Next Article in Journal
The Role of TLR-2 in Lethal COVID-19 Disease Involving Medullary and Resident Lung Megakaryocyte Up-Regulation in the Microthrombosis Mechanism
Previous Article in Journal
Multiple Myeloma: The Role of Autologous Stem Cell Transplantation in the Era of Immunotherapy
Previous Article in Special Issue
Mesenchymal Stem Cell Exosomes Enhance Posterolateral Spinal Fusion in a Rat Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Strategies in Mesenchymal Stem Cell-Based Cardiovascular Therapeutics

by
Rishabh Kumar
1,
Nitin Mishra
1,
Talan Tran
2,
Munish Kumar
1,*,
Sivakumar Vijayaraghavalu
3 and
Narasimman Gurusamy
2,*
1
Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
2
Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328-2018, USA
3
Department of Life Sciences, Manipur University, Imphal 795003, India
*
Authors to whom correspondence should be addressed.
Cells 2024, 13(10), 855; https://doi.org/10.3390/cells13100855
Submission received: 29 January 2024 / Revised: 13 May 2024 / Accepted: 15 May 2024 / Published: 17 May 2024
(This article belongs to the Collection Stem Cells in Tissue Engineering and Regeneration)

Abstract

:
Cardiovascular diseases continue to challenge global health, demanding innovative therapeutic solutions. This review delves into the transformative role of mesenchymal stem cells (MSCs) in advancing cardiovascular therapeutics. Beginning with a historical perspective, we trace the development of stem cell research related to cardiovascular diseases, highlighting foundational therapeutic approaches and the evolution of cell-based treatments. Recognizing the inherent challenges of MSC-based cardiovascular therapeutics, which range from understanding the pro-reparative activity of MSCs to tailoring patient-specific treatments, we emphasize the need to refine the pro-regenerative capacity of these cells. Crucially, our focus then shifts to the strategies of the fourth generation of cell-based therapies: leveraging the secretomic prowess of MSCs, particularly the role of extracellular vesicles; integrating biocompatible scaffolds and artificial sheets to amplify MSCs’ potential; adopting three-dimensional ex vivo propagation tailored to specific tissue niches; harnessing the promise of genetic modifications for targeted tissue repair; and institutionalizing good manufacturing practice protocols to ensure therapeutic safety and efficacy. We conclude with reflections on these advancements, envisaging a future landscape redefined by MSCs in cardiovascular regeneration. This review offers both a consolidation of our current understanding and a view toward imminent therapeutic horizons.

1. Introduction

Cardiovascular diseases remain leading causes of morbidity and mortality worldwide, presenting an unrelenting challenge to the medical community [1]. In recent decades, traditional treatments, including pharmaceuticals, lifestyle modifications, and surgical interventions, have undeniably advanced. However, the complex nature of cardiovascular diseases, which are characterized by damage to the myocardium, limited regenerative potential, and progressive heart failure, demands innovative therapeutic approaches [2]. Stem cells, with their intrinsic ability to self-renew and differentiate into various cell types, have presented a promising avenue for cardiovascular regeneration [3]. The initial optimism surrounding stem cell-based therapies stemmed from the prospect of regenerating damaged myocardial tissue, thereby potentially reversing the effects of conditions such as myocardial infarction [3].
Mesenchymal stem cells (MSCs) have been at the forefront of this research, primarily due to their multilineage differentiation potential, immunomodulatory properties, and relative ease of isolation from various tissues [4]. Early studies focused on the direct transplantation of MSCs into damaged cardiac tissue, with the aim of replacing lost cardiomyocytes and restoring heart function [4]. However, as the field evolved, it became evident that the therapeutic benefits of MSCs extended beyond mere cell replacement. MSCs were found to play a pivotal role in modulating the cardiac microenvironment through paracrine signaling, thus aiding in tissue repair and reducing inflammation [4]. The next phase of research shifted toward understanding these intricate mechanisms, paving the way for the second and third generations of cell-based therapies, which focused on enhancing the efficacy and delivery of stem cell-derived factors [5]. Today, as we stand at the precipice of the fourth generation of stem cell-based therapies, the emphasis is on harnessing the full therapeutic potential of MSCs, fine-tuning their properties, and developing innovative strategies to address the multifaceted challenges of cardiovascular diseases.
To conduct a thorough review of MSC-based cardiovascular therapeutics, we systematically selected and analyzed relevant articles using databases like PubMed, Scopus, and Web of Science. Our search was guided by specific keywords related to MSCs, cardiovascular diseases, and stem cell therapies. We included the most recent peer-reviewed articles, reviews, and clinical trial reports, focusing on their relevance, innovation, and contributions to the field. Emphasis was placed on articles detailing emerging technologies in tissue engineering, genetic modifications of MSCs, and cell-free therapies. Each article was critically evaluated for methodological soundness and its implications for future research and clinical applications, ensuring a comprehensive overview of the latest advancements and gaps in MSC-based cardiovascular therapies.

2. Past and Present Stem Cell-Based Therapeutic Approaches

The historical journey of stem cell therapies in cardiovascular medicine began when scientists explored bone marrow transplantation for the treatment of blood disorders [6]. The discovery of embryonic stem cells toward the end of the 20th century opened up new possibilities for regenerative medicine.
Among the earliest and most significant developments was the application of bone marrow-derived stem cells for myocardial repair. Pioneering studies such as the BOOST trial [7] underscored the potential of bone marrow-derived mononuclear cells to enhance left ventricular ejection fraction following myocardial infarction. Despite these promising results, subsequent larger trials like REPAIR-AMI presented a more nuanced picture, showing modest improvements in specific patient subsets and raising questions about the efficacy and applicability of these therapies [8,9]. Embryonic stem cells, known for their pluripotency, emerged as a promising candidate in cardiovascular therapeutics [10]. However, their use was fraught with ethical concerns, especially regarding the use of embryonic material [11]. Additionally, the risk of teratoma formation and immunological complications associated with embryonic stem cells posed significant hurdles to their direct clinical application [12]. These challenges necessitated a shift toward more ethically acceptable and patient-specific approaches.

2.1. Induced Pluripotent Stem Cells (iPSCs)

The development of induced pluripotent stem cells (iPSCs) by Shinya Yamanaka in 2006 significantly advanced stem cell research, offering a potent alternative to embryonic stem cells without associated ethical concerns [13]. iPSCs are reprogrammed from adult cells to a pluripotent state, facilitating the generation of patient-specific cells that bypass immunological issues inherent in embryonic sources [14]. iPSC-derived cardiomyocytes have emerged as crucial tools in drug testing, disease modeling, and cardiac therapy [15,16]. Due to their ability to differentiate into cardiomyocytes, iPSCs have opened new pathways for cardiac repair and regeneration, showcasing their utility in personalized medicine for designing patient-specific treatments [17]. Moreover, iPSC-derived cardiac cells have been instrumental in advancing our understanding of cardiac diseases at a molecular level, enhancing disease modeling and fostering the development of targeted therapies [18,19].
To enhance the efficacy, safety, and delivery of iPSC-based therapies for cardiovascular applications, several innovative approaches are being pursued. (a) Three-Dimensional bioprinting: This technology that constructs functional tissues that mimic natural cardiovascular structures, allowing for the precise placement of iPSC-derived cells in a biomaterial matrix. It enhances cell maturation, integration, and function in a controlled environment [20]. (b) Tissue engineering: Techniques are developed to create viable cardiac tissue grafts from iPSCs, using biodegradable scaffolds that support cell growth and integration. These tissues can replace a damaged myocardium and incorporate features like electrical conductivity to boost functional integration [21,22]. (c) Genetic modifications: iPSCs are genetically modified to improve survival, proliferation, and differentiation. Techniques like CRISPR/Cas9 are employed to enhance cellular properties and reduce sources of risk such as immunogenicity [23]. (d) Delivery mechanisms: Innovative delivery methods like catheter-based injections, magnetic targeting, and microcarriers are used to enhance the precision and retention of iPSC-derived cells in the myocardium [24]. Despite progress, challenges like ensuring long-term cell survival, preventing immune rejection, and adhering to regulatory standards remain [24]. Ongoing research aims to improve safety profiles and optimize cellular functions, moving toward more effective, safe, and customized iPSC-based cardiovascular therapies [25].

2.2. Cardiosphere-Derived Cells

Cardiosphere-derived cells (CDCs) are sourced from cardiac biopsies and hold significant potential for cardiac regeneration, bridging traits between MSCs and cardiac progenitor cells [26]. These cells have been integral in promoting angiogenesis, reducing fibrosis, and modulating immune responses in post-myocardial infarction scenarios, with their paracrine effects, particularly through the secretion of exosomes and microRNAs, playing pivotal roles in cardiac repair [26]. The CADUCEUS trial notably demonstrated the safety and effectiveness of CDCs in myocardial infarction patients by significantly reducing scar mass and increasing viable heart tissue, highlighting their potential in enhancing myocardial regeneration [8]. Apart from CDCs, other types of cardiac progenitor cells have been explored for their regenerative potential. A study by Mishra et al. [10] demonstrated the therapeutic potential of a unique population of cardiac progenitor cells identified by their expression of Islet-1 in repairing a damaged myocardium. These cells were found to contribute to the formation of new cardiac muscle and blood vessels in animal models, opening new avenues for clinical applications in human heart repair [10].

2.3. Endothelial Progenitor Cells

The angiogenic capabilities of endothelial progenitor cells (EPCs) are essential to restoring vascular health [27]. Derived from bone marrow, EPCs repair and maintain the endothelial lining and are crucial for treating cardiovascular diseases marked by endothelial dysfunction [27]. They express specific markers such as CD34, VEGFR2, and CD133, which facilitate their identification and differentiation within the vascular system [28]. Therapeutically, EPCs are utilized in novel treatments like EPC-capture stents, which enhance vascular healing by attracting EPCs to damaged sites, thereby promoting endothelialization and reducing stent thrombosis risks [29]. Additionally, optimizing EPC mobilization and homing—which are affected by physical activity, oxidative stress, and pharmacological interventions—is a vibrant research area, pushing forward cell-based therapies that promise new treatments for complex cardiovascular ailments [30].

2.4. MSCs

MSCs are multipotent stromal cells capable of differentiating into a variety of cell types, such as osteoblasts, chondrocytes, myocytes, and adipocytes [31]. Originating from sources including bone marrow, adipose tissue, and umbilical cord blood, MSCs are characterized by their ability to self-renew, minimal immunogenicity, and pronounced immunomodulatory properties, making them particularly valuable for regenerative medicine applications, notably in cardiovascular therapeutics [31]. In the realm of cardiovascular disease treatment, MSCs offer several advantageous properties: they modulate immune responses to reduce inflammation; attenuate fibrosis, which can impair cardiac function; promote angiogenesis, which is necessary for tissue repair and improved blood circulation; and potentially differentiate into cardiomyocyte-like cells to directly aid cardiac repair [32]. While preclinical studies affirm MSCs’ efficacy in enhancing cardiac function post myocardial infarction through engraftment and the facilitation of cardiac repair, their clinical utility is tempered by challenges such as the low retention rate and survival of transplanted cells within the ischemic myocardial environment [33,34,35,36,37,38]. In recent years, focus has shifted toward optimizing the therapeutic potential of stem cells. This involves ensuring better cell survival post transplantation, enhancing the cells’ reparative properties, understanding patient-specific factors, and developing combinatorial therapies involving scaffolds, exosomes, and tissue engineering [39].
Ongoing research and clinical trials continue to explore MSCs’ therapeutic potential, underscoring their significant prospective impact on the treatment of myocardial infarction and other cardiac ailments. The MSC-HF Trial, a randomized, double-blind, placebo-controlled study, evaluated the 4-year outcomes of intramyocardial injections of autologous bone marrow-derived MSCs in patients with ischemic heart failure [40]. It included 60 patients with significant heart failure, showing that MSC treatment led to significant improvements in left ventricular end-systolic volume, ejection fraction, stroke volume, and myocardial mass over 12 months, along with reduced scar tissue and enhanced quality of life [40]. After four years, the data indicated fewer hospitalizations for angina, with no adverse effects noted [40]. The C-CURE Trial assessed the safety and efficacy of MSCs treated with a cardiogenic cocktail in chronic heart failure patients [41]. This multicenter, randomized trial showed improvements in cardiac function, 6-minute walk distance, and composite clinical scores, underscoring the potential of cardiopoietic stem cell therapy [41]. The CHART-1 Trial, another randomized, double-blind study, did not meet its primary efficacy endpoint but revealed potential benefits in specific patient subgroups with certain baseline ventricular volumes, pointing to the possibility of targeted therapy [42]. The ongoing DREAM-HF Trial, the largest among the reviewed studies, is examining the efficacy and safety of mesenchymal precursor cells in advanced chronic heart failure [43]. Although not all primary endpoints were met, there were indications of reduced hospital readmissions and improved cardiac function in certain subgroups, highlighting the nuanced potential of MSC therapies in heart failure [43]. These studies collectively advance the understanding of MSC-based therapies in cardiac care, showing significant promise and guiding future research toward more targeted and personalized approaches.

2.5. MSC-Derived Extracellular Vesicles

Extracellular vesicles (EVs) derived from MSCs have emerged as promising therapeutic agents in the field of cardiovascular diseases [44,45]. EVs, encompassing exosomes and microvesicles, are nanosized lipid bilayer-enclosed structures capable of transferring proteins, lipids, and nucleic acids between cells, thereby modulating cellular functions and responses [45]. In the context of cardiovascular diseases, MSC-derived EVs exhibit potential in enhancing angiogenesis, reducing fibrosis, and modulating inflammatory responses, which are crucial for myocardial repair and regeneration [46]. The therapeutic efficacy of EVs is largely attributed to their ability to mirror the regenerative and reparative properties of their parent MSCs while offering an advantage in terms of safety and feasibility as they lack the risks associated with stem cell implantation, such as tumorigenicity and immune rejection [47]. This study has investigated the use of cardiomyocyte-targeting exosomes loaded with microRNA-302 (miR302) for treating myocardial ischemia/reperfusion injury, finding that these engineered exosomes enhanced cardiomyocyte proliferation and activity and reduced heart damage in vitro and in mice [47]. Recent advances have focused on optimizing the isolation, characterization, and scalability of EV production under good manufacturing practice (GMP) conditions [48]. Furthermore, research is exploring the potential of engineering EVs to enhance their targeting efficiency and therapeutic payload, making them a highly versatile and promising tool in regenerative medicine for cardiovascular therapies [48].

3. Biological Properties of MSCs

The therapeutic potential of MSCs in cardiovascular diseases derives from their unique biological properties and actions, which are essential for tissue repair, and researchers are seeking insights into optimizing their clinical use. This section details the key attributes and mechanisms that make them suitable for cardiovascular therapies.

3.1. Multipotency and Differentiation

MSCs have the ability to differentiate into cardiomyocytes, endothelial cells, and smooth muscle cells, key components in cardiac tissue repair and vascular regeneration [31]. For MSCs, the process of differentiating into these lineages is governed by a complex interplay of signaling pathways and environmental cues. For instance, the transformation of MSCs into cardiomyocytes often involves the modulation of pathways like Wnt/β-catenin, JAK/STAT, and transforming growth factor-beta (TGF-β) [49,50,51]. A study by Quevedo et al. [52] highlights how MSCs can be induced to differentiate into cardiomyocyte-like cells, subsequently integrating into cardiac tissue and contributing to the restoration of myocardial function in heart failure models. MSC differentiation into endothelial cells, which are essential for angiogenesis and vascular repair, is typically regulated by factors such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2, and TGF-β, which activate endothelial markers like von Willebrand factor (vWF), PECAM-1, and vascular endothelial cadherin [53,54]. Similarly, MSC differentiation into smooth muscle cells involves cues from the TGF-β superfamily and PDGF-BB, promoting the expression of smooth muscle markers like α-SMA and calponin [55,56].
Further studies [57] have shed light on the molecular underpinnings of these differentiation pathways. These insights are crucial for developing targeted therapies. For example, understanding the specific signaling molecules and transcription factors, such as stromal-derived factor-1 (SDF-1) and bone morphogenic protein-2, involved in MSC differentiation can lead to the development of biomaterials or scaffold-based approaches that mimic the natural niches of these cells, thereby enhancing their differentiation efficiency and therapeutic efficacy in situ [58]. Moreover, genetic engineering techniques, such as CRISPR/Cas9 and RNA interference, are being explored for the manipulation of these molecular pathways, enhancing the precision of and control over MSC differentiation [59]. The following studies present significant advancements in the field of MSC differentiation and therapeutic applications facilitated by CRISPR/Cas9 technology. Shahabipour et al. [60] developed a CRISPR/Cas9-mediated strategy to insert a DMP1 promoter-driven GFP-DsRed reporter into MSCs, providing a real-time indicator of osteoblast differentiation. Another study [61] explored the role of extracellular vesicles from human-induced pluripotent stem cell-derived MSCs in protecting against renal ischemia–reperfusion injury, highlighting a novel anti-necroptosis mechanism mediated by SP1 delivery and subsequent sphingosine kinase 1 activation. Meshitsuka et al. [62] demonstrated that the CRISPR/Cas9 and AAV-mediated insertion of a B2 microglobulin-HLA-G fusion gene into MSCs could prevent allogeneic rejection, enhancing their utility in off-the-shelf cell therapies. MSC genome editing through a CRISPR-Cas9 ribonucleoprotein delivery method reduced cytotoxicity and enhanced their therapeutic potential [63]. Collectively, these studies underscore the versatility of CRISPR/Cas9 in enhancing MSC applications through precise genetic modifications and functional enhancements.

3.2. Immunomodulation

Immunomodulation by MSCs involves the attenuation of inflammation and disease progression [64]. MSCs can interact with various components of the immune system, modulating their activity and thereby reducing inflammation and promoting tissue repair [65]. The immunomodulatory function of MSCs is multifaceted, involving both the secretion of soluble factors and direct cell-to-cell interactions [32]. These cells can secrete a wide range of cytokines, chemokines, and growth factors, such as TGF-β, PGE2, IL-10, and hepatocyte growth factor (HGF), which collectively contribute to the suppression of pro-inflammatory responses and the promotion of anti-inflammatory environments [64]. This secretome alters the behavior of various immune cells, including T cells, B cells, natural killer cells, and dendritic cells, leading to a reduction in inflammatory cytokine production and the suppression of T cell proliferation and cytotoxicity [66].
Prockop and Oh [67] described MSCs as “guardians of inflammation”, highlighting their role in sensing and suppressing excessive inflammatory responses. This description underscores the dynamic and responsive nature of MSCs in modulating the immune response, adapting to the specific inflammatory milieu they encounter. TGF-β contributes to the attenuation of T lymphocyte proliferation by inducing G1 cell cycle arrest through the Jak-1/Stat-5 signaling pathway [68]. MSC-derived EVs deliver thrombospondin 1 (TSP1), which can suppress NK cell activity by modulating TGF-β/Smad signaling [69]. Shi et al. [70] elucidated interactions between MSCs and immune cells, such as the induction of regulatory T cells (Tregs) and the alteration of macrophage phenotypes from a pro-inflammatory (M1) to a regenerative and anti-inflammatory (M2) state. This shift is crucial in mitigating chronic inflammation and facilitating the healing process in cardiovascular tissues.
Furthermore, MSCs can exert immunomodulatory effects through direct cell-to-cell contact involving various adhesion molecules and interactions with immune cell receptors [66]. This contact-dependent mechanism is particularly important in the context of MSCs’ interactions with T cells and antigen-presenting cells, where it can lead to anergy or tolerance, further contributing to a reduction in inflammation [66]. The ability of MSCs to modulate the immune response is not only pivotal in treating inflammatory cardiovascular conditions but also enhances the compatibility and success of other stem cell-based therapies [32]. By creating a more conducive environment for tissue repair and regeneration, MSCs’ immunomodulatory properties can be leveraged to improve outcomes in a wide range of cardiovascular interventions [32].

3.3. Paracrine Signaling

MSCs secrete a diverse array of bioactive molecules, including growth factors, cytokines, and extracellular vesicles, which interact with surrounding cells and tissues to induce regenerative processes [71]. MSCs produce growth factors such as VEGF, HGF, insulin-like growth factor (IGF), and platelet-derived growth factor (PDGF) [72]. These factors play critical roles in promoting angiogenesis, the formation of new blood vessels, which is essential for repairing ischemic heart tissues. Angiogenesis facilitates the delivery of oxygen and nutrients to damaged areas, thereby supporting tissue regeneration [72]. Additionally, MSCs secrete cytokines and chemokines that modulate the local immune environment. These include anti-inflammatory cytokines like interleukin-10 (IL-10) and TGF-β, which help mitigate inflammation and fibrosis in the heart, conditions commonly associated with various cardiovascular pathologies [32]. Harrell et al. [73] provide deeper insights into the specific paracrine factors secreted by MSCs and their effects on cardiac tissues.

3.4. Extracellular Vesicles and Secretome

MSC-derived EVs, encompassing exosomes and microvesicles, serve as critical conveyors of bioactive molecules including microRNAs, proteins, and lipids that can exert profound influences on recipient cells and tissues [74]. EVs from MSCs are equipped with a diverse array of signaling molecules such as microRNAs that can regulate gene expression in recipient cells, leading to altered cellular behavior such as enhanced survival, reduced apoptosis, and increased angiogenic potential [74]. This capacity to modulate gene expression is crucial in orchestrating tissue repair mechanisms, especially in an ischemic and injured myocardium where the restoration of vascular supply and cellular function is essential [74].
Sahoo et al. [75] demonstrated the proangiogenic activity of EVs derived from human CD34+ stem cells, highlighting their potential in promoting the formation of new blood vessels in damaged cardiac tissue. This angiogenic property is particularly valuable in post-myocardial infarction scenarios in which the restoration of blood flow to ischemic areas is a key factor in limiting infarct size and preserving heart function. Further research by Phinney and Pittenger [76] and Baglio et al. [77] delved into the complex composition of these vesicles. Their studies elucidated the diverse range of proteins, lipids, and nucleic acids present in EVs, each contributing to their regenerative potential. For example, certain proteins within MSC-derived EVs are known to activate signaling pathways involved in cell survival and proliferation, while specific lipids may play a role in membrane interactions and fusion with target cells.
These studies also emphasize the role of the MSC secretome, which includes not only EVs but also soluble factors such as cytokines and growth factors. This secretome can modulate the immune response, reduce inflammation, and enhance the regenerative capacity of heart tissue by recruiting and activating resident cardiac progenitor cells. Understanding the molecular constituents of MSC-derived EVs and their secretome, and how these constituents interact with and influence cardiac cells, opens up novel avenues for therapeutic interventions. By harnessing and potentially engineering these vesicles, it is possible to develop targeted therapies that deliver specific regenerative molecules directly to the site of injury, thereby enhancing the repair and regeneration of cardiac tissues in a more efficient and controlled manner.
Recent studies have highlighted the specific role of miRNAs in modulating the functions and therapeutic potential of MSCs. Zheng et al. [78] found that exosomal miR-9-5p from iPSC-derived MSCs mitigates doxorubicin-induced cardiomyopathy by preventing cardiomyocyte senescence, primarily through the inhibition of the VPO1/ERK signaling pathway. Human umbilical cord-derived MSCs alleviated myocardial fibrosis and restored miRNA-133a expression in diabetic cardiomyopathy, which positively influenced fibrosis markers and inflammatory mediators in a diabetic mouse model [79]. Additionally, the immunoregulatory properties of exosomal miRNAs from bone marrow MSCs overexpressing IDO1 demonstrated their potential to modulate immune responses and improve allogeneic heart transplantation outcomes by influencing key immune-related proteins and miRNAs [80]. Zhu et al. [81] reported that suppressing miR-873-5p rejuvenates aging MSCs, enhancing their functionality and therapeutic efficacy for myocardial infarction repair through modulating autophagy via the AMPK signaling pathway.
In addition to the differentiation of MSCs and the secretion of paracrine factors, MSCs can facilitate cardiac repair through the transfer of mitochondria to damaged cardiac cells [38,82]. This organelle transfer can help rescue injured cardiomyocytes, improve their functional performance, and enhance cell survival under stress conditions [82]. This mechanism could be pivotal in cardiac repair as it directly addresses cellular energy deficits encountered post-injury [82]. Further, MSCs can stimulate the regeneration of endogenous cardiomyocytes through the activation and recruitment of local cardiac stem cells, which can then differentiate and replace damaged myocardial tissue [37,83].

3.5. Angiogenesis

MSCs promote the formation of new blood vessels and help restore blood supply to ischemic or damaged heart tissue [84]. They achieve this primarily through the secretion of angiogenic factors such as VEGF, FGF, and HGF [84]. These factors stimulate the proliferation and migration of endothelial cells and enhance the process of angiogenesis. Gnecchi et al. [85] showed that MSCs overexpressing Akt1 enhance myocardial protection post infarction through paracrine mechanisms, reducing apoptosis and improving cardiac function, proposing their secreted factors as potential therapeutic agents for ischemic damage. Kinnaird et al. [86] have shown that MSC secrete arteriogenic cytokines like VEGF and bFGF, enhancing the proliferation of vascular cells through paracrine mechanisms rather than direct incorporation into vessels. In a mouse model, MSC injection improved limb perfusion and functionality and reduced muscle atrophy and fibrosis, underscoring the therapeutic potential of MSCs for collateral remodeling and recovery after ischemic injury [86].

3.6. Anti-Fibrotic Effects

Cardiac fibrosis, characterized by the excessive deposition of extracellular matrix (ECM) components, primarily fibrillar collagens like types I and III along with other ECM constituents such as fibronectin and elastin, leads to the stiffening of the heart muscle, impairing its function [87]. MSCs exert anti-fibrotic effects by inhibiting fibroblast proliferation and ECM deposition by promoting the secretion of matrix metalloproteinases, which break down ECM components via cardiac fibroblasts [88]. Kou et al. [89] showed that MSC-derived EVs are effective in modulating immune responses and facilitating the regeneration of various tissues, including those damaged by fibrosis. However, challenges in preparing MSC-EVs, such as ensuring consistent quality and overcoming heterogeneity, are noted as significant hurdles to their clinical application [89]. MSCs release factors such as hepatocyte growth factor (HGF) and prostaglandin E2 (PGE2), which have been shown to directly inhibit the proliferation and activation of fibroblasts into myofibroblasts [88,90]. MSCs directly inhibit the synthesis of TGF-β and also alter downstream SMAD signaling, which is crucial for the transcriptional activation of fibrotic genes [91]. MSC-derived EVs containing microRNAs such as miR-378 and miR-27b have been shown to suppress TGF-β signaling, thereby inhibiting the fibrotic response [92]. Understanding these mechanisms is crucial for developing targeted therapies to treat cardiac fibrosis, a common aftermath of various heart diseases, including myocardial infarction and hypertensive heart disease [93].

4. Homing and Migration of MSCs

MSCs have an intrinsic ability to specifically target and migrate to sites of injury or inflammation, such as damaged cardiac tissues, following systemic administration. This process is intricately regulated by a series of molecular signals involving chemokines, their receptors, and adhesion molecules [94]. Chemokines are a family of small cytokines or signaling proteins secreted by cells. The interactions between these chemokines and their receptors on MSCs play pivotal roles in guiding the migration of MSCs to injured heart tissue [95]. Chemokine receptors, such as CXCR4, CCR2, and CCR7, expressed on MSCs allow them to respond to the gradient of chemokines released from the injured or inflamed tissue, a phenomenon akin to a cellular GPS system [95].
Ruster et al. [96] demonstrated the coordinated rolling and adhesion behavior of MSCs on endothelial cells, which is a critical step in the homing process. This interaction is mediated by selectins and their ligands as well as integrins, which facilitate the initial weak binding (rolling) of MSCs on the endothelial surface, followed by firmer adhesion [96]. This adhesion is necessary for MSCs to transmigrate across the endothelial barrier and reach the site of injury [96]. Further studies by Yau et al. [97] have emphasized the roles of specific chemokines, such as SDF-1 (stromal cell-derived factor-1) and its interaction with CXCR4 on MSCs. SDF-1 is upregulated in damaged cardiac tissues and acts as a strong chemoattractant for MSCs, guiding their migration to these sites [97]. In addition to understanding these natural homing mechanisms, research is also focused on how to enhance this ability for therapeutic applications. Approaches such as preconditioning MSCs (with hypoxia or pharmacological agents), genetic modification to overexpress certain chemokine receptors, or even the use of magnetic nanoparticles for guided delivery, are being explored [94]. By enhancing their homing efficiency, it is possible to increase the therapeutic efficacy of MSCs, ensuring a greater number of these cells reach and engraft in damaged tissue, thereby augmenting repair and regeneration processes [94].

Stimulation of Homing and Recruitment of Stem Cells

The process of stem cell homing and recruitment involves the directed migration and engraftment of stem cells to sites of injury. Recent evidence indicates that chemokine receptor CXCR4 signaling in endothelial progenitor cells is impaired in individuals with coronary artery disease [98]. This impairment leads to reduced neovascularization, highlighting the potential of targeting CXCR4 to improve outcomes in coronary artery disease [98]. Furthermore, the SDF-1/CXCR4 axis has emerged as a promising therapeutic target for ischemic heart disease [99]. Enhancing SDF-1/CXCR4 signaling through various molecular mechanisms, including gene transfer, may augment EPC migration and improve therapeutic efficacy [99].
The development of chemokine-coated scaffolds and the use of nanoparticles sensitive to reactive oxygen species for targeted delivery to injured tissues are innovative approaches being explored [100]. Such nanoparticles have shown promise in targeting CXCR12 in damaged cardiac tissue, potentially enhancing the homing and effectiveness of stem cell therapy [100]. The integration of Artificial Intelligence (AI) in healthcare has opened new avenues for advancing stem cell therapies. AI-assisted drug synthesis and delivery could potentially surpass traditional methods in efficiency and specificity. AI applications in stem cell therapy are poised to deepen our understanding of MSC mechanisms and address existing challenges, thereby enhancing the effectiveness of treatments [101].
Despite over two decades of research and significant advancements, cell-based therapy for cardiovascular diseases faces persistent challenges and uncertainties. Major hurdles include an insufficient number of engrafted stem/progenitor cells, low survival rates in damaged tissue, and the impaired reparative capacity of these cells in patients with cardiovascular diseases [102]. To achieve successful cardiovascular repair through cell-based therapy, these obstacles must be addressed. A safe, effective, and broadly applicable cell-based therapy for cardiovascular diseases necessitates further research and the optimization of stem-/progenitor-cell-based treatments.

5. Challenges and Needs in MSC-Based Cardiovascular Therapeutics

The multipotential nature of MSCs is particularly valuable for treating cardiovascular disorders due to their specific differentiation into cell types crucial for cardiovascular therapy [103,104,105].

5.1. Challenges

The immunomodulatory capabilities of MSCs are subject of research in treating immunodeficiency diseases, such as graft-versus-host disease and Crohn’s disease. However, the use of MSCs in conditions like multiple sclerosis remains experimental and should be approached with caution [106,107,108]. Despite the potential of MSC-based therapies, challenges have arisen, notably concerning the quality and delivery of MSCs, as well as a lack of established guidelines for culture protocols and biosafety [109,110]. Critical stages in MSC therapy involve the isolation and culture of cells in vitro followed by their in vivo delivery, presenting challenges like impaired homing ability, poor cell retention, and the risk of the overexpression of chemokines and cytokines in targeted areas [45,57,58].
The efficacy of MSCs is also influenced by factors such as the donor’s and recipient’s ages, medical histories, and genetic predispositions [111,112]. Obtaining enough healthy MSCs from individuals with conditions like diabetes or rheumatoid arthritis is particularly challenging as these conditions can affect the quality of MSCs [113]. Additionally, there have been concerns regarding the safety of MSC therapies, such as the development of tumors, which might be attributed to the inherent characteristics of MSCs rather than graft rejection [114].

5.2. Overcoming Limitations

To address these challenges, MSCs are now classified as advanced therapy medicinal products. Guidelines from the American Code of Federal Regulations and the Food and Drug Administration, along with GMPs, have been established to guide the culture method, isolation method, quality assurance method, and delivery protocol used and the overall safety of MSC-based therapies [115,116]. These guidelines aim to enhance the in vivo and in vitro efficacy and viability of MSCs. Recent studies, like the one conducted by Codinach et al. [117], have demonstrated the efficacy of bioprocess engineering in the separation, expansion, validation, and manufacture of bone marrow-derived MSCs for clinical use. Their research on 48 batches of iliac crest bone marrow samples for autologous transplantation highlights the importance of standardized processes in MSC therapy, including collection, isolation, trypsinization, and quality control, ensuring the safety, functionality, and potency of MSCs [117].
Adult-tissue-derived MSCs exhibit significant variability due to donor differences, impacting their proliferation, differentiation, and immunomodulatory capacities. This variability challenges the consistency and therapeutic efficacy of MSC-derived products like exosomes. In contrast, iPSC-derived MSCs, which originate from clonal lines, provide a more standardized source that diminishes variability and enhances the scalability and reproducibility of MSC production [118]. These cells are produced under stringent GMP conditions, ensuring rigorous quality control standards for safety, consistency, and efficacy [118]. iPSC-derived MSCs have demonstrated promising results in clinical trials for conditions such as refractory graft-versus-host disease (GVHD), and a Phase 1 trial highlights the potential of iPSC-derived MSCs to surmount challenges of heterogeneity in adult-tissue-derived MSCs, favoring their use in therapeutic applications that require high consistency and scalability [118].

6. Emerging Strategies in MSC-Based Tissue Engineering and Regeneration

6.1. Cell-Free Approaches: The Power of the Secretome

The MSC secretome, which encompasses a range of bioactive factors, including growth factors, cytokines, and chemokines, contributes to immunomodulation, homeostasis, tissue repair, and regeneration [119,120,121]. The secretome has gained attention in wound healing due to its direct involvement in cell proliferation, migration, and tissue repair [122,123,124]. The process of extracting the secretome from MSCs involves isolating and cultivating the cells in a suitable medium. After appropriate cultivation, the MSC-derived secretome is collected, followed by centrifugation and filtration to ensure purity [125]. Characterization of the secretome proteins is conducted using proteomic techniques such as the shotgun method, ELISA, or Western blotting [126].
Prior to clinical application, the secretome undergoes rigorous testing for efficacy, cell viability, proliferation, wound-healing capabilities [127,128,129,130], and cardiovascular diseases [57,131]. Cell viability assays are conducted using model cells like human epithelial stem cells, and the results are analyzed through fluorescence microscopy. Proliferation assays and colorimetric assessments are also performed to further validate the therapeutic potential of the secretome [127,128,129,130]. The efficacy of the secretome in wound healing is evaluated both in vitro and in vivo [132]. For in vitro studies, tissue defects are treated with varying concentrations of the secretome, and cell migration is assessed using fluorescence analysis [132]. In vivo studies involve applying the secretome to damaged skin in animal models, followed by a histological examination post regeneration [132]. Various delivery methods, including intravenous injection and direct injection into specific tissues, have been explored for secretome administration [133]. While these methods have shown positive effects in animal models, challenges remain, particularly in developing effective culture techniques and standardized protocols for isolation, culture, and distribution to ensure safety and minimize potential complications [133].
The secretome presents a potential therapeutic approach in cell-free treatments which offers advantages such as reductions in the risk of immunological rejection and tumorigenic potential. Its application in tissue regeneration is particularly promising due to its compatibility and ease of delivery, addressing some limitations of traditional stem cell-based therapies. However, further research is needed to establish standardized guidelines and validate its efficacy as a biological element in tissue regeneration [134].

6.2. Scaffold-Based Therapeutics: Enhancing Stem Cell Potential

The development of scaffold-based therapeutics represents a significant advance in tissue engineering, offering promising strategies for repairing damaged cardiac tissues and restoring their structure and function [135,136]. These scaffolds, derived from both synthetic and natural biomaterials, are crucial in regenerating diverse tissues including bone, cartilage, ligaments, neural tissues, skin, skeletal muscle, and blood vessels [137,138]. They typically employ biodegradable natural polymers such as collagen, fibrin, gelatin, hyaluronic acid, and poly(lactic-co-glycolic) acid [139,140,141]. The primary objective of biomaterial scaffold-based techniques is to support tissue repair and regeneration. This is achieved by incorporating therapeutic cells into a porous 3D scaffold enriched with growth factors or signaling molecules, creating a conducive environment for cell infiltration, proliferation, and differentiation [142].

Biomaterials

Cells, sourced from allogeneic, syngeneic, xenogeneic, or autologous origins, are initially isolated from biopsies. These cells are then cultured in bioreactors, cell culture systems, or in vitro settings for controlled growth and expansion. Expanded cells are seeded onto scaffolds infused with growth factors and nutrients, leading to the formation of new tissues. These tissues, which are integrated within the scaffold, are designed to replace damaged tissues in patients [143,144]. In tissue engineering, natural polymers like chitosan, collagen, alginate, silk fibroin, hyaluronan, and gelatin are extensively used for cartilage regeneration. These materials facilitate the growth of new chondral tissue at defect sites due to their biocompatibility, biodegradability, minimal immune response, and effective cell interaction [145]. Natural polysaccharides, which are preferred over synthetic polymers, potentially minimize immune reactions and promote cartilage growth through specific biological pathways [146,147].
The success of biomaterials in tissue engineering depends on their structural and functional compatibility with target tissues and cell types. This necessity stems from the distinct physical and chemical properties inherent to different tissues and cells [148,149]. Factors such as the hydrophobicity, chemical composition, and charge of biomaterial surfaces significantly influence their biological activity [150,151]. Biomaterials interact intricately with the 3D microenvironments of targeted tissues [152,153,154]. For tissues subjected to mechanical stress or weight bearing, like bones or teeth, biomaterials with robust mechanical properties are essential [155]. Conversely, soft tissues like skin and internal organs require biomaterials with characteristics such as porosity, softness, and high viscosity [156]. This complex interplay between biomaterial properties and tissue-specific needs underscores the critical role of biomaterials in orchestrating successful tissue regeneration processes.

6.3. Three-Dimensional Ex Vivo Propagation and Pre-Treatment

The transition from traditional 2D cultures to 3D environments represents a significant advancement in MSC propagation. Three-dimensional cultures better replicate the natural environment of MSCs, enhancing their differentiation toward cardiac [157,158,159] and skeleton-related tissues [160]. Despite extensive research on 3D MSC culture, challenges persist, including replicating the natural ECM milieu, eliminating residual hazardous solvents, achieving uniform cell distribution, and maintaining cell viability [160,161]. Scaffold-based culture systems promote MSC–matrix interaction, while scaffold-free cultures rely on the cells themselves to establish a suitable microenvironment [162]. The growing body of research on 3D MSC culture suggests its therapeutic potential, but it requires more meticulous attention and critical analysis compared to 2D culture. The relative scarcity of standardized protocols for 3D culture, in contrast to the well-established 2D culture methods, presents an additional challenge [163,164].
Optimizing MSC-based treatments involves strategies that do not necessarily rely on increasing the dosage or frequency of administration. The primary goal is to achieve optimal results without adverse systemic effects. These optimization strategies are divided into genetic and non-genetic modifications [165,166]; the latter are also known as MSC pre-treatments. MSCs can be pre-activated to enhance their functional potential by simulating either their physiological or pathological microenvironment [166].

6.3.1. Physiological Microenvironment Simulation Pre-Activation

Typically, in vitro MSC cultures are exposed to an oxygen tension of about 21%. Under hypoxic conditions, MSCs, particularly adipose-derived MSCs, have been shown to maintain their undifferentiated state and express higher levels of multipotent stem cell markers (Oct4, Sox2, and Nanog) without significant morphological or surface marker changes [167,168,169].

6.3.2. Pathological Microenvironment Simulation Pre-Activation

Mimicking the inflammatory microenvironment using cytokines like TNF-α, INF-γ, IL-1β, IL-17A, and IL-25 can enhance the immunomodulatory function of MSCs [170]. Pre-activation with growth factors such as basic fibroblast growth factor (bFGF) and chemokine ligands like SDF-1 has shown promise in maintaining stem cell properties and aiding tissue regeneration [171,172]. Pre-activation with bioactive compounds, either natural or synthetic, in MSC pre-activation is gaining interest [173,174,175,176,177]. These compounds can enhance MSC survival, immunomodulation, and cardiac repair. Notable examples include trimetazidine, tadalafil, atorvastatin for MSC modulation, and iron chelator deferoxamine and treprostinil for enhancing immunomodulatory functions [173,174,175,176,177]. The process of optimizing MSCs for enhanced therapeutic outcomes in cardiovascular treatments has been illustrated in Figure 1.

6.4. Genetic Modifications: Tailoring MSCs for Targeted Repair

Genetic modifications of MSCs for targeted repair are performed using both viral vectors and non-viral methods, enhancing specificity and efficacy in regenerative treatments. The ease of isolation and expansion of MSCs in vitro makes them suitable candidates for transfection and targeted recruitment at inflammation sites [178,179,180].

6.4.1. Viral Vector-Mediated Genetic Modification

Retroviruses, using RNA as genetic material, integrate their genome into the host cell DNA [181]. This process involves the fusion of the viral lipid envelope with the host cell membrane, followed by the reverse transcription of viral RNA into DNA and its integration into the host genome [181]. Retroviruses have been used to modify MSCs efficiently, allowing for the production of large quantities of modified cells [182]. For example, a study successfully transferred genes like Foxa1 and Hnf4a into BMSCs using retroviral methods [183]. Lentiviruses, which are capable of infecting non-dividing cells, can carry 8–9 kb of genetic material. Unlike retroviruses, lentiviruses integrate more slowly and less disruptively into the host cell genome. Their high efficiency and stability rates make them preferred tools for MSC modification [184,185,186]. The transplantation of MSCs modified with the TNFR gene via a recombinant adeno-associated virus improved left ventricular function after myocardial infarction by reducing inflammation and apoptosis [187].

6.4.2. Non-Viral Methods of Genetic Modification

Electroporation, sonotransfection, and nucleofection are physical methods for introducing genetic material into MSCs [188,189]. Electroporation has shown high efficiency, with the optimal conditions for human MSCs being a pulse magnitude of 1500 V for 20 ms, resulting in 78% viability and 50% efficiency [188]. Nucleofection, a specialized form of electroporation, facilitates the direct transfer of plasmid DNA into the cell nucleus [188]. Chemical methods include the use of synthetic vectors like cationic lipids, polymers, nanoparticles, and cell-penetrating peptides for gene transfer [190]. Chemical methods offer the advantage of large-scale manufacturing and reduced side effects compared to viral vectors [190]. Genetically engineered MSCs overexpressing stromal cell-derived factor-1α [191] or hypoxia-inducible factor 1-α [192], Insulin-like growth factor 1, and hepatocyte growth factor [193] have shown potential in cardiovascular recovery. MSCs modified to express specific factors demonstrated enhanced efficacy in reducing inflammatory responses and fibrosis in a model of Chagas disease [194]. Additionally, MSCs overexpressing microRNA, such as miR-126, have been shown to increase pro-angiogenic factors, thus improving the treatment efficacy in infarcted hearts [195].

6.5. Mechanobiologically Mediated Differentiation of Stem Cells

Shear stress from fluid flow crucially directs MSCs and EPCs toward an endothelial phenotype, elevating endothelial markers like CD31, vWF, and vascular endothelial cadherin due to mechanotransduction pathways that translate mechanical signals into biochemical cues [54]. The application of laminar shear stress to MSCs significantly increased cardiomyocyte differentiation [196] and endothelial markers, promoting endothelium-like functions such as tubule formation and low density lipoprotein uptake [197]. Additionally, mechanical strains, such as cyclic stretch, also guide stem cells toward endothelial lineages, particularly in EPCs, enhancing their migration and tubulogenesis, which mirrors the dynamics within blood vessels [198]. This integration of mechanobiological stimuli is essential for the effective differentiation of MSCs and EPCs into functional ECs, underscoring the significance of physical cues in vascular tissue engineering and the potential of these cells in regenerative medicine for cardiovascular disorders [27,54]. The optimization of cell culture conditions, microenvironmental factors, and the application of mechanical and electrical stimulations to enhance myogenic differentiation has been emphasized [199,200].
A comparative overview of emerging MSC-based therapeutic strategies in tissue engineering and regeneration is summarized in Table 1.

6.6. GMPs in Stem Cell-Based Therapeutics

MSCs must comply with current GMP standards, ensuring their readiness for further manufacturing steps. GMPs, enforced by agencies like the US Food and Drug Administration (FDA), set forth rigorous rules to guarantee that products are consistently produced and controlled according to quality standards [201]. Adherence to GMP standards is a fundamental requirement for obtaining marketing authorization for stem cell-based therapeutics [202]. This compliance not only accelerates the development of regenerative medicine products but also potentially reduces associated costs [203]. Global regulations and quality assurance guidelines provide detailed procedures for manufacturing, testing, and quality assurance. Adhering to these standards is crucial for producing raw materials that meet stringent criteria for purity, potency, consistency, and stability [203]. Compliance with these guidelines is vital for maintaining the safety and high quality of raw materials used in stem cell-based therapies, thereby upholding the integrity and efficacy of the final therapeutic products [203].

7. Conclusions

Emerging strategies in MSC-based cardiovascular therapeutics focus on sophisticated, targeted approaches that move beyond traditional cell transplantation to more refined treatments. Harnessing the MSC secretome, developing biocompatible scaffolds, employing genetic modifications, and utilizing extracellular vesicles and three-dimensional tissue modeling are key advances. These strategies aim for higher efficacy, reduced side effects, and personalized patient care. The integration of tissue engineering, nanotechnology, and precision medicine is set to enhance these therapies further, ensuring their safety, quality, and reproducibility through standardized protocols and GMP-compliant procedures. This evolving landscape promises transformative impacts on cardiovascular medicine, driven by in-depth MSC biology research and technological innovations.

Author Contributions

Conceptualization, N.G.; writing—original draft preparation, R.K., N.M. and T.T.; writing—review and editing, N.G., M.K. and S.V.; funding acquisition, N.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by funding from the American Heart Association, grant #940594/Narasimman Gurusamy/2022.

Acknowledgments

The authors are thankful for the support of the Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update from the GBD 2019 Study. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef] [PubMed]
  2. Hashimoto, H.; Olson, E.N.; Bassel-Duby, R. Therapeutic approaches for cardiac regeneration and repair. Nat. Rev. Cardiol. 2018, 15, 585–600. [Google Scholar] [CrossRef] [PubMed]
  3. Zakrzewski, W.; Dobrzyński, M.; Szymonowicz, M.; Rybak, Z. Stem cells: Past, present, and future. Stem Cell Res. Ther. 2019, 10, 68. [Google Scholar] [CrossRef] [PubMed]
  4. Han, Y.; Li, X.; Zhang, Y.; Han, Y.; Chang, F.; Ding, J. Mesenchymal Stem Cells for Regenerative Medicine. Cells 2019, 8, 886. [Google Scholar] [CrossRef] [PubMed]
  5. Aly, R.M. Current state of stem cell-based therapies: An overview. Stem Cell Investig. 2020, 7, 8. [Google Scholar] [CrossRef]
  6. Thomas, E.D.; Lochte, H.L., Jr.; Lu, W.C.; Ferrebee, J.W. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N. Engl. J. Med. 1957, 257, 491–496. [Google Scholar] [CrossRef] [PubMed]
  7. Wollert, K.C.; Meyer, G.P.; Lotz, J.; Ringes-Lichtenberg, S.; Lippolt, P.; Breidenbach, C.; Fichtner, S.; Korte, T.; Hornig, B.; Messinger, D.; et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: The BOOST randomised controlled clinical trial. Lancet 2004, 364, 141–148. [Google Scholar] [CrossRef]
  8. Makkar, R.R.; Smith, R.R.; Cheng, K.; Malliaras, K.; Thomson, L.E.; Berman, D.; Czer, L.S.; Marbán, L.; Mendizabal, A.; Johnston, P.V.; et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): A prospective, randomised phase 1 trial. Lancet 2012, 379, 895–904. [Google Scholar] [CrossRef] [PubMed]
  9. Schächinger, V.; Erbs, S.; Elsässer, A.; Haberbosch, W.; Hambrecht, R.; Hölschermann, H.; Yu, J.; Corti, R.; Mathey, D.G.; Hamm, C.W.; et al. Improved clinical outcome after intracoronary administration of bone-marrow-derived progenitor cells in acute myocardial infarction: Final 1-year results of the REPAIR-AMI trial. Eur. Heart J. 2006, 27, 2775–2783. [Google Scholar] [CrossRef]
  10. Mishra, R.; Vijayan, K.; Colletti, E.J.; Harrington, D.A.; Matthiesen, T.S.; Simpson, D.; Goh, S.K.; Walker, B.L.; Almeida-Porada, G.; Wang, D.; et al. Characterization and functionality of cardiac progenitor cells in congenital heart patients. Circulation 2011, 123, 364–373. [Google Scholar] [CrossRef]
  11. Sugarman, J. Ethics and stem cell therapeutics for cardiovascular disease. Prog. Cardiovasc. Dis. 2007, 50, 1–6. [Google Scholar] [CrossRef]
  12. Liu, L.; Wu, Q.; Wang, Z.; Niu, B.; Jiao, Y.; An, H. APE1 promotes embryonic stem cell proliferation and teratoma formation by regulating GDNF/GFRα1 axis. Reprod. Biol. 2023, 23, 100792. [Google Scholar] [CrossRef] [PubMed]
  13. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed]
  14. 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] [PubMed]
  15. Kussauer, S.; David, R.; Lemcke, H. hiPSCs Derived Cardiac Cells for Drug and Toxicity Screening and Disease Modeling: What Micro- Electrode-Array Analyses Can Tell Us. Cells 2019, 8, 1331. [Google Scholar] [CrossRef] [PubMed]
  16. Bonilauri, B.; Shin, H.S.; Htet, M.; Yan, C.D.; Witteles, R.M.; Sallam, K.; Wu, J.C. Generation of two induced pluripotent stem cell lines from patients with cardiac amyloidosis carrying heterozygous transthyretin (TTR) mutation. Stem Cell Res. 2023, 72, 103215. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, J.; Wilson, G.F.; Soerens, A.G.; Koonce, C.H.; Yu, J.; Palecek, S.P.; Thomson, J.A.; Kamp, T.J. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ. Res. 2009, 104, e30–e41. [Google Scholar] [CrossRef] [PubMed]
  18. Gurusamy, N.; Rajasingh, S.; Sigamani, V.; Rajasingh, R.; Isai, D.G.; Czirok, A.; Bittel, D.; Rajasingh, J. Noonan syndrome patient-specific induced cardiomyocyte model carrying SOS1 gene variant c.1654A>G. Exp. Cell Res. 2021, 400, 112508. [Google Scholar] [CrossRef] [PubMed]
  19. Song, M.; Choi, D.B.; Im, J.S.; Song, Y.N.; Kim, J.H.; Lee, H.; An, J.; Kim, A.; Choi, H.; Kim, J.C.; et al. Modeling acute myocardial infarction and cardiac fibrosis using human induced pluripotent stem cell-derived multi-cellular heart organoids. Cell Death Dis. 2024, 15, 308. [Google Scholar] [CrossRef]
  20. Liang, S.; Su, Y.; Yao, R. 3D Bioprinting of Induced Pluripotent Stem Cells and Disease Modeling. Handb. Exp. Pharmacol. 2023, 281, 29–56. [Google Scholar] [CrossRef]
  21. Yu, D.; Wang, X.; Ye, L. Cardiac Tissue Engineering for the Treatment of Myocardial Infarction. J. Cardiovasc. Dev. Dis. 2021, 8, 153. [Google Scholar] [CrossRef]
  22. Riegler, J.; Tiburcy, M.; Ebert, A.; Tzatzalos, E.; Raaz, U.; Abilez, O.J.; Shen, Q.; Kooreman, N.G.; Neofytou, E.; Chen, V.C.; et al. Human Engineered Heart Muscles Engraft and Survive Long Term in a Rodent Myocardial Infarction Model. Circ. Res. 2015, 117, 720–730. [Google Scholar] [CrossRef]
  23. Wang, D.; Mou, H.; Li, S.; Li, Y.; Hough, S.; Tran, K.; Li, J.; Yin, H.; Anderson, D.G.; Sontheimer, E.J.; et al. Adenovirus-Mediated Somatic Genome Editing of Pten by CRISPR/Cas9 in Mouse Liver in Spite of Cas9-Specific Immune Responses. Hum. Gene Ther. 2015, 26, 432–442. [Google Scholar] [CrossRef]
  24. Lemcke, H.; Voronina, N.; Steinhoff, G.; David, R. Recent Progress in Stem Cell Modification for Cardiac Regeneration. Stem Cells Int. 2018, 2018, 1909346. [Google Scholar] [CrossRef]
  25. Bian, W.; Chen, W.; Nguyen, T.; Zhou, Y.; Zhang, J. miR-199a Overexpression Enhances the Potency of Human Induced-Pluripotent Stem-Cell-Derived Cardiomyocytes for Myocardial Repair. Front. Pharmacol. 2021, 12, 673621. [Google Scholar] [CrossRef] [PubMed]
  26. Smith, R.R.; Barile, L.; Cho, H.C.; Leppo, M.K.; Hare, J.M.; Messina, E.; Giacomello, A.; Abraham, M.R.; Marbán, E. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation 2007, 115, 896–908. [Google Scholar] [CrossRef] [PubMed]
  27. Qiu, Y.; Zhang, C.; Zhang, G.; Tao, J. Endothelial progenitor cells in cardiovascular diseases. Aging Med. 2018, 1, 204–208. [Google Scholar] [CrossRef] [PubMed]
  28. Salybekov, A.A.; Kobayashi, S.; Asahara, T. Characterization of Endothelial Progenitor Cell: Past, Present, and Future. Int. J. Mol. Sci. 2022, 23, 7697. [Google Scholar] [CrossRef]
  29. Park, K.S.; Kang, S.N.; Kim, D.H.; Kim, H.B.; Im, K.S.; Park, W.; Hong, Y.J.; Han, D.K.; Joung, Y.K. Late endothelial progenitor cell-capture stents with CD146 antibody and nanostructure reduce in-stent restenosis and thrombosis. Acta Biomater. 2020, 111, 91–101. [Google Scholar] [CrossRef]
  30. Mitsiou, G.; Tokmakidis, S.P.; Dinas, P.C.; Smilios, I.; Nanas, S. Endothelial progenitor cell mobilization based on exercise volume in patients with cardiovascular disease and healthy individuals: A systematic review and meta-analysis. Eur. Heart J. Open 2022, 2, oeac078. [Google Scholar] [CrossRef]
  31. Guo, Y.; Yu, Y.; Hu, S.; Chen, Y.; Shen, Z. The therapeutic potential of mesenchymal stem cells for cardiovascular diseases. Cell Death Dis. 2020, 11, 349. [Google Scholar] [CrossRef]
  32. Wang, Y.; Qi, Z.; Yan, Z.; Ji, N.; Yang, X.; Gao, D.; Hu, L.; Lv, H.; Zhang, J.; Li, M. Mesenchymal Stem Cell Immunomodulation: A Novel Intervention Mechanism in Cardiovascular Disease. Front. Cell Dev. Biol. 2021, 9, 742088. [Google Scholar] [CrossRef]
  33. Yan, W.; Xia, Y.; Zhao, H.; Xu, X.; Ma, X.; Tao, L. Stem cell-based therapy in cardiac repair after myocardial infarction: Promise, challenges, and future directions. J. Mol. Cell. Cardiol. 2024, 188, 1–14. [Google Scholar] [CrossRef]
  34. Mastrolia, I.; Foppiani, E.M.; Murgia, A.; Candini, O.; Samarelli, A.V.; Grisendi, G.; Veronesi, E.; Horwitz, E.M.; Dominici, M. Challenges in Clinical Development of Mesenchymal Stromal/Stem Cells: Concise Review. Stem Cells Transl. Med. 2019, 8, 1135–1148. [Google Scholar] [CrossRef]
  35. Angoulvant, D.; Ivanes, F.; Ferrera, R.; Matthews, P.G.; Nataf, S.; Ovize, M. Mesenchymal stem cell conditioned media attenuates in vitro and ex vivo myocardial reperfusion injury. J. Heart Lung Transplant. 2011, 30, 95–102. [Google Scholar] [CrossRef]
  36. Rehman, A.; Nigam, A.; Laino, L.; Russo, D.; Todisco, C.; Esposito, G.; Svolacchia, F.; Giuzio, F.; Desiderio, V.; Ferraro, G. Mesenchymal Stem Cells in Soft Tissue Regenerative Medicine: A Comprehensive Review. Medicina 2023, 59, 1449. [Google Scholar] [CrossRef] [PubMed]
  37. Siu, C.W.; Liao, S.Y.; Liu, Y.; Lian, Q.; Tse, H.F. Stem cells for myocardial repair. Thromb. Haemost. 2010, 104, 6–12. [Google Scholar] [CrossRef] [PubMed]
  38. Schulman, I.H.; Balkan, W.; Hare, J.M. Mesenchymal Stem Cell Therapy for Aging Frailty. Front. Nutr. 2018, 5, 108. [Google Scholar] [CrossRef] [PubMed]
  39. Hoang, D.M.; Pham, P.T.; Bach, T.Q.; Ngo, A.T.L.; Nguyen, Q.T.; Phan, T.T.K.; Nguyen, G.H.; Le, P.T.T.; Hoang, V.T.; Forsyth, N.R.; et al. Stem cell-based therapy for human diseases. Signal Transduct. Target. Ther. 2022, 7, 272. [Google Scholar] [CrossRef]
  40. Mathiasen, A.B.; Qayyum, A.A.; Jørgensen, E.; Helqvist, S.; Kofoed, K.F.; Haack-Sørensen, M.; Ekblond, A.; Kastrup, J. Bone marrow-derived mesenchymal stromal cell treatment in patients with ischaemic heart failure: Final 4-year follow-up of the MSC-HF trial. Eur. J. Heart Fail. 2020, 22, 884–892. [Google Scholar] [CrossRef]
  41. Bartunek, J.; Behfar, A.; Dolatabadi, D.; Vanderheyden, M.; Ostojic, M.; Dens, J.; El Nakadi, B.; Banovic, M.; Beleslin, B.; Vrolix, M.; et al. Cardiopoietic stem cell therapy in heart failure: The C-CURE (Cardiopoietic stem Cell therapy in heart failURE) multicenter randomized trial with lineage-specified biologics. J. Am. Coll. Cardiol. 2013, 61, 2329–2338. [Google Scholar] [CrossRef] [PubMed]
  42. Bartunek, J.; Terzic, A.; Davison, B.A.; Filippatos, G.S.; Radovanovic, S.; Beleslin, B.; Merkely, B.; Musialek, P.; Wojakowski, W.; Andreka, P.; et al. Cardiopoietic cell therapy for advanced ischaemic heart failure: Results at 39 weeks of the prospective, randomized, double blind, sham-controlled CHART-1 clinical trial. Eur. Heart J. 2017, 38, 648–660. [Google Scholar] [CrossRef] [PubMed]
  43. Borow, K.M.; Yaroshinsky, A.; Greenberg, B.; Perin, E.C. Phase 3 DREAM-HF Trial of Mesenchymal Precursor Cells in Chronic Heart Failure. Circ. Res. 2019, 125, 265–281. [Google Scholar] [CrossRef] [PubMed]
  44. Ma, J.; Chen, L.; Zhu, X.; Li, Q.; Hu, L.; Li, H. Mesenchymal stem cell-derived exosomal miR-21a-5p promotes M2 macrophage polarization and reduces macrophage infiltration to attenuate atherosclerosis. Acta Biochim. Biophys. Sin. 2021, 53, 1227–1236. [Google Scholar] [CrossRef] [PubMed]
  45. Pan, Y.; Wu, W.; Jiang, X.; Liu, Y. Mesenchymal stem cell-derived exosomes in cardiovascular and cerebrovascular diseases: From mechanisms to therapy. Biomed. Pharmacother. 2023, 163, 114817. [Google Scholar] [CrossRef] [PubMed]
  46. Sahoo, S.; Losordo, D.W. Exosomes and cardiac repair after myocardial infarction. Circ. Res. 2014, 114, 333–344. [Google Scholar] [CrossRef] [PubMed]
  47. Gu, J.; You, J.; Liang, H.; Zhan, J.; Gu, X.; Zhu, Y. Engineered bone marrow mesenchymal stem cell-derived exosomes loaded with miR302 through the cardiomyocyte specific peptide can reduce myocardial ischemia and reperfusion (I/R) injury. J. Transl. Med. 2024, 22, 168. [Google Scholar] [CrossRef] [PubMed]
  48. Dang, X.T.T.; Kavishka, J.M.; Zhang, D.X.; Pirisinu, M.; Le, M.T.N. Extracellular Vesicles as an Efficient and Versatile System for Drug Delivery. Cells 2020, 9, 2191. [Google Scholar] [CrossRef] [PubMed]
  49. Shen, X.; Pan, B.; Zhou, H.; Liu, L.; Lv, T.; Zhu, J.; Huang, X.; Tian, J. Differentiation of mesenchymal stem cells into cardiomyocytes is regulated by miRNA-1-2 via WNT signaling pathway. J. Biomed. Sci. 2017, 24, 29. [Google Scholar] [CrossRef]
  50. Xu, H.; Yang, Y.J.; Qian, H.Y.; Tang, Y.D.; Wang, H.; Zhang, Q. Rosuvastatin treatment activates JAK-STAT pathway and increases efficacy of allogeneic mesenchymal stem cell transplantation in infarcted hearts. Circ. J. 2011, 75, 1476–1485. [Google Scholar] [CrossRef]
  51. He, X.; Li, L.; Tang, M.; Zeng, Y.; Li, H.; Yu, X. Biomimetic electrical stimulation induces rat bone marrow mesenchymal stem cells to differentiate into cardiomyocyte-like cells via TGF-beta 1 in vitro. Prog. Biophys. Mol. Biol. 2019, 148, 47–53. [Google Scholar] [CrossRef] [PubMed]
  52. Quevedo, H.C.; Hatzistergos, K.E.; Oskouei, B.N.; Feigenbaum, G.S.; Rodriguez, J.E.; Valdes, D.; Pattany, P.M.; Zambrano, J.P.; Hu, Q.; McNiece, I.; et al. Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc. Natl. Acad. Sci. USA 2009, 106, 14022–14027. [Google Scholar] [CrossRef] [PubMed]
  53. Khaki, M.; Salmanian, A.H.; Abtahi, H.; Ganji, A.; Mosayebi, G. Mesenchymal Stem Cells Differentiate to Endothelial Cells Using Recombinant Vascular Endothelial Growth Factor—A. Rep. Biochem. Mol. Biol. 2018, 6, 144–150. [Google Scholar] [PubMed]
  54. Huang, Y.; Qian, J.Y.; Cheng, H.; Li, X.M. Effects of shear stress on differentiation of stem cells into endothelial cells. World J. Stem Cells 2021, 13, 894–913. [Google Scholar] [CrossRef] [PubMed]
  55. Harris, L.J.; Abdollahi, H.; Zhang, P.; McIlhenny, S.; Tulenko, T.N.; DiMuzio, P.J. Differentiation of adult stem cells into smooth muscle for vascular tissue engineering. J. Surg. Res. 2011, 168, 306–314. [Google Scholar] [CrossRef] [PubMed]
  56. Narita, Y.; Yamawaki, A.; Kagami, H.; Ueda, M.; Ueda, Y. Effects of transforming growth factor-beta 1 and ascorbic acid on differentiation of human bone-marrow-derived mesenchymal stem cells into smooth muscle cell lineage. Cell Tissue Res. 2008, 333, 449–459. [Google Scholar] [CrossRef] [PubMed]
  57. Ranganath, S.H.; Levy, O.; Inamdar, M.S.; Karp, J.M. Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell 2012, 10, 244–258. [Google Scholar] [CrossRef] [PubMed]
  58. Safina, I.; Embree, M.C. Biomaterials for recruiting and activating endogenous stem cells in situ tissue regeneration. Acta Biomater. 2022, 143, 26–38. [Google Scholar] [CrossRef] [PubMed]
  59. Allemailem, K.S.; Almatroodi, S.A.; Almatroudi, A.; Alrumaihi, F.; Al Abdulmonem, W.; Al-Megrin, W.A.I.; Aljamaan, A.N.; Rahmani, A.H.; Khan, A.A. Recent Advances in Genome-Editing Technology with CRISPR/Cas9 Variants and Stimuli-Responsive Targeting Approaches within Tumor Cells: A Future Perspective of Cancer Management. Int. J. Mol. Sci. 2023, 24, 7052. [Google Scholar] [CrossRef]
  60. Shahabipour, F.; Oskuee, R.K.; Shokrgozar, M.A.; Naderi-Meshkin, H.; Goshayeshi, L.; Bonakdar, S. CRISPR/Cas9 mediated GFP-human dentin matrix protein 1 (DMP1) promoter knock-in at the ROSA26 locus in mesenchymal stem cell for monitoring osteoblast differentiation. J. Gene Med. 2020, 22, e3288. [Google Scholar] [CrossRef]
  61. Yuan, X.; Li, D.; Chen, X.; Han, C.; Xu, L.; Huang, T.; Dong, Z.; Zhang, M. Extracellular vesicles from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect against renal ischemia/reperfusion injury via delivering specificity protein (SP1) and transcriptional activating of sphingosine kinase 1 and inhibiting necroptosis. Cell Death Dis. 2017, 8, 3200. [Google Scholar] [CrossRef] [PubMed]
  62. Meshitsuka, S.; Ninomiya, R.; Nagamura-Inoue, T.; Okada, T.; Futami, M.; Tojo, A. CRISPR/Cas9 and AAV mediated insertion of β2 microglobulin-HLA-G fusion gene protects mesenchymal stromal cells from allogeneic rejection and potentiates the use for off-the-shelf cell therapy. Regen. Ther. 2022, 21, 442–452. [Google Scholar] [CrossRef] [PubMed]
  63. Han, A.R.; Shin, H.R.; Kwon, J.; Lee, S.B.; Lee, S.E.; Kim, E.Y.; Kweon, J.; Chang, E.J.; Kim, Y.; Kim, S.W. Highly efficient genome editing via CRISPR-Cas9 ribonucleoprotein (RNP) delivery in mesenchymal stem cells. BMB Rep. 2024, 57, 60–65. [Google Scholar] [CrossRef] [PubMed]
  64. Song, N.; Scholtemeijer, M.; Shah, K. Mesenchymal Stem Cell Immunomodulation: Mechanisms and Therapeutic Potential. Trends Pharmacol. Sci. 2020, 41, 653–664. [Google Scholar] [CrossRef] [PubMed]
  65. Jiang, W.; Xu, J. Immune modulation by mesenchymal stem cells. Cell Prolif. 2020, 53, e12712. [Google Scholar] [CrossRef] [PubMed]
  66. Huang, Y.; Wu, Q.; Tam, P.K.H. Immunomodulatory Mechanisms of Mesenchymal Stem Cells and Their Potential Clinical Applications. Int. J. Mol. Sci. 2022, 23, 10023. [Google Scholar] [CrossRef] [PubMed]
  67. Prockop, D.J.; Oh, J.Y. Mesenchymal stem/stromal cells (MSCs): Role as guardians of inflammation. Mol. Ther. 2012, 20, 14–20. [Google Scholar] [CrossRef] [PubMed]
  68. Bright, J.J.; Kerr, L.D.; Sriram, S. TGF-beta inhibits IL-2-induced tyrosine phosphorylation and activation of Jak-1 and Stat 5 in T lymphocytes. J. Immunol. 1997, 159, 175–183. [Google Scholar] [CrossRef] [PubMed]
  69. Fan, Y.; Herr, F.; Vernochet, A.; Mennesson, B.; Oberlin, E.; Durrbach, A. Human Fetal Liver Mesenchymal Stem Cell-Derived Exosomes Impair Natural Killer Cell Function. Stem Cells Dev. 2019, 28, 44–55. [Google Scholar] [CrossRef]
  70. Shi, Y.; Su, J.; Roberts, A.I.; Shou, P.; Rabson, A.B.; Ren, G. How mesenchymal stem cells interact with tissue immune responses. Trends Immunol. 2012, 33, 136–143. [Google Scholar] [CrossRef]
  71. Han, Y.; Yang, J.; Fang, J.; Zhou, Y.; Candi, E.; Wang, J.; Hua, D.; Shao, C.; Shi, Y. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduct. Target. Ther. 2022, 7, 92. [Google Scholar] [CrossRef] [PubMed]
  72. Ahangar, P.; Mills, S.J.; Cowin, A.J. Mesenchymal Stem Cell Secretome as an Emerging Cell-Free Alternative for Improving Wound Repair. Int. J. Mol. Sci. 2020, 21, 7038. [Google Scholar] [CrossRef]
  73. Harrell, C.R.; Fellabaum, C.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Molecular Mechanisms Responsible for Therapeutic Potential of Mesenchymal Stem Cell-Derived Secretome. Cells 2019, 8, 467. [Google Scholar] [CrossRef] [PubMed]
  74. Varderidou-Minasian, S.; Lorenowicz, M.J. Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: Challenges and opportunities. Theranostics 2020, 10, 5979–5997. [Google Scholar] [CrossRef] [PubMed]
  75. Sahoo, S.; Klychko, E.; Thorne, T.; Misener, S.; Schultz, K.M.; Millay, M.; Ito, A.; Liu, T.; Kamide, C.; Agrawal, H.; et al. Exosomes from human CD34+ stem cells mediate their proangiogenic paracrine activity. Circ. Res. 2011, 109, 724–728. [Google Scholar] [CrossRef]
  76. Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef] [PubMed]
  77. Baglio, S.R.; Pegtel, D.M.; Baldini, N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front. Physiol. 2012, 3, 359. [Google Scholar] [CrossRef] [PubMed]
  78. Zheng, H.; Liang, X.; Liu, B.; Huang, X.; Shen, Y.; Lin, F.; Chen, J.; Gao, X.; He, H.; Li, W.; et al. Exosomal miR-9-5p derived from iPSC-MSCs ameliorates doxorubicin-induced cardiomyopathy by inhibiting cardiomyocyte senescence. J. Nanobiotechnol. 2024, 22, 195. [Google Scholar] [CrossRef]
  79. Liu, B.; Wei, Y.; He, J.; Feng, B.; Chen, Y.; Guo, R.; Griffin, M.D.; Hynes, S.O.; Shen, S.; Liu, Y.; et al. Human umbilical cord-derived mesenchymal stromal cells improve myocardial fibrosis and restore miRNA-133a expression in diabetic cardiomyopathy. Stem Cell Res. Ther. 2024, 15, 120. [Google Scholar] [CrossRef]
  80. Zheng, R.; Wu, X.; Li, S.; Chen, X.; Yan, D.; He, J. Mechanism Exploration on the Immunoregulation of Allogeneic Heart Transplantation Rejection in Rats with Exosome miRNA and Proteins from Overexpressed IDO1 BMSCs. Cell Transplant. 2024, 33, 9636897241245796. [Google Scholar] [CrossRef]
  81. Zhu, W.; Du, W.; Duan, R.; Liu, Y.; Zong, B.; Jin, X.; Dong, Z.; Wang, H.; Shahab, S.; Wang, H.; et al. miR-873-5p Suppression Reinvigorates Aging Mesenchymal Stem Cells and Improves Cardiac Repair after Myocardial Infarction. ACS Pharmacol. Transl. Sci. 2024, 7, 743–756. [Google Scholar] [CrossRef] [PubMed]
  82. Vignais, M.L.; Levoux, J.; Sicard, P.; Khattar, K.; Lozza, C.; Gervais, M.; Mezhoud, S.; Nakhle, J.; Relaix, F.; Agbulut, O.; et al. Transfer of Cardiac Mitochondria Improves the Therapeutic Efficacy of Mesenchymal Stem Cells in a Preclinical Model of Ischemic Heart Disease. Cells 2023, 12, 582. [Google Scholar] [CrossRef] [PubMed]
  83. 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.; et al. Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circ. Res. 2010, 107, 913–922. [Google Scholar] [CrossRef]
  84. Tao, H.; Han, Z.; Han, Z.C.; Li, Z. Proangiogenic Features of Mesenchymal Stem Cells and Their Therapeutic Applications. Stem Cells Int. 2016, 2016, 1314709. [Google Scholar] [CrossRef] [PubMed]
  85. Gnecchi, M.; He, H.; Liang, O.D.; Melo, L.G.; Morello, F.; Mu, H.; Noiseux, N.; Zhang, L.; Pratt, R.E.; Ingwall, J.S.; et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat. Med. 2005, 11, 367–368. [Google Scholar] [CrossRef] [PubMed]
  86. Kinnaird, T.; Stabile, E.; Burnett, M.S.; Shou, M.; Lee, C.W.; Barr, S.; Fuchs, S.; Epstein, S.E. Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation 2004, 109, 1543–1549. [Google Scholar] [CrossRef]
  87. Kong, P.; Christia, P.; Frangogiannis, N.G. The pathogenesis of cardiac fibrosis. Cell Mol. Life Sci. 2014, 71, 549–574. [Google Scholar] [CrossRef]
  88. Mias, C.; Lairez, O.; Trouche, E.; Roncalli, J.; Calise, D.; Seguelas, M.H.; Ordener, C.; Piercecchi-Marti, M.D.; Auge, N.; Salvayre, A.N.; et al. Mesenchymal stem cells promote matrix metalloproteinase secretion by cardiac fibroblasts and reduce cardiac ventricular fibrosis after myocardial infarction. Stem Cells 2009, 27, 2734–2743. [Google Scholar] [CrossRef]
  89. Kou, M.; Huang, L.; Yang, J.; Chiang, Z.; Chen, S.; Liu, J.; Guo, L.; Zhang, X.; Zhou, X.; Xu, X.; et al. Mesenchymal stem cell-derived extracellular vesicles for immunomodulation and regeneration: A next generation therapeutic tool? Cell Death Dis. 2022, 13, 580. [Google Scholar] [CrossRef]
  90. Chen, T.S.; Liou, S.Y.; Lin, H.H.; Hung, M.Y.; Lin, C.C.; Lin, Y.M.; Lin, K.H.; Padma, V.V.; Yao, C.H.; Kuo, W.W.; et al. Oral administration of green tea Epigallocatechin-3-gallate reduces oxidative stress and enhances restoration of cardiac function in diabetic rats receiving autologous transplantation of adipose-derived stem cells. Arch. Physiol. Biochem. 2021, 127, 82–89. [Google Scholar] [CrossRef]
  91. Meng, K.; Cai, H.; Cai, S.; Hong, Y.; Zhang, X. Adiponectin Modified BMSCs Alleviate Heart Fibrosis via Inhibition TGF-beta1/Smad in Diabetic Rats. Front. Cell Dev. Biol. 2021, 9, 644160. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, W.Y.; Sun, H.H.; Sun, P.F. MicroRNA-378 attenuates myocardial fibrosis by inhibiting MAPK/ERK pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4398–4405. [Google Scholar] [CrossRef] [PubMed]
  93. Gubert, F.; da Silva, J.S.; Vasques, J.F.; de Jesus Gonçalves, R.G.; Martins, R.S.; de Sá, M.P.L.; Mendez-Otero, R.; Zapata-Sudo, G. Mesenchymal Stem Cells Therapies on Fibrotic Heart Diseases. Int. J. Mol. Sci. 2021, 22, 7447. [Google Scholar] [CrossRef]
  94. Ullah, M.; Liu, D.D.; Thakor, A.S. Mesenchymal Stromal Cell Homing: Mechanisms and Strategies for Improvement. iScience 2019, 15, 421–438. [Google Scholar] [CrossRef] [PubMed]
  95. Hocking, A.M. The Role of Chemokines in Mesenchymal Stem Cell Homing to Wounds. Adv. Wound Care 2015, 4, 623–630. [Google Scholar] [CrossRef]
  96. Rüster, B.; Göttig, S.; Ludwig, R.J.; Bistrian, R.; Müller, S.; Seifried, E.; Gille, J.; Henschler, R. Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood 2006, 108, 3938–3944. [Google Scholar] [CrossRef]
  97. Yau, T.M.; Pagani, F.D.; Mancini, D.M.; Chang, H.L.; Lala, A.; Woo, Y.J.; Acker, M.A.; Selzman, C.H.; Soltesz, E.G.; Kern, J.A.; et al. Intramyocardial Injection of Mesenchymal Precursor Cells and Successful Temporary Weaning from Left Ventricular Assist Device Support in Patients with Advanced Heart Failure: A Randomized Clinical Trial. JAMA 2019, 321, 1176–1186. [Google Scholar] [CrossRef] [PubMed]
  98. Walter, D.H.; Haendeler, J.; Reinhold, J.; Rochwalsky, U.; Seeger, F.; Honold, J.; Hoffmann, J.; Urbich, C.; Lehmann, R.; Arenzana-Seisdesdos, F.; et al. Impaired CXCR4 signaling contributes to the reduced neovascularization capacity of endothelial progenitor cells from patients with coronary artery disease. Circ. Res. 2005, 97, 1142–1151. [Google Scholar] [CrossRef]
  99. Chen, L.; Wu, F.; Xia, W.H.; Zhang, Y.Y.; Xu, S.Y.; Cheng, F.; Liu, X.; Zhang, X.Y.; Wang, S.M.; Tao, J. CXCR4 gene transfer contributes to in vivo reendothelialization capacity of endothelial progenitor cells. Cardiovasc. Res. 2010, 88, 462–470. [Google Scholar] [CrossRef]
  100. He, F.; Luo, P.F.; Tang, T.; Zhang, F.; Fang, H.; Ji, S.Z.; Sun, Y.; Wu, G.S.; Pan, B.H.; Huo, Z.B.; et al. Targeted release of stromal cell-derived factor-1α by reactive oxygen species-sensitive nanoparticles results in bone marrow stromal cell chemotaxis and homing, and repair of vascular injury caused by electrical burns. PLoS ONE 2018, 13, e0194298. [Google Scholar] [CrossRef]
  101. Adeshina, Y.O.; Deeds, E.J.; Karanicolas, J. Machine learning classification can reduce false positives in structure-based virtual screening. Proc. Natl. Acad. Sci. USA 2020, 117, 18477–18488. [Google Scholar] [CrossRef] [PubMed]
  102. Terashvili, M.; Bosnjak, Z.J. Stem Cell Therapies in Cardiovascular Disease. J. Cardiothorac. Vasc. Anesth. 2019, 33, 209–222. [Google Scholar] [CrossRef] [PubMed]
  103. Xu, S.; Qiu, Y.; Tao, J. The challenges and optimization of cell-based therapy for cardiovascular disease. J. Transl. Int. Med. 2021, 9, 234–238. [Google Scholar] [CrossRef] [PubMed]
  104. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [PubMed]
  105. Midha, S.; Jain, K.G.; Bhaskar, N.; Kaur, A.; Rawat, S.; Giri, S.; Basu, B.; Mohanty, S. Tissue-specific mesenchymal stem cell-dependent osteogenesis in highly porous chitosan-based bone analogs. Stem Cells Transl. Med. 2021, 10, 303–319. [Google Scholar] [CrossRef] [PubMed]
  106. Martínez-Carrasco, R.; Sánchez-Abarca, L.I.; Nieto-Gómez, C.; Martín García, E.; Sánchez-Guijo, F.; Argüeso, P.; Aijón, J.; Hernández-Galilea, E.; Velasco, A. Subconjunctival injection of mesenchymal stromal cells protects the cornea in an experimental model of GVHD. Ocul. Surf. 2019, 17, 285–294. [Google Scholar] [CrossRef] [PubMed]
  107. Petrou, P.; Gothelf, Y.; Argov, Z.; Gotkine, M.; Levy, Y.S.; Kassis, I.; Vaknin-Dembinsky, A.; Ben-Hur, T.; Offen, D.; Abramsky, O.; et al. Safety and Clinical Effects of Mesenchymal Stem Cells Secreting Neurotrophic Factor Transplantation in Patients with Amyotrophic Lateral Sclerosis: Results of Phase 1/2 and 2a Clinical Trials. JAMA Neurol. 2016, 73, 337–344. [Google Scholar] [CrossRef] [PubMed]
  108. Zhao, K.; Liu, Q. The clinical application of mesenchymal stromal cells in hematopoietic stem cell transplantation. J. Hematol. Oncol. 2016, 9, 46. [Google Scholar] [CrossRef] [PubMed]
  109. Conrad, C.; Niess, H.; Huss, R.; Huber, S.; von Luettichau, I.; Nelson, P.J.; Ott, H.C.; Jauch, K.W.; Bruns, C.J. Multipotent mesenchymal stem cells acquire a lymphendothelial phenotype and enhance lymphatic regeneration in vivo. Circulation 2009, 119, 281–289. [Google Scholar] [CrossRef]
  110. Haga, H.; Yan, I.K.; Takahashi, K.; Wood, J.; Zubair, A.; Patel, T. Tumour cell-derived extracellular vesicles interact with mesenchymal stem cells to modulate the microenvironment and enhance cholangiocarcinoma growth. J. Extracell. Vesicles 2015, 4, 24900. [Google Scholar] [CrossRef]
  111. Wang, T.; Zhang, J.; Liao, J.; Zhang, F.; Zhou, G. Donor genetic backgrounds contribute to the functional heterogeneity of stem cells and clinical outcomes. Stem Cells Transl. Med. 2020, 9, 1495–1499. [Google Scholar] [CrossRef] [PubMed]
  112. Alves, H.; van Ginkel, J.; Groen, N.; Hulsman, M.; Mentink, A.; Reinders, M.; van Blitterswijk, C.; de Boer, J. A mesenchymal stromal cell gene signature for donor age. PLoS ONE 2012, 7, e42908. [Google Scholar] [CrossRef] [PubMed]
  113. Pachón-Peña, G.; Serena, C.; Ejarque, M.; Petriz, J.; Duran, X.; Oliva-Olivera, W.; Simó, R.; Tinahones, F.J.; Fernández-Veledo, S.; Vendrell, J. Obesity Determines the Immunophenotypic Profile and Functional Characteristics of Human Mesenchymal Stem Cells from Adipose Tissue. Stem Cells Transl. Med. 2016, 5, 464–475. [Google Scholar] [CrossRef]
  114. Dlouhy, B.J.; Awe, O.; Rao, R.C.; Kirby, P.A.; Hitchon, P.W. Autograft-derived spinal cord mass following olfactory mucosal cell transplantation in a spinal cord injury patient: Case report. J. Neurosurg. Spine 2014, 21, 618–622. [Google Scholar] [CrossRef] [PubMed]
  115. Ducret, M.; Fabre, H.; Farges, J.C.; Degoul, O.; Atzeni, G.; McGuckin, C.; Forraz, N.; Mallein-Gerin, F.; Perrier-Groult, E. Production of Human Dental Pulp Cells with a Medicinal Manufacturing Approach. J. Endod. 2015, 41, 1492–1499. [Google Scholar] [CrossRef] [PubMed]
  116. Torre, M.L.; Lucarelli, E.; Guidi, S.; Ferrari, M.; Alessandri, G.; De Girolamo, L.; Pessina, A.; Ferrero, I. Ex vivo expanded mesenchymal stromal cell minimal quality requirements for clinical application. Stem Cells Dev. 2015, 24, 677–685. [Google Scholar] [CrossRef] [PubMed]
  117. Codinach, M.; Blanco, M.; Ortega, I.; Lloret, M.; Reales, L.; Coca, M.I.; Torrents, S.; Doral, M.; Oliver-Vila, I.; Requena-Montero, M.; et al. Design and validation of a consistent and reproducible manufacture process for the production of clinical-grade bone marrow-derived multipotent mesenchymal stromal cells. Cytotherapy 2016, 18, 1197–1208. [Google Scholar] [CrossRef]
  118. Bloor, A.J.C.; Patel, A.; Griffin, J.E.; Gilleece, M.H.; Radia, R.; Yeung, D.T.; Drier, D.; Larson, L.S.; Uenishi, G.I.; Hei, D.; et al. Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: A phase I, multicenter, open-label, dose-escalation study. Nat. Med. 2020, 26, 1720–1725. [Google Scholar] [CrossRef] [PubMed]
  119. Brown, P.T.; Handorf, A.M.; Jeon, W.B.; Li, W.J. Stem cell-based tissue engineering approaches for musculoskeletal regeneration. Curr. Pharm. Des. 2013, 19, 3429–3445. [Google Scholar] [CrossRef]
  120. Narayanan, G.; Bhattacharjee, M.; Nair, L.S.; Laurencin, C.T. Musculoskeletal Tissue Regeneration: The Role of the Stem Cells. Regen. Eng. Transl. Med. 2017, 3, 133–165. [Google Scholar] [CrossRef]
  121. Vizoso, F.J.; Eiro, N.; Cid, S.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int. J. Mol. Sci. 2017, 18, 1852. [Google Scholar] [CrossRef] [PubMed]
  122. Pawitan, J.A. Prospect of stem cell conditioned medium in regenerative medicine. BioMed Res. Int. 2014, 2014, 965849. [Google Scholar] [CrossRef] [PubMed]
  123. Konala, V.B.; Mamidi, M.K.; Bhonde, R.; Das, A.K.; Pochampally, R.; Pal, R. The current landscape of the mesenchymal stromal cell secretome: A new paradigm for cell-free regeneration. Cytotherapy 2016, 18, 13–24. [Google Scholar] [CrossRef]
  124. Caccia, D.; Dugo, M.; Callari, M.; Bongarzone, I. Bioinformatics tools for secretome analysis. Biochim. Biophys. Acta 2013, 1834, 2442–2453. [Google Scholar] [CrossRef] [PubMed]
  125. Cases-Perera, O.; Blanco-Elices, C.; Chato-Astrain, J.; Miranda-Fernández, C.; Campos, F.; Crespo, P.V.; Sánchez-Montesinos, I.; Alaminos, M.; Martín-Piedra, M.A.; Garzón, I. Development of secretome-based strategies to improve cell culture protocols in tissue engineering. Sci. Rep. 2022, 12, 10003. [Google Scholar] [CrossRef]
  126. Tran, C.; Damaser, M.S. Stem cells as drug delivery methods: Application of stem cell secretome for regeneration. Adv. Drug Deliv. Rev. 2015, 82–83, 1–11. [Google Scholar] [CrossRef]
  127. García-García, Ó.D.; El Soury, M.; González-Quevedo, D.; Sánchez-Porras, D.; Chato-Astrain, J.; Campos, F.; Carriel, V. Histological, Biomechanical, and Biological Properties of Genipin-Crosslinked Decellularized Peripheral Nerves. Int. J. Mol. Sci. 2021, 22, 674. [Google Scholar] [CrossRef]
  128. Irastorza-Lorenzo, A.; Sánchez-Porras, D.; Ortiz-Arrabal, O.; de Frutos, M.J.; Esteban, E.; Fernández, J.; Janer, A.; Campos, A.; Campos, F.; Alaminos, M. Evaluation of Marine Agarose Biomaterials for Tissue Engineering Applications. Int. J. Mol. Sci. 2021, 22, 1923. [Google Scholar] [CrossRef]
  129. Martin-Piedra, M.A.; Garzon, I.; Oliveira, A.C.; Alfonso-Rodriguez, C.A.; Carriel, V.; Scionti, G.; Alaminos, M. Cell viability and proliferation capability of long-term human dental pulp stem cell cultures. Cytotherapy 2014, 16, 266–277. [Google Scholar] [CrossRef]
  130. Sánchez-Porras, D.; Durand-Herrera, D.; Paes, A.B.; Chato-Astrain, J.; Verplancke, R.; Vanfleteren, J.; Sánchez-López, J.D.; García-García, Ó.D.; Campos, F.; Carriel, V. Ex Vivo Generation and Characterization of Human Hyaline and Elastic Cartilaginous Microtissues for Tissue Engineering Applications. Biomedicines 2021, 9, 292. [Google Scholar] [CrossRef]
  131. Constantin, A.; Comarița, I.K.; Alexandru, N.; Filippi, A.; Bojin, F.; Gherghiceanu, M.; Vîlcu, A.; Nemecz, M.; Niculescu, L.S.; Păunescu, V.; et al. Stem cell-derived extracellular vesicles reduce the expression of molecules involved in cardiac hypertrophy-In a model of human-induced pluripotent stem cell-derived cardiomyocytes. Front. Pharmacol. 2022, 13, 1003684. [Google Scholar] [CrossRef] [PubMed]
  132. Silveira, B.M.; Ribeiro, T.O.; Freitas, R.S.; Carreira, A.C.O.; Gonçalves, M.S.; Sogayar, M.; Meyer, R.; Birbrair, A.; Fortuna, V. Secretome from human adipose-derived mesenchymal stem cells promotes blood vessel formation and pericyte coverage in experimental skin repair. PLoS ONE 2022, 17, e0277863. [Google Scholar] [CrossRef] [PubMed]
  133. Daneshmandi, L.; Shah, S.; Jafari, T.; Bhattacharjee, M.; Momah, D.; Saveh-Shemshaki, N.; Lo, K.W.; Laurencin, C.T. Emergence of the Stem Cell Secretome in Regenerative Engineering. Trends Biotechnol. 2020, 38, 1373–1384. [Google Scholar] [CrossRef]
  134. Md Fadilah, N.I.; Mohd Abdul Kader Jailani, M.S.; Badrul Hisham, M.A.I.; Sunthar Raj, N.; Shamsuddin, S.A.; Ng, M.H.; Fauzi, M.B.; Maarof, M. Cell secretomes for wound healing and tissue regeneration: Next generation acellular based tissue engineered products. J. Tissue Eng. 2022, 13, 20417314221114273. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, J.; Li, J.; Qu, X.; Liu, Y.; Sun, L.; Harada, A.; Hua, Y.; Sougawa, N.; Tabata, A.; Liu, L.; et al. Development of composite functional tissue sheets using hiPSC-CMs and hADSCs to improve the cardiac function after myocardial infarction. Bioact. Mater. 2024, 37, 533–548. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, J.; Li, J.; Qu, X.; Liu, Y.; Harada, A.; Hua, Y.; Yoshida, N.; Ishida, M.; Tabata, A.; Sun, L.; et al. Development of a thick and functional human adipose-derived stem cell tissue sheet for myocardial infarction repair in rat hearts. Stem Cell Res. Ther. 2023, 14, 380. [Google Scholar] [CrossRef]
  137. Park, S.R.; Kim, S.R.; Im, J.B.; Park, C.H.; Lee, H.Y.; Hong, I.S. 3D stem cell-laden artificial endometrium: Successful endometrial regeneration and pregnancy. Biofabrication 2021, 13, 045012. [Google Scholar] [CrossRef] [PubMed]
  138. Mazzoni, E.; Iaquinta, M.R.; Lanzillotti, C.; Mazziotta, C.; Maritati, M.; Montesi, M.; Sprio, S.; Tampieri, A.; Tognon, M.; Martini, F. Bioactive Materials for Soft Tissue Repair. Front. Bioeng. Biotechnol. 2021, 9, 613787. [Google Scholar] [CrossRef]
  139. Rice, J.J.; Martino, M.M.; De Laporte, L.; Tortelli, F.; Briquez, P.S.; Hubbell, J.A. Engineering the regenerative microenvironment with biomaterials. Adv. Healthc. Mater. 2013, 2, 57–71. [Google Scholar] [CrossRef]
  140. Wang, C.Y.; Hong, P.D.; Wang, D.H.; Cherng, J.H.; Chang, S.J.; Liu, C.C.; Fang, T.J.; Wang, Y.W. Polymeric Gelatin Scaffolds Affect Mesenchymal Stem Cell Differentiation and Its Diverse Applications in Tissue Engineering. Int. J. Mol. Sci. 2020, 21, 8632. [Google Scholar] [CrossRef]
  141. Bonferoni, M.C.; Caramella, C.; Catenacci, L.; Conti, B.; Dorati, R.; Ferrari, F.; Genta, I.; Modena, T.; Perteghella, S.; Rossi, S.; et al. Biomaterials for Soft Tissue Repair and Regeneration: A Focus on Italian Research in the Field. Pharmaceutics 2021, 13, 1341. [Google Scholar] [CrossRef] [PubMed]
  142. Nikolova, M.P.; Chavali, M.S. Recent advances in biomaterials for 3D scaffolds: A review. Bioact. Mater. 2019, 4, 271–292. [Google Scholar] [CrossRef] [PubMed]
  143. Salerno, A.; Netti, P.A. Review on Computer-Aided Design and Manufacturing of Drug Delivery Scaffolds for Cell Guidance and Tissue Regeneration. Front. Bioeng. Biotechnol. 2021, 9, 682133. [Google Scholar] [CrossRef] [PubMed]
  144. Olson, J.L.; Atala, A.; Yoo, J.J. Tissue engineering: Current strategies and future directions. Chonnam Med. J. 2011, 47, 1–13. [Google Scholar] [CrossRef] [PubMed]
  145. Alkaya, D.; Gurcan, C.; Kilic, P.; Yilmazer, A.; Gurman, G. Where is human-based cellular pharmaceutical R&D taking us in cartilage regeneration? 3 Biotech 2020, 10, 161. [Google Scholar] [CrossRef] [PubMed]
  146. Mohammadinejad, R.; Kumar, A.; Ranjbar-Mohammadi, M.; Ashrafizadeh, M.; Han, S.S.; Khang, G.; Roveimiab, Z. Recent Advances in Natural Gum-Based Biomaterials for Tissue Engineering and Regenerative Medicine: A Review. Polymers 2020, 12, 176. [Google Scholar] [CrossRef] [PubMed]
  147. Bharadwaz, A.; Jayasuriya, A.C. Recent trends in the application of widely used natural and synthetic polymer nanocomposites in bone tissue regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 110, 110698. [Google Scholar] [CrossRef] [PubMed]
  148. Abbas, Y.; Brunel, L.G.; Hollinshead, M.S.; Fernando, R.C.; Gardner, L.; Duncan, I.; Moffett, A.; Best, S.; Turco, M.Y.; Burton, G.J.; et al. Generation of a three-dimensional collagen scaffold-based model of the human endometrium. Interface Focus 2020, 10, 20190079. [Google Scholar] [CrossRef] [PubMed]
  149. Peressotti, S.; Koehl, G.E.; Goding, J.A.; Green, R.A. Self-Assembling Hydrogel Structures for Neural Tissue Repair. ACS Biomater. Sci. Eng. 2021, 7, 4136–4163. [Google Scholar] [CrossRef]
  150. Echeverria Molina, M.I.; Malollari, K.G.; Komvopoulos, K. Design Challenges in Polymeric Scaffolds for Tissue Engineering. Front. Bioeng. Biotechnol. 2021, 9, 617141. [Google Scholar] [CrossRef]
  151. Pearce, A.K.; O’Reilly, R.K. Polymers for Biomedical Applications: The Importance of Hydrophobicity in Directing Biological Interactions and Application Efficacy. Biomacromolecules 2021, 22, 4459–4469. [Google Scholar] [CrossRef] [PubMed]
  152. Barthes, J.; Özçelik, H.; Hindié, M.; Ndreu-Halili, A.; Hasan, A.; Vrana, N.E. Cell microenvironment engineering and monitoring for tissue engineering and regenerative medicine: The recent advances. BioMed Res. Int. 2014, 2014, 921905. [Google Scholar] [CrossRef] [PubMed]
  153. Nicolas, J.; Magli, S.; Rabbachin, L.; Sampaolesi, S.; Nicotra, F.; Russo, L. 3D Extracellular Matrix Mimics: Fundamental Concepts and Role of Materials Chemistry to Influence Stem Cell Fate. Biomacromolecules 2020, 21, 1968–1994. [Google Scholar] [CrossRef] [PubMed]
  154. Antmen, E.; Vrana, N.E.; Hasirci, V. The role of biomaterials and scaffolds in immune responses in regenerative medicine: Macrophage phenotype modulation by biomaterial properties and scaffold architectures. Biomater. Sci. 2021, 9, 8090–8110. [Google Scholar] [CrossRef] [PubMed]
  155. Mastrogiacomo, S.; Dou, W.; Jansen, J.A.; Walboomers, X.F. Magnetic Resonance Imaging of Hard Tissues and Hard Tissue Engineered Bio-substitutes. Mol. Imaging Biol. 2019, 21, 1003–1019. [Google Scholar] [CrossRef] [PubMed]
  156. Metcalfe, A.D.; Ferguson, M.W. Tissue engineering of replacement skin: The crossroads of biomaterials, wound healing, embryonic development, stem cells and regeneration. J. R. Soc. Interface 2007, 4, 413–437. [Google Scholar] [CrossRef]
  157. Campostrini, G.; Windt, L.M.; van Meer, B.J.; Bellin, M.; Mummery, C.L. Cardiac Tissues from Stem Cells: New Routes to Maturation and Cardiac Regeneration. Circ. Res. 2021, 128, 775–801. [Google Scholar] [CrossRef] [PubMed]
  158. Abbasgholizadeh, R.; Islas, J.F.; Navran, S.; Potaman, V.N.; Schwartz, R.J.; Birla, R.K. A Highly Conductive 3D Cardiac Patch Fabricated Using Cardiac Myocytes Reprogrammed from Human Adipogenic Mesenchymal Stem Cells. Cardiovasc. Eng. Technol. 2020, 11, 205–218. [Google Scholar] [CrossRef] [PubMed]
  159. Schmuck, E.G.; Mulligan, J.D.; Ertel, R.L.; Kouris, N.A.; Ogle, B.M.; Raval, A.N.; Saupe, K.W. Cardiac fibroblast-derived 3D extracellular matrix seeded with mesenchymal stem cells as a novel device to transfer cells to the ischemic myocardium. Cardiovasc. Eng. Technol. 2014, 5, 119–131. [Google Scholar] [CrossRef]
  160. Habanjar, O.; Diab-Assaf, M.; Caldefie-Chezet, F.; Delort, L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int. J. Mol. Sci. 2021, 22, 12200. [Google Scholar] [CrossRef]
  161. Williams, D.F. There is no such thing as a biocompatible material. Biomaterials 2014, 35, 10009–10014. [Google Scholar] [CrossRef]
  162. Cosgrove, B.D.; Mui, K.L.; Driscoll, T.P.; Caliari, S.R.; Mehta, K.D.; Assoian, R.K.; Burdick, J.A.; Mauck, R.L. N-cadherin adhesive interactions modulate matrix mechanosensing and fate commitment of mesenchymal stem cells. Nat. Mater. 2016, 15, 1297–1306. [Google Scholar] [CrossRef] [PubMed]
  163. Ezquerra, S.; Zuleta, A.; Arancibia, R.; Estay, J.; Aulestia, F.; Carrion, F. Functional Properties of Human-Derived Mesenchymal Stem Cell Spheroids: A Meta-Analysis and Systematic Review. Stem Cells Int. 2021, 2021, 8825332. [Google Scholar] [CrossRef] [PubMed]
  164. Kusuma, G.D.; Li, A.; Zhu, D.; McDonald, H.; Inocencio, I.M.; Chambers, D.C.; Sinclair, K.; Fang, H.; Greening, D.W.; Frith, J.E.; et al. Effect of 2D and 3D Culture Microenvironments on Mesenchymal Stem Cell-Derived Extracellular Vesicles Potencies. Front. Cell Dev. Biol. 2022, 10, 819726. [Google Scholar] [CrossRef]
  165. Asahara, T.; Kalka, C.; Isner, J.M. Stem cell therapy and gene transfer for regeneration. Gene Ther. 2000, 7, 451–457. [Google Scholar] [CrossRef]
  166. Mitrecic, D.; Nicaise, C.; Klimaschewski, L.; Gajovic, S.; Bohl, D.; Pochet, R. Genetically modified stem cells for the treatment of neurological diseases. Front. Biosci. (Elite Ed.) 2012, 4, 1170–1181. [Google Scholar] [CrossRef] [PubMed]
  167. Choi, J.R.; Pingguan-Murphy, B.; Wan Abas, W.A.; Noor Azmi, M.A.; Omar, S.Z.; Chua, K.H.; Wan Safwani, W.K. Impact of low oxygen tension on stemness, proliferation and differentiation potential of human adipose-derived stem cells. Biochem. Biophys. Res. Commun. 2014, 448, 218–224. [Google Scholar] [CrossRef]
  168. Yamamoto, Y.; Fujita, M.; Tanaka, Y.; Kojima, I.; Kanatani, Y.; Ishihara, M.; Tachibana, S. Low oxygen tension enhances proliferation and maintains stemness of adipose tissue-derived stromal cells. Biores Open Access 2013, 2, 199–205. [Google Scholar] [CrossRef] [PubMed]
  169. Lee, J.H.; Yoon, Y.M.; Lee, S.H. Hypoxic Preconditioning Promotes the Bioactivities of Mesenchymal Stem Cells via the HIF-1α-GRP78-Akt Axis. Int. J. Mol. Sci. 2017, 18, 1320. [Google Scholar] [CrossRef]
  170. Seo, Y.; Shin, T.H.; Kim, H.S. Current Strategies to Enhance Adipose Stem Cell Function: An Update. Int. J. Mol. Sci. 2019, 20, 3827. [Google Scholar] [CrossRef]
  171. Kurogoushi, R.; Hasegawa, T.; Akazawa, Y.; Iwata, K.; Sugimoto, A.; Yamaguchi-Ueda, K.; Miyazaki, A.; Narwidina, A.; Kawarabayashi, K.; Kitamura, T.; et al. Fibroblast growth factor 2 suppresses the expression of C-C motif chemokine 11 through the c-Jun N-terminal kinase pathway in human dental pulp-derived mesenchymal stem cells. Exp. Ther. Med. 2021, 22, 1356. [Google Scholar] [CrossRef] [PubMed]
  172. Ratajczak, M.Z.; Zuba-Surma, E.; Kucia, M.; Reca, R.; Wojakowski, W.; Ratajczak, J. The pleiotropic effects of the SDF-1-CXCR4 axis in organogenesis, regeneration and tumorigenesis. Leukemia 2006, 20, 1915–1924. [Google Scholar] [CrossRef]
  173. Wisel, S.; Khan, M.; Kuppusamy, M.L.; Mohan, I.K.; Chacko, S.M.; Rivera, B.K.; Sun, B.C.; Hideg, K.; Kuppusamy, P. Pharmacological preconditioning of mesenchymal stem cells with trimetazidine (1-[2,3,4-trimethoxybenzyl]piperazine) protects hypoxic cells against oxidative stress and enhances recovery of myocardial function in infarcted heart through Bcl-2 expression. J. Pharmacol. Exp. Ther. 2009, 329, 543–550. [Google Scholar] [CrossRef] [PubMed]
  174. Haider, H.; Lee, Y.J.; Jiang, S.; Ahmed, R.P.; Ryon, M.; Ashraf, M. Phosphodiesterase inhibition with tadalafil provides longer and sustained protection of stem cells. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1395–H1404. [Google Scholar] [CrossRef] [PubMed]
  175. Song, L.; Yang, Y.J.; Dong, Q.T.; Qian, H.Y.; Gao, R.L.; Qiao, S.B.; Shen, R.; He, Z.X.; Lu, M.J.; Zhao, S.H.; et al. Atorvastatin enhance efficacy of mesenchymal stem cells treatment for swine myocardial infarction via activation of nitric oxide synthase. PLoS ONE 2013, 8, e65702. [Google Scholar] [CrossRef] [PubMed]
  176. Li, M.; Jiang, Y.; Hou, Q.; Zhao, Y.; Zhong, L.; Fu, X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: Current status and future prospects. Stem Cell Res. Ther. 2022, 13, 146. [Google Scholar] [CrossRef] [PubMed]
  177. Dai, G.; Xu, Q.; Luo, R.; Gao, J.; Chen, H.; Deng, Y.; Li, Y.; Wang, Y.; Yuan, W.; Wu, X. Atorvastatin treatment improves effects of implanted mesenchymal stem cells: Meta-analysis of animal models with acute myocardial infarction. BMC Cardiovasc. Disord. 2015, 15, 170. [Google Scholar] [CrossRef] [PubMed]
  178. Salem, H.K.; Thiemermann, C. Mesenchymal stromal cells: Current understanding and clinical status. Stem Cells 2010, 28, 585–596. [Google Scholar] [CrossRef]
  179. DelaRosa, O.; Lombardo, E. Modulation of adult mesenchymal stem cells activity by toll-like receptors: Implications on therapeutic potential. Mediat. Inflamm. 2010, 2010, 865601. [Google Scholar] [CrossRef]
  180. Tran, T.; Cruz, C.; Chan, A.; Awad, S.; Rajasingh, J.; Deth, R.; Gurusamy, N. Mesenchymal Stem Cell-Derived Long Noncoding RNAs in Cardiac Injury and Repair. Cells 2023, 12, 2268. [Google Scholar] [CrossRef]
  181. Nisole, S.; Saïb, A. Early steps of retrovirus replicative cycle. Retrovirology 2004, 1, 9. [Google Scholar] [CrossRef] [PubMed]
  182. Dahlberg, J.E. An overview of retrovirus replication and classification. Adv. Vet. Sci. Comp. Med. 1988, 32, 1–35. [Google Scholar] [CrossRef] [PubMed]
  183. Nitta, S.; Kusakari, Y.; Yamada, Y.; Kubo, T.; Neo, S.; Igarashi, H.; Hisasue, M. Conversion of mesenchymal stem cells into a canine hepatocyte-like cells by Foxa1 and Hnf4a. Regen. Ther. 2020, 14, 165–176. [Google Scholar] [CrossRef] [PubMed]
  184. Subbramanian, R.A.; Cohen, E.A. Molecular biology of the human immunodeficiency virus accessory proteins. J. Virol. 1994, 68, 6831–6835. [Google Scholar] [CrossRef]
  185. Naldini, L.; Blömer, U.; Gallay, P.; Ory, D.; Mulligan, R.; Gage, F.H.; Verma, I.M.; Trono, D. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 1996, 272, 263–267. [Google Scholar] [CrossRef] [PubMed]
  186. Santoni de Sio, F.R.; Gritti, A.; Cascio, P.; Neri, M.; Sampaolesi, M.; Galli, C.; Luban, J.; Naldini, L. Lentiviral vector gene transfer is limited by the proteasome at postentry steps in various types of stem cells. Stem Cells 2008, 26, 2142–2152. [Google Scholar] [CrossRef] [PubMed]
  187. Bao, C.; Guo, J.; Lin, G.; Hu, M.; Hu, Z. TNFR gene-modified mesenchymal stem cells attenuate inflammation and cardiac dysfunction following MI. Scand. Cardiovasc. J. 2008, 42, 56–62. [Google Scholar] [CrossRef] [PubMed]
  188. Abdul Halim, N.S.; Fakiruddin, K.S.; Ali, S.A.; Yahaya, B.H. A comparative study of non-viral gene delivery techniques to human adipose-derived mesenchymal stem cell. Int. J. Mol. Sci. 2014, 15, 15044–15060. [Google Scholar] [CrossRef] [PubMed]
  189. Feril, L.B., Jr. Ultrasound-mediated gene transfection. Methods Mol. Biol. 2009, 542, 179–194. [Google Scholar] [CrossRef]
  190. Zhou, H.; He, Y.; Xiong, W.; Jing, S.; Duan, X.; Huang, Z.; Nahal, G.S.; Peng, Y.; Li, M.; Zhu, Y.; et al. MSC based gene delivery methods and strategies improve the therapeutic efficacy of neurological diseases. Bioact. Mater. 2023, 23, 409–437. [Google Scholar] [CrossRef]
  191. Tang, J.; Wang, J.; Yang, J.; Kong, X.; Zheng, F.; Guo, L.; Zhang, L.; Huang, Y. Mesenchymal stem cells over-expressing SDF-1 promote angiogenesis and improve heart function in experimental myocardial infarction in rats. Eur. J. Cardiothorac. Surg. 2009, 36, 644–650. [Google Scholar] [CrossRef] [PubMed]
  192. Hnatiuk, A.P.; Ong, S.G.; Olea, F.D.; Locatelli, P.; Riegler, J.; Lee, W.H.; Jen, C.H.; De Lorenzi, A.; Giménez, C.S.; Laguens, R.; et al. Allogeneic Mesenchymal Stromal Cells Overexpressing Mutant Human Hypoxia-Inducible Factor 1-α (HIF1-α) in an Ovine Model of Acute Myocardial Infarction. J. Am. Heart Assoc. 2016, 5, e003714. [Google Scholar] [CrossRef]
  193. Gómez-Mauricio, G.; Moscoso, I.; Martín-Cancho, M.F.; Crisóstomo, V.; Prat-Vidal, C.; Báez-Díaz, C.; Sánchez-Margallo, F.M.; Bernad, A. Combined administration of mesenchymal stem cells overexpressing IGF-1 and HGF enhances neovascularization but moderately improves cardiac regeneration in a porcine model. Stem Cell Res. Ther. 2016, 7, 94. [Google Scholar] [CrossRef] [PubMed]
  194. Silva, D.N.; Souza, B.S.F.; Vasconcelos, J.F.; Azevedo, C.M.; Valim, C.X.R.; Paredes, B.D.; Rocha, V.P.C.; Carvalho, G.B.; Daltro, P.S.; Macambira, S.G.; et al. Granulocyte-Colony Stimulating Factor-Overexpressing Mesenchymal Stem Cells Exhibit Enhanced Immunomodulatory Actions through the Recruitment of Suppressor Cells in Experimental Chagas Disease Cardiomyopathy. Front. Immunol. 2018, 9, 1449. [Google Scholar] [CrossRef]
  195. Huang, F.; Zhu, X.; Hu, X.Q.; Fang, Z.F.; Tang, L.; Lu, X.L.; Zhou, S.H. Mesenchymal stem cells modified with miR-126 release angiogenic factors and activate Notch ligand Delta-like-4, enhancing ischemic angiogenesis and cell survival. Int. J. Mol. Med. 2013, 31, 484–492. [Google Scholar] [CrossRef]
  196. Raman, N.; Imran, S.A.M.; Ahmad Amin Noordin, K.B.; Wan Kamarul Zaman, W.S.; Nordin, F. Mechanotransduction of mesenchymal stem cells (MSCs) during cardiomyocytes differentiation. Heliyon 2022, 8, e11624. [Google Scholar] [CrossRef] [PubMed]
  197. Wu, C.C.; Chao, Y.C.; Chen, C.N.; Chien, S.; Chen, Y.C.; Chien, C.C.; Chiu, J.J.; Linju Yen, B. Synergism of biochemical and mechanical stimuli in the differentiation of human placenta-derived multipotent cells into endothelial cells. J. Biomech. 2008, 41, 813–821. [Google Scholar] [CrossRef]
  198. Ravishankar, P.; Tandon, I.; Balachandran, K. Effect of Cyclic Uniaxial Mechanical Strain on Endothelial Progenitor Cell Differentiation. Cardiovasc. Eng. Technol. 2022, 13, 872–885. [Google Scholar] [CrossRef]
  199. Sivaraman, S.; Ravishankar, P.; Rao, R.R. Differentiation and Engineering of Human Stem Cells for Smooth Muscle Generation. Tissue Eng. Part. B Rev. 2023, 29, 1–9. [Google Scholar] [CrossRef]
  200. Bravo-Olín, J.; Martínez-Carreón, S.A.; Francisco-Solano, E.; Lara, A.R.; Beltran-Vargas, N.E. Analysis of the role of perfusion, mechanical, and electrical stimulation in bioreactors for cardiac tissue engineering. Bioprocess Biosyst. Eng. 2024. [Google Scholar] [CrossRef]
  201. Chouaib, B.; Haack-Sørensen, M.; Chaubron, F.; Cuisinier, F.; Collart-Dutilleul, P.Y. Towards the Standardization of Mesenchymal Stem Cell Secretome-Derived Product Manufacturing for Tissue Regeneration. Int. J. Mol. Sci. 2023, 24, 12594. [Google Scholar] [CrossRef] [PubMed]
  202. Gouveia, B.G.; Rijo, P.; Gonçalo, T.S.; Reis, C.P. Good manufacturing practices for medicinal products for human use. J. Pharm. Bioallied Sci. 2015, 7, 87–96. [Google Scholar] [CrossRef] [PubMed]
  203. Olsen, T.R.; Ng, K.S.; Lock, L.T.; Ahsan, T.; Rowley, J.A. Peak MSC-Are We There Yet? Front. Med. 2018, 5, 178. [Google Scholar] [CrossRef]
Figure 1. Optimization strategies for enhanced MSC therapeutic efficacy in cardiovascular treatments. This diagram depicts the process of enhancing the efficacy of mesenchymal stem cells (MSCs) for cardiovascular applications. Unmodified MSCs face a hostile cellular environment, leading to reduced survival, premature senescence, and increased cell death, which collectively result in decreased therapeutic efficacy. On the other hand, MSCs undergoing preparation following GMP standards or genetically modified MSCs or MSCs preconditioned with growth factors, cytokines, or hypoxic conditions overcome the limitations of the unmodified state. The resultant modified MSCs exhibit enhanced adaptation to the injury environment, increased survival, and improved capacity for tissue regeneration, thus significantly improving therapeutic outcomes. The figure highlights the critical role of MSC optimization in advancing regenerative cardiovascular medicine. The figure above was partly generated using Servier Medical Art, provided by Servier, licensed under an unported Creative Commons Attribution 3.0 license.
Figure 1. Optimization strategies for enhanced MSC therapeutic efficacy in cardiovascular treatments. This diagram depicts the process of enhancing the efficacy of mesenchymal stem cells (MSCs) for cardiovascular applications. Unmodified MSCs face a hostile cellular environment, leading to reduced survival, premature senescence, and increased cell death, which collectively result in decreased therapeutic efficacy. On the other hand, MSCs undergoing preparation following GMP standards or genetically modified MSCs or MSCs preconditioned with growth factors, cytokines, or hypoxic conditions overcome the limitations of the unmodified state. The resultant modified MSCs exhibit enhanced adaptation to the injury environment, increased survival, and improved capacity for tissue regeneration, thus significantly improving therapeutic outcomes. The figure highlights the critical role of MSC optimization in advancing regenerative cardiovascular medicine. The figure above was partly generated using Servier Medical Art, provided by Servier, licensed under an unported Creative Commons Attribution 3.0 license.
Cells 13 00855 g001
Table 1. Overview of emerging MSC-based therapeutic strategies in tissue engineering and regeneration.
Table 1. Overview of emerging MSC-based therapeutic strategies in tissue engineering and regeneration.
MethodFeasibilityAdvantagesDisadvantagesReferences
Cell-Free Approach (Secretome)The MSC secretome encompasses a range of bioactive factors, including growth factors, cytokines, and chemokines, which contribute to cell proliferation, migration, and tissue repair.1. Risk of immunological rejection is minimized;
2. Secretome offers a cell-free treatment option;
3. High compatibility with host tissues;
4. Ease of delivery;
5. Lower tumorigenic potential compared to cellular therapies.
Limited by the absence of standardized protocols for secretome preparation[57,121,122,123,124,125,131]
Scaffold-based TherapeuticsScaffold-based treatments have emerged as a notable breakthrough in tissue engineering, providing promising approaches for healing injured tissues and reinstating their structure and function.Essential for regenerating a variety of tissues, providing structural support and a conducive environment for cell attachment and growth.Potential for host versus graft rejection and suboptimal mechanical properties that may not withstand long-term stress[135,136,137,138,139,140,141]
Three-Dimensional Ex Vivo PropagationThe aggregation of MSCs in a three-dimensional (3D) structure enhanced several biological characteristics, such as the ability to differentiate into multiple cell lineages, the production of therapeutic factors, and the ability to withstand ischemic conditions.Enhances differentiation toward skeleton-related tissues and the production of therapeutic factors.Challenges include replicating the ECM, maintaining uniform cell distribution, and ensuring high cell viability[157,158,159,160,161]
Physiological and Pathological Microenvironment ActivationExposing MSCs to varying oxygen tensions and inflammatory cytokines simulates physiological and pathological conditions, respectively.Physiological activation maintains stemness under hypoxia; pathological activation enhances immunomodulation and tissue regeneration responses via cytokines and growth factors.Precise environmental control is needed to simulate conditions effectively, posing operational challenges[167,168,169,170,171,172,173,174,175,176,177]
Genetic ModificationGenetic modifications of MSCs for targeted repair are performed using both viral vectors and non-viral methods to enhance specificity and efficacy.1. High efficiency and stability in viral gene delivery;
2. Reduced inflammatory responses and fibrosis in disease models;
3. Boosted therapeutic effectiveness for infarcted hearts with miR-126.
Side effects from viral-mediated delivery and high costs[184,185,186,187,191,192,193]
Mechanobiology-Mediated DifferentiationShear stress and mechanical strains direct MSCs and EPCs toward an endothelial phenotype, enhancing endothelial markers and functions.1. Promotes endothelial-like functions, enhancing tubule formation and LDL uptake;
2. Guides stem cells toward endothelial lineages, enhancing migration and tubulogenesis, which are crucial for vascular tissue engineering.
Requires precise control over mechanical conditions to ensure effective differentiation and functional outcomes in ECs[54,196,197,198,199,200]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kumar, R.; Mishra, N.; Tran, T.; Kumar, M.; Vijayaraghavalu, S.; Gurusamy, N. Emerging Strategies in Mesenchymal Stem Cell-Based Cardiovascular Therapeutics. Cells 2024, 13, 855. https://doi.org/10.3390/cells13100855

AMA Style

Kumar R, Mishra N, Tran T, Kumar M, Vijayaraghavalu S, Gurusamy N. Emerging Strategies in Mesenchymal Stem Cell-Based Cardiovascular Therapeutics. Cells. 2024; 13(10):855. https://doi.org/10.3390/cells13100855

Chicago/Turabian Style

Kumar, Rishabh, Nitin Mishra, Talan Tran, Munish Kumar, Sivakumar Vijayaraghavalu, and Narasimman Gurusamy. 2024. "Emerging Strategies in Mesenchymal Stem Cell-Based Cardiovascular Therapeutics" Cells 13, no. 10: 855. https://doi.org/10.3390/cells13100855

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop