36, 1744–1753. [Google Scholar] [CrossRef]
  60. Teerlink, J.R.; Metra, M.; Filippatos, G.S.; Davison, B.A.; Bartunek, J.; Terzic, A.; Gersh, B.J.; Povsic, T.J.; Henry, T.D.; Alexandre, B.; Homsy, C.; Edwards, C.; Seron, A.; Wijns, W.; Cotter, G.; CHART Investigators. Benefit of cardiopoietic mesenchymal stem cell therapy on left ventricular remodelling: results from the Congestive Heart Failure Cardiopoietic Regenerative Therapy (CHART-1) study. Eur J Heart Fail. 2017, 19, 1520–1529. [Google Scholar] [CrossRef]
  61. Perin, E.C.; Borow, K.M.; Silva, G.V.; DeMaria, A.N.; Marroquin, O.C.; Huang, P.P.; Traverse, J.H.; Krum, H.; Skerrett, D.; Zheng, Y.; Willerson, J.T.; Itescu, S.; Henry, T.D. A Phase II Dose-Escalation Study of Allogeneic Mesenchymal Precursor Cells in Patients With Ischemic or Nonischemic Heart Failure. Circ Res. 2015, 117, 576–584. [Google Scholar] [CrossRef]
  62. Kastrup, J.; Haack-Sørensen, M.; Juhl, M.; Harary Søndergaard, R.; Follin, B.; Drozd Lund, L.; Mønsted Johansen, E.; Ali Qayyum, A.; Bruun Mathiasen, A.; Jørgensen, E.; Helqvist, S.; Jørgen Elberg, J.; Bruunsgaard, H.; Ekblond, A. Cryopreserved Off-the-Shelf Allogeneic Adipose-Derived Stromal Cells for Therapy in Patients with Ischemic Heart Disease and Heart Failure-A Safety Study. Stem Cells Transl Med. 2017, 6, 1963–1971. [Google Scholar] [CrossRef] [PubMed]
  63. Kastrup, J.; Schou, M.; Gustafsson, I.; Nielsen, O.W.; Møgelvang, R.; Kofoed, K.F.; Kragelund, C.; Hove, J.D.; Fabricius-Bjerre, A.; Heitman, M.; Haack-Sørensen, M.; Lund, L.D.; Johansen, E.M.; Qayyum, A.A.; Mathiasen, A.B.; Ekblond, A. Rationale and Design of the First Double- Blind, Placebo-Controlled Trial with Allogeneic Adipose Tissue-Derived Stromal Cell Therapy in Patients with Ischemic Heart Failure: A Phase II Danish Multicentre Study. Stem Cells Int. 2017, 2017, 8506370. [Google Scholar] [CrossRef]
  64. Bartolucci, J.; Verdugo, F.J.; González, P.L.; Larrea, R.E.; Abarzua, E.; Goset, C.; Rojo, P.; Palma, I.; Lamich, R.; Pedreros, P.A.; Valdivia, G.; Lopez, V.M.; Nazzal, C.; Alcayaga-Miranda, F.; Cuenca, J.; Brobeck, M.J.; Patel, A.N.; Figueroa, F.E.; Khoury, M. Safety and Efficacy of the Intravenous Infusion of Umbilical Cord Mesenchymal Stem Cells in Patients With Heart Failure: A Phase 1/2 Randomized Controlled Trial (RIMECARD Trial [Randomized Clinical Trial of Intravenous Infusion Umbilical Cord Mesenchymal Stem Cells on Cardiopathy]). Circ Res. 2017, 121, 1192–1204. [Google Scholar] [CrossRef] [PubMed]
  65. Tompkins, B.A.; Rieger, A.C.; Florea, V.; Banerjee, M.N.; Natsumeda, M.; Nigh, E.D.; Landin, A.M.; Rodriguez, G.M.; Hatzistergos, K.E.; Schulman, I.H.; Hare, J.M. Comparison of Mesenchymal Stem Cell Efficacy in Ischemic Versus Nonischemic Dilated Cardiomyopathy. J Am Heart Assoc. 2018, 7, e008460. [Google Scholar] [CrossRef] [PubMed]
  66. Heldman, A.W.; DiFede, D.L.; Fishman, J.E.; Zambrano, J.P.; Trachtenberg, B.H.; Karantalis, V.; Mushtaq, M.; Williams, A.R.; Suncion, V.Y.; McNiece, I.K.; Ghersin, E.; Soto, V.; Lopera, G.; Miki, R.; Willens, H.; Hendel, R.; Mitrani, R.; Pattany, P.; Feigenbaum, G.; Oskouei, B.; Byrnes, J.; Lowery, M.H.; Sierra, J.; Pujol, M.V.; Delgado, C.; Gonzalez, P.J.; Rodriguez, J.E.; Bagno, L.L.; Rouy, D.; Altman, P.; Foo, C.W.; da Silva, J.; Anderson, E.; Schwarz, R.; Mendizabal, A.; Hare, J.M. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA. 2014, 311, 62–73. [Google Scholar] [CrossRef]
  67. Karantalis, V.; DiFede, D.L.; Gerstenblith, G.; Pham, S.; Symes, J.; Zambrano, J.P.; Fishman, J.; Pattany, P.; McNiece, I.; Conte, J.; Schulman, S.; Wu, K.; Shah, A.; Breton, E.; Davis- Sproul, J.; Schwarz, R.; Feigenbaum, G.; Mushtaq, M.; Suncion, V.Y.; Lardo, A.C.; Borrello, I.; Mendizabal, A.; Karas, T.Z.; Byrnes, J.; Lowery, M.; Heldman, A.W.; Hare, J.M. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The Prospective Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trial. Circ Res. 2014, 114, 1302–1310. [Google Scholar] [CrossRef]
  68. Hare, J.M.; Fishman, J.E.; Gerstenblith, G.; DiFede Velazquez, D.L.; Zambrano, J.P.; Suncion, V.Y.; Tracy, M.; Ghersin, E.; Johnston, P.V.; Brinker, J.A.; Breton, E.; Davis- Sproul, J.; Schulman, I.H.; Byrnes, J.; Mendizabal, A.M.; Lowery, M.H.; Rouy, D.; Altman, P.; Wong Po Foo, C.; Ruiz, P.; Amador, A.; Da Silva, J.; McNiece, I.K.; Heldman, A.W.; George, R.; Lardo, A. Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012, 308, 2369–2379. [Google Scholar] [CrossRef]
  69. Xiao, W.; Guo, S.; Gao, C.; Dai, G.; Gao, Y.; Li, M.; Wang, X.; Hu, D. A Randomized Comparative Study on the Efficacy of Intracoronary Infusion of Autologous Bone Marrow Mononuclear Cells and Mesenchymal Stem Cells in Patients With Dilated Cardiomyopathy. Int Heart J. 2017, 58, 238–244. [Google Scholar] [CrossRef]
  70. Florea, V.; Rieger, A.C.; DiFede, D.L.; El-Khorazaty, J.; Natsumeda, M.; Banerjee, M.N.; Tompkins, B.A.; Khan, A.; Schulman, I.H.; Landin, A.M.; Mushtaq, M.; Golpanian, S.; Lowery, M.H.; Byrnes, J.J.; Hendel, R.C.; Cohen, M.G.; Valasaki, K.; Pujol, M.V.; Ghersin, E.; Miki, R.; Delgado, C.; Abuzeid, F.; Vidro-Casiano, M.; Saltzman, R.G.; DaFonseca, D.; Caceres, L.V.; Ramdas, K.N.; Mendizabal, A.; Heldman, A.W.; Mitrani, R.D.; Hare, J.M. Dose Comparison Study of Allogeneic Mesenchymal Stem Cells in Patients With Ischemic Cardiomyopathy (The TRIDENT Study). Circ Res. 2017, 121, 1279–1290. [Google Scholar] [CrossRef]
  71. Subherwal, S.; Patel, M.R.; Kober, L.; Peterson, E.D.; Bhatt, D.L.; Gislason, G.H.; Olsen, A.M.; Jones, W.S.; Torp- Pedersen, C.; Fosbol, E.L. Peripheral artery disease is a coronary heart disease risk equivalent among both men and women: results from a nationwide study. Eur J Prev Cardiol. 2015, 22, 317–325. [Google Scholar] [CrossRef]
  72. Lu, D.; Chen, B.; Liang, Z.; Deng, W.; Jiang, Y.; Li, S.; Xu, J.; Wu, Q.; Zhang, Z.; Xie, B.; Chen, S. Comparison of bone marrow mesenchymal stem cells with bone marrow- derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Res Clin Pract. 2011, 92, 26–36. [Google Scholar] [CrossRef]
  73. Gupta, P.K.; Chullikana, A.; Parakh, R.; Desai, S.; Das, A.; Gottipamula, S.; Krishnamurthy, S.; Anthony, N.; Pherwani, A.; Majumdar, A.S. A double blind randomized placebo controlled phase I/II study assessing the safety and efficacy of allogeneic bone marrow derived mesenchymal stem cell in critical limb ischemia. J Transl Med. 2013, 11, 143. [Google Scholar] [CrossRef]
  74. Bura, A.; Planat-Benard, V.; Bourin, P.; Silvestre, J.S.; Gross, F.; Grolleau, J.L.; Saint-Lebese, B.; Peyrafitte, J.A.; Fleury, S.; Gadelorge, M.; Taurand, M.; Dupuis-Coronas, S.; Leobon, B.; Casteilla, L. Phase I trial: the use of autologous cultured adipose-derived stroma/stem cells to treat patients with non-revascularizable critical limb ischemia. Cytotherapy. 2014, 16, 245–257. [Google Scholar] [CrossRef] [PubMed]
  75. Xie, B.; Luo, H.; Zhang, Y.; Wang, Q.; Zhou, C.; Xu, D. Autologous Stem Cell Therapy in Critical Limb Ischemia: A Meta-Analysis of Randomized Controlled Trials. Stem Cells Int. 2018, 2018, 7528464. [Google Scholar] [CrossRef]
  76. Sacco, R.L.; Kasner, S.E.; Broderick, J.P.; Caplan, L.R.; Connors, J.J.; Culebras, A.; Elkind, M.S.; George, M.G.; Hamdan, A.D.; Higashida, R.T.; Hoh, B.L.; Janis, L.S.; Kase, C.S.; Kleindorfer, D.O.; Lee, J.M.; Moseley, M.E.; Peterson, E.D.; Turan, T.N.; Valderrama, A.L.; Vinters, H.V.; American Heart Association Stroke Council, Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Radiology and Intervention; Council on Cardiovascular and Stroke Nursing; Council on Epidemiology and Prevention; Council on Peripheral Vascular Disease; Council on Nutrition, Physical Activity and Metabolism. An updated definition of stroke for the 21st century: a statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2013, 44, 2064–2089. [Google Scholar] [CrossRef] [PubMed]
  77. Johnson, W.; Onuma, O.; Owolabi, M.; Sachdev, S. Stroke: a global response is needed. Bull World Health Organ. 2016, 94, 634-634A. [Google Scholar] [CrossRef]
  78. Steinberg, G.K.; Kondziolka, D.; Wechsler, L.R.; Lunsford, L.D.; Coburn, M.L.; Billigen, J.B.; Kim, A.S.; Johnson, J.N.; Bates, D.; King, B.; Case, C.; McGrogan, M.; Yankee, E.W.; Schwartz, N.E. Clinical Outcomes of Transplanted Modified Bone Marrow-Derived Mesenchymal Stem Cells in Stroke: A Phase 1/2a Study. Stroke. 2016, 47, 1817–1824. [Google Scholar] [CrossRef] [PubMed]
  79. Hess, D.C.; Wechsler, L.R.; Clark, W.M.; Savitz, S.I.; Ford, G.A.; Chiu, D.; Yavagal, D.R.; Uchino, K.; Liebeskind, D.S.; Auchus, A.P.; Sen, S.; Sila, C.A.; Vest, J.D.; Mays, R.W. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017, 16, 360–368. [Google Scholar] [CrossRef]
  80. Levy, M.L.; Crawford, J.R.; Dib, N.; Verkh, L.; Tankovich, N.; Cramer, S.C. Phase I/II Study of Safety and Preliminary Efficacy of Intravenous Allogeneic Mesenchymal Stem Cells in Chronic Stroke. Stroke. 2019, 50, 2835–2841. [Google Scholar] [CrossRef] [PubMed]
  81. Savitz, S.I.; Yavagal, D.; Rappard, G.; Likosky, W.; Rutledge, N.; Graffagnino, C.; Alderazi, Y.; Elder, J.A.; Chen, P.R.; Budzik RFJr Tarrel, R.; Huang, D.Y.; Hinson, J.M., Jr. A Phase 2 Randomized, Sham-Controlled Trial of Internal Carotid Artery Infusion of Autologous Bone Marrow-Derived ALD-401 Cells in Patients With Recent Stable Ischemic Stroke (RECOVER-Stroke). Circulation. 2019, 139, 192–205. [Google Scholar] [CrossRef]
  82. Lee, J.S.; Hong, J.M.; Moon, G.J.; Lee, P.H.; Ahn, Y.H.; Bang, O.Y.; STARTING collaborators. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells. 2010, 28, 1099–1106. [Google Scholar] [CrossRef] [PubMed]
  83. Ljujic, B.; Milovanovic, M.; Volarevic, V.; Murray, B.; Bugarski, D.; Przyborski, S.; Arsenijevic, N.; Lukic, M.L.; Stojkovic, M. Human mesenchymal stem cells creating an immunosuppressive environment and promote breast cancer in mice. Sci Rep. 2013, 3, 2298. [Google Scholar] [CrossRef]
  84. Gazdic, M.; Simovic Markovic, B.; Jovicic, N.; Misirkic- Marjanovic, M.; Djonov, V.; Jakovljevic, V.; Arsenijevic, N.; Lukic, M.L.; Volarevic, V. Mesenchymal Stem Cells Promote Metastasis of Lung Cancer Cells by Downregulating Systemic Antitumor Immune Response. Stem Cells Int. 2017, 2017, 6294717. [Google Scholar] [CrossRef] [PubMed]
  85. Volarevic, V.; Markovic, B.S.; Gazdic, M.; Volarevic, A.; Jovicic, N.; Arsenijevic, N.; Armstrong, L.; Djonov, V.; Lako, M.; Stojkovic, M. Ethical and Safety Issues of Stem Cell-Based Therapy. Int J Med Sci. 2018, 15, 36–45. [Google Scholar] [CrossRef]
  86. Miloradovic, D.; Miloradovic, D.; Markovic, B.S.; Acovic, A.; Harrell, C.R.; Djonov, V.; Arsenijevic, N.; Volarevic, V. The Effects of Mesenchymal Stem Cells on Antimelanoma Immunity Depend on the Timing of Their Administration. Stem Cells Int. 2020, 2020, 8842659. [Google Scholar] [CrossRef]
Figure 1. The main mechanisms responsible for MSC- based treatment of CVDs. Transplanted MSCs can contribute to cardiovascular repair and regeneration by forming endothelial cells (ECs), smooth muscle cells (SMCs) and cardiomyocytes (CMs) (differentiation). Additionally, MSCs produce a broad range of cytokines, chemokines, and growth factors that resolve inflammation, inhibit apoptosis and fibrosis, promote angiogenesis, and induce endogenous cardiomyocyte regeneration.
Figure 1. The main mechanisms responsible for MSC- based treatment of CVDs. Transplanted MSCs can contribute to cardiovascular repair and regeneration by forming endothelial cells (ECs), smooth muscle cells (SMCs) and cardiomyocytes (CMs) (differentiation). Additionally, MSCs produce a broad range of cytokines, chemokines, and growth factors that resolve inflammation, inhibit apoptosis and fibrosis, promote angiogenesis, and induce endogenous cardiomyocyte regeneration.
Jmms 08 00023 g001
Table 1. Clinical trials using MSCs to treat cardiac diseases.
Table 1. Clinical trials using MSCs to treat cardiac diseases.
Jmms 08 00023 i001
Table 2. Clinical trials using MSCs in peripheral arterial disease and stroke therapies.
Table 2. Clinical trials using MSCs in peripheral arterial disease and stroke therapies.
Jmms 08 00023 i002

Share and Cite

MDPI and ACS Style

Miloradovic, D.R.; Pavlovic, D.R.; Stojkovic, M.B.; Bojic, S.B.; Volarevic, V.B.; Jankovic, M.M.G.; Ljujic, B.T. Therapeutic Efficacy of Mesenchymal Stem Cells for Cardiovascular Diseases. J. Mind Med. Sci. 2021, 8, 179-190. https://doi.org/10.22543/7674.82.P179190

AMA Style

Miloradovic DR, Pavlovic DR, Stojkovic MB, Bojic SB, Volarevic VB, Jankovic MMG, Ljujic BT. Therapeutic Efficacy of Mesenchymal Stem Cells for Cardiovascular Diseases. Journal of Mind and Medical Sciences. 2021; 8(2):179-190. https://doi.org/10.22543/7674.82.P179190

Chicago/Turabian Style

Miloradovic, Dragana Radoje, Dragica Radoje Pavlovic, Miodrag Bozidar Stojkovic, Sanja Bratislav Bojic, Vladislav Bogdan Volarevic, Marina Milosav Gazdic Jankovic, and Biljana Tomislav Ljujic. 2021. "Therapeutic Efficacy of Mesenchymal Stem Cells for Cardiovascular Diseases" Journal of Mind and Medical Sciences 8, no. 2: 179-190. https://doi.org/10.22543/7674.82.P179190

APA Style

Miloradovic, D. R., Pavlovic, D. R., Stojkovic, M. B., Bojic, S. B., Volarevic, V. B., Jankovic, M. M. G., & Ljujic, B. T. (2021). Therapeutic Efficacy of Mesenchymal Stem Cells for Cardiovascular Diseases. Journal of Mind and Medical Sciences, 8(2), 179-190. https://doi.org/10.22543/7674.82.P179190

Article Metrics

Back to TopTop