Skip to main content

Genetically modified mesenchymal stromal cells: a cell-based therapy offering more efficient repair after myocardial infarction

Abstract

Myocardial infarction (MI) is a serious complication of coronary artery disease. This condition is common worldwide and has a profound impact on patients' lives and quality of life. Despite significant advances in the treatment of heart disease in modern medicine, the efficient treatment of MI still faces a number of challenges. Problems such as scar formation and loss of myocardial function after a heart attack still limit patients' recovery. Therefore, the search for a new therapeutic tool that can promote repair and regeneration of myocardial tissue has become crucial. In this context, mesenchymal stromal cells (MSCs) have attracted much attention as a potential therapeutic tool. MSCs are a class of adult stem cells with multidirectional differentiation potential, derived from bone marrow, fat, placenta and other tissues, and possessing properties such as self-renewal and immunomodulation. The application of MSCs may provide a new direction for the treatment of MI. These stem cells have the potential to differentiate into cardiomyocytes and vascular endothelial cells in damaged tissue and to repair and protect myocardial tissue through anti-inflammatory, anti-fibrotic and pro-neovascularization mechanisms. However, the clinical results of MSCs transplantation for the treatment of MI are less satisfactory due to the limitations of the native function of MSCs. Genetic modification has overcome problems such as the low survival rate of transplanted MSCs in vivo and enhanced their functions of promoting neovascularization and differentiation into cardiomyocytes, paving the way for them to become an effective tool for repair therapy after MI. In previous studies, MSCs have shown some therapeutic potential in experimental animals and preliminary clinical trials. This review aims to provide readers with a comprehensive and in-depth understanding to promote the wider application of engineering MSCs in the field of MI therapy, offering new hope for recovery and improved survival of cardiac patients.

Introduction

Cardiovascular disease (CVD) is a major health threat worldwide, and Myocardial infarction (MI) is included in this group of diseases characterized by high morbidity and mortality [1]. MI is pathologically characterized by the death of myocardial cells resulting from prolonged ischemia of the myocardium [2]. If left untreated, it will lead to irreversible myocardial loss, scar formation or even replacement by fibrotic tissue, ultimately resulting in heart failure [3,4,5,6]. Current clinical treatments, including pharmacological thrombolysis, percutaneous coronary intervention, and coronary artery bypass grafting, have significantly improved symptom relief and patients' quality of life. However, these treatments still face limitations when it comes to regenerating and repairing myocardial tissue [7,8,9].

Research on mesenchymal stromal cells (MSCs) transplantation for the treatment of MI has been conducted for several years, and its role in myocardial protection, immunomodulation, and promotion of angiogenesis has been gradually confirmed, showing a broad therapeutic perspectives [10,11,12]. Owing to unfavorable conditions within infarcted myocardium that significantly compromise survival rates of transplanted cells, primary strategies to enhance therapeutic efficacy include pre-treatment of transplanted cells, optimization of myocardial microenvironment, and integration of genetic engineering with tissue engineering, among other approaches [13,14,15,16,17]. Among them, the optimization method based on genetically engineering cells has a number of advantages, for example, MSCs come from a wide range of sources, which are easy to obtain and expand, and do not involve the controversy of ethics and morality; and by modulating the expression of specific genes, the ability of MSCs to promote angiogenesis and anti-apoptosis can be improved, which significantly improves the transplantation effect [18,19,20].

This review consolidates recent advancements and future prospects of genetically modified MSCs-based therapies for MI treatment, while also examining the mechanisms and effects associated with transplantation of genetically modified MSCs in addressing this condition. Finally, the current challenges and strategies are discussed to provide reference and guidance for further advancing research and clinical applications in this field. Based on these opportunities and challenges, the prospect of clinical application of genetically modified MSCs transplantation for MI remains promising. Through continued in-depth research and constant innovation, it is believed that this treatment will bring better quality of life and therapeutic efficacy to patients with MI.

The pathophysiological process of MI and treatment

The most common type of MI is type I MI, defined as MI caused by atherosclerotic thrombotic CVD [21]. There are many risk factors that can trigger MI (Fig. 1). Initially, damage occurs to the endothelium of coronary arteries, often resulting from narrowing or rupture associated with atherosclerosis [22]. In the damaged area, platelets aggregate and form a thrombus, leading to a blockage of the coronary artery, which may be partial or complete, thus preventing blood flow to the myocardium leading to myocardial ischemia [23]. At this point, there is an inadequate supply of oxygen and energy to the cardiac muscle cells, and under ischemic conditions, these cells begin to suffer damage. Ischemia causes loss of function and structural damage to cardiomyocytes. Metabolic processes within the cells are disrupted and cell membrane permeability is increased, leading to disturbances in the exchange of substances between the inside and outside of the cell [24]. At this point, the cells may experience reversible damage, but if ischemia continues, irreversible damage develops. If ischemia persists for a longer period of time, irreversible cellular damage leads to cardiomyocyte death and the formation of an infarct zone [25]. Size and location of the infarct zone are determined by the extent and site of coronary artery obstruction. Dead cardiomyocytes release intracellular components that initiate an inflammatory response, attracting immune cells to the infarct zone, which subsequently clear away dead cells and tissue debris [26]. Over time, the inflammation subsides and collagen fibers are gradually deposited, forming scar tissue that replaces the dead cardiomyocytes [27]. This process may lead to a permanent weakening of myocardial function, affecting the heart's ability to pump blood.

Fig. 1
figure 1

Major risk factors for MI include an irrational diet, which can lead to high blood cholesterol and high blood pressure. Extreme emotions, including overexertion and sadness, can cause rapid short-term ischemia of the heart muscle, while overexertion can deprive the heart of oxygen by prolonged heavy work. Smoking is strongly associated with thrombosis, and alcohol abuse increases oxygen consumption by the heart muscle through increased heart rate and blood pressure

The main goals of conventional drug therapy are to relieve symptoms, improve heart function, and prevent reinfarction. These include anticoagulants, antiplatelet agents, and beta-receptor antagonists [28]. Although these treatments can help patients maintain their lives and slow the progression of the disease to some extent, they cannot reverse the tissue damage and scarring caused by a heart attack to the extent that normal heart function cannot be restored and patients may continue to experience problems such as heart failure. Conventional treatments have not been effective in stopping the inflammatory response that interferes with the repair process in the infarcted area.

Many basic studies have been conducted on MSCs-based repair therapy after MI, and most of the preclinical studies have achieved good results. However, recent clinical trials have unveiled several drawbacks associated with original MSCs, revealing that their clinical efficacy is constrained by inherent native functions and falls short of anticipated levels. Based on this status quo, it is very difficult to meet the approval criteria for patented drugs and to advance the clinical translation of MSCs.

Biological properties of MSCs and MI

MSCs are adult stem cells whose biological properties make them a potential tool for repair therapy after MI [29, 30]. They are derived from a variety of tissues, such as bone marrow, adipose tissue, umbilical cord, and placenta, encompassing a wide range of sources [31]. This diversity of sources provides flexibility for clinical applications, allowing MSCs to be derived from the patient's own or other suitable sources [32]. The types of MSCs commonly investigated in human clinical trials are bone marrow-derived MSCs (BM-MSCs), adipose-derived MSCs (AD-MSCs), and umbilical cord-derived MSCs (UC-MSCs) [33]. Of these, the main ones used for post-MI cardiac repair studies are BM-MSCs and UC-MSCs. Subgroup analyses of cell sources showed that UC-MSCs improved ejection fraction of heart more significantly than BM-MSCs and AD-MSCs, indicating MSCs derived various tissues had tissue heterogeneity in therapeutic outcomes [34].

MSCs are characterized by their multidirectional differentiation potential and self-renewal capacity [35]. The multidirectional differentiation potential of MSCs allows them to differentiate into various cell types, such as cardiomyocytes and vascular endothelial cells. This characteristic is anticipated to facilitate the process of myocardial repair following MI [36,37,38]. MSCs not only excel in their differentiation potential, but their immunomodulatory capacity is also a distinguishing feature of their biological properties [39]. MSCs have the capability to enhance inflammation when the immune system is not sufficiently active, and they can also inhibit inflammation when the immune system is overactive, in order to prevent auto-aggression [40]. This makes MSCs a powerful tool in the treatment of autoimmune and inflammatory diseases [41]. The modulation of the inflammatory microenvironment is an important therapeutic target in post MI repair therapy, and MSCs can reduce further myocardial damage by immunomodulation through paracrine effects and by acting on immune cells [11, 36, 42].

The natural repair and regeneration capabilities of MSCs provide a solid foundation for genetic modification (Fig. 2) [18]. Firstly, they can differentiate into cardiomyocytes to fill in the damaged areas [43]. Secondly, MSCs promote repair of myocardial infarcted areas by secreting growth factors such as vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1), and immunomodulatory factors in order to act as a promoter of angiogenesis, modulator of inflammatory responses, and stimulator of matrix remodelling [44]. In addition, MSCs exhibit anti-apoptotic and anti-fibrotic properties, which are important for tissue repair after MI [45]. By inhibiting apoptosis, MSCs help maintain the survival of cardiomyocytes and slow down the process of tissue damage [46]. Meanwhile, inhibition of fibrosis helps to reduce scar tissue formation and inhibit adverse remodelling [47].

Fig. 2
figure 2

Sources of MSCs and their roles in repair therapy after MI

Genetic engineering-based transformation of MSCs

The current challenges for MSCs cardiac therapies are to improve their survival and engraftment in injured myocardium and to enhance their ability to promote cardiac regeneration [48]. A variety of approaches have been used to enhance the function of naive MSCs to suit therapeutic needs (Table 1).

Table 1 Methods used to enhance the function of MSCs and their therapeutic effects

In recent years, genetic engineering, a cutting-edge field at the forefront of scientific and technological progress, has provided an innovative approach to repair therapy after MI [18]. The essence of this approach resides in regulating MSCs at the molecular level via genetic manipulation, which enhances cellular function and improves therapeutic outcomes in MI [56]. Genetic engineering techniques used to modify MSCs include gene editing and transgenic techniques (Table 2). In the modification of MSCs, guide RNAs can be designed to achieve precise editing of target genes by gene editing techniques, leading to gene knockout, modification or substitution. Transgenic technology, on the other hand, uses vectors such as plasmids and viruses to deliver target genes into the interior of MSCs and ensure stable delivery of exogenous genes so that MSCs can constantly express or inhibit specific proteins under specific conditions to achieve precise regulation of cellular functions, and vectors carrying specific drugs can also be introduced into MSCs so that they can become drug carriers and achieve targeted release of therapeutic substances [57]. In addition, reverse transcription gene technology can be used to introduce specific genes to enhance myocardial tissue repair and regeneration by MSCs. Lentiviruses, retroviruses, adeno-associated viruses (AAVs), and adenoviruses can all be used as viral vectors for genetically modified MSCs [57]. Small interfering RNA (SiRNA) gene silencing and microRNA (miRNA) are also common genetic engineering techniques, where miRNA plays an important role in gene modification of MSCs by post-transcriptional inhibition of the target messenger RNA (mRNA) to alter gene expression [58].

Table 2 Current basic research on gene-modified cell therapy for MI

Improving survival of grafted cells

The myocardial lesion areas of MI patients often exhibit a complex state of ischemia, hypoxia and chronic inflammation, and this poor host microenvironment is highly susceptible to induce the expression of apoptosis-related genes in transplanted MSCs, thereby adversely affecting their survival [73]. Secondly, the biological effects of MSCs from different individual sources vary widely, and MSCs from older donors tend to be more susceptible to apoptosis as a result of senescence and apoptosis due to the downregulation of biological functions caused by aging [74,75,76]. An increasing number of studies have shown that the survival of transplanted MSCs in infarcted myocardium can be effectively improved by genetically engineering MSCs.

As early as 2003, Mangi et al. used isolated retroviral transduction of rat MSCs to overexpress the proto-survival gene protein kinase Bα (Akt1), significantly increased survival of MSCs after transplantation into rat ischemic myocardium, with inhibition of cardiac remodeling and near restoration of normal function [77]. Subsequent studies further confirmed this effect and suggested that these beneficial effects may be due to paracrine effects of MSCs, although the exact mechanism remained to be investigated [78]. Akt upregulation by stromal cell-derived Factor-1alpha (SDF-1α) prevented apoptosis and MSCs transduced by lentiviral vector have reduced apoptosis and increased survival after injection into infarcted myocardium [79]. Deng et al. also demonstrated that overexpression of cellular repressor of E1A-stimulated genes activated Akt to down-regulate the expression of p53, and the transcriptional activation of p53 led to the expression of pro-apoptotic proteins including BCL2-Associated X (Bax) and cell death proteins, and thus effectively inhibited apoptosis in MSCs under conditions of hypoxia and serum deprivation in vitro [80]. To further improve the survival of MSCs, Chen et al. transduced MSCs using isolated AAVs to overexpress Akt1 and Wnt Family Member 11 (Wnt11), and found that the transplanted cells showed reduced apoptosis and increased survival, as well as greater cardiac differentiation and survival potential [81]. During this period, a team transfected BM-MSCs with the fibroblast growth factor-2 (FGF-2) gene and found that their secretion of FGF-2 increased under hypoxic conditions, their survival was significantly improved and the expression of the anti-apoptotic gene B-cell lymphoma/leukemia 2 was increased [82]. Secreted Frizzled-related protein 2 (sFRP2) was thought to be a key mediator of MSCs-mediated myocardial repair, and overexpression of sFRP2 enhanced the resistance to apoptosis as well as increased the implantation rate of MSCs, and these effects were achieved by inhibiting the Wnt and Bone morphogenic protein signaling pathways [83]. Overexpression of mid-range factor (MK) inhibited MSCs apoptosis induced by hypoxia and glucose deprivation in vitro, and implantation of MSCs overexpressing MK into the hearts of myocardial infarcted rats was observed to result in an increase in survival, as well as a reduction in infarct size and an improvement in cardiac function [84]. GATA binding protein 4 (GATA-4) played a regulatory role in the differentiation, growth and survival of a wide range of cell types, and Li et al. found that BM-MSCs overexpressing GATA-4 showed increased survival in ischemic myocardium, an effect that may be attributed to the anti-apoptotic effect of GATA-4 in addition to the upregulation of insulin-like growth factor-1 (IGF-1) levels in MSCs [85]. B-cell Lymphoma-extra-large (Bcl-xL) was an important factor in the regulation of apoptosis and belongs to the B-cell lymphoma-2 (Bcl-2) family of proteins, and overexpression of Bcl-xL in rat MSCs by gene modification improved the implantation rate of MSCs as well as their survival in the ischemic and hypoxic microenvironment, in addition to enhancing the pro-angiogenic capacity of MSCs [86]. Heme oxygenase-1 (HO-1) was an anti-apoptotic and anti-inflammatory enzyme, and Zeng et al. used recombinant adenoviruses to transfect MSCs and found that they improved the survival of MSCs in the ischemic myocardium of infarcted rats by improving the mRNA levels of inflammatory cytokines and reducing the levels of the pro-apoptotic protein Bax, mainly through the release of paracrine factors that bind to HO-1 protein [87]. In response to the upregulation of specific chemokines in infarcted myocardium and the very low expression of corresponding chemokine receptors in MSCs, Huang et al. genetically engineered MSCs to overexpress Chemokine (c–c motif) receptor 1 and observed a reduction in the number of apoptotic cardiomyocytes and infarcted area after transplantation into ischemic myocardium [88]. Integrin-linked kinase (ILK) was an important pleiotropic protein that regulated cell survival, proliferation, differentiation and angiogenesis. Mu et al. injected ILK-transfected MSCs into myocardial infarcted pigs and found that MSCs homed to the infarcted myocardium was significantly enhanced and apoptosis was effectively reduced [89]. The LIM homology cassette transcription factor islet-1 played a crucial role in the development of cardiac embryos and adult resident cardiac stem cells. Xiang et al. found that ISL1 overexpressed human mesenchymal stromal cells (HMSCs) enhanced hMSCs survival and paracrine function in hMSCs transplanted into myocardial infarcted rats, and that this effect may be mediated through insulin-like growth factor binding protein 3 [60]. Follicle suppressor-like 1 (Fstl 1) has been described as a novel cardiomyocyte survival-promoting factor. Shen et al. found that transduction of MSCs with Fstl 1 recombinant lentiviruses attenuated myocardial extracellular matrix (ECM) deposition and inflammatory cell infiltration in ischemic hearts and improved the survival of transplanted cells [90]. Irisin has anti-apoptotic, increased cell viability and antioxidative stress cardioprotective effects and was the extracellular domain of Fibronectin type III domain-containing protein 5 (FNDC5), and overexpression of FNDC5 by genetic modification of BM-MSCs prevented hypoxia-induced increased in apoptosis and consequently improved MSCs survival after transplantation [67]. Octamer-binding protein 4 (Oct4) can promote the release of cytoprotective factors such as Basic fibroblast growth factor (BFGF), survivin and Bcl-2, and can reduce the expression of apoptosis-associated proteins Bax and caspase-3, and Huang et al. found that β-catenin was an upstream regulatory target that can be overexpressed to maintain the survival of transplanted MSCs [91]. Fas apoptosis inhibitory molecule (FAIM) was involved in the regulation of exogenous apoptotic pathways, Chen et al. found that FAIM-overexpressed MSCs improved the survival of transplanted MSCs in myocardial infarcted mice by inhibiting the C-Jun N-terminal kinase-mediated ubiquitinated proteasome-dependent Cellular-FLICE inhibitory protein degradation pathway [59].

Based on the fact that overexpression of anti-apoptotic genes can improve the survival rate of MSCs, it was suggested that silencing of pro-apoptotic genes might have the same effect. Therefore, a recombinant human caspase-8 small hairpin RNA adenovirus was prepared to transfect MSCs and transplanted into the ischemic myocardium of infarcted rats, and the results showed that it significantly reduced the activity and expression of caspase-8 in the border zone of the infarcted myocardium and also increased the survival rate of the transplanted MSCs [92]. Apoptosis signal-regulated kinase 1 (ASK1) has been implicated in the development of oxidative stress-associated pathologies in the injured heart, and Lee et al. found that hypoxia-induced activation of ASK1 signaling could be attenuated by microRNA-301a overexpression in human adipose-derived stem cells to inhibit apoptosis and improve the survival of transplanted stem cells [93]. Chen et al. found that after injection of BMSCs overexpressing microRNA-133 into the hearts of rats with MI, apoptosis of MSCs in hypoxic conditions was significantly reduced, inflammation and fibrosis in the infarcted heart were suppressed, and cardiac function was improved [94]. Mitochondrial integrity played an important role in the anti-apoptotic process of hMSCs, which prevented the release of various pro-apoptotic factors, and regulation of the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway helped to maintain the integrity and function of mitochondria [80]. Leptin conferred mitochondrial integrity to hMSCs by enhancing the accumulation of Optic atrophy 1, so that leptin overexpression enhanced the survival of MSCs in an ischemic environment [95]. Ischemia and hypoxia usually led to mitochondrial dysfunction, which in turn leads to apoptosis, and it was found that ELABELA regulated Hypoxic/ischemic-induced mitochondrial dysfunction through apelin receptor, thereby improving the anti-apoptotic capacity and increasing the viability of MSCs [96].

Senescence has become a prominent area of investigation, where mesenchymal stromal cells were observed to transition into a senescent state due to factors such as telomere dysfunction, genomic lesions, chromatin structural irregularities, and the engagement of active mitotic signaling pathways [75, 97]. Concurrently, these cells face an inevitable diminishment in their immunoregulatory functions [98]. In addition to this, the proliferative, differentiation, and migratory capabilities of MSCs also diminished accordingly [99, 100]. Consequently, this adversely affected the survival rate and reparative capabilities of the transplanted cells [101]. Sirtuin 3 (SIRT3) was localized in mitochondria and was associated with oxidative stress and longevity, and overexpression of SIRT3 enhanced the antioxidant capacity of Old hMSCs as a mean of increasing the number of survivors after transplantation into damaged myocardium [102]. MicroRNA-10a (MiR-10a) was significantly reduced in O-hBM-MSCs, and overexpression of miR-10a increased survival of transplanted hBM-MSCs in infarcted mouse hearts by activating AKT [103]. Erb-B2 receptor tyrosine kinase 4 (ERBB4)-overexpressing aged MSCs were more resistant to oxidative stress-induced cell death, and the increased expression of their phosphorylated AKT and extracellular signal-regulated kinase (ERK) under hypoxic conditions effectively reduced apoptosis [104]. Senescent BM-MSCs have reduced autophagy levels and are less tolerant to hypoxia, so their apoptosis rate was higher under hypoxic conditions compared to the younger group. Yang et al. found that knocking down IGF-1 increased their autophagy levels, which facilitated their survival when transplanted into infarcted myocardium [105]. In conclusion, modification of MSCs by genetic engineering is a reliable way to increase their survival rate. Inverse regulation of N6-methyladenosine by ALKB homologue 5 (ALKBH5) promoted cellular senescence, so down-regulation of ALKBH5 rejuvenated aged MSCs and increased their survival in infarcted hearts [55].

Promoting angiogenesis

Myocardial perfusion was closely related to the survival of cardiomyocytes. Therefore, hemodialysis after MI played an important role in the recovery of cardiac function, and previous studies have shown that MSCs can differentiate into vascular endothelial cells for angiogenesis [106, 107].

VEGF and angiopoietin-1 (Ang1) played a therapeutic role by promoting angiogenesis, but their therapeutic effects in a mouse model of Acute myocardial infarction (AMI) were found to be weaker than in a group of transplanted MSCs [108]. One team suggested transfecting MSCs with VEGF to improve the therapeutic effect, and found that transplanting them into damaged hearts not only promoted angiogenesis, but also helped to improve post-infarction cardiac remodeling and cardiac systolic and diastolic function [109]. In the hearts of rats with MI treated with HO-1-MSCs, the expression level of VEGF was more significantly enhanced and the density of micro vessels was significantly increased [87]. In addition, autologous MSCs transplanted with angiopoietin gene transfection showed increased neointima formation and synergistic effects on cardiac perfusion and functional improvement in a porcine model of chronic ischemia [110]. Based on the fact that MSCs overexpressing the Akt gene showed improved survival and engraftment in infarcted myocardium, Jiang et al. transduced MSCs co-overexpressing Ang-1 and Akt to enhance the viability of remaining cardiomyocytes and transplanted cells by promoting angiogenesis [111]. Subsequent studies have demonstrated its long-term stable efficacy [112]. Zhang et al. to maximized the effect of SDF-1α on cell migration and regeneration, adenoviral transduction of male rat MSCs to overexpress C-X-C motif chemokine receptor type 4 (CXCR4) was transplanted into myocardial infarcted rats and found to be significantly increased in infarcted areas and peri-infarcts, effectively promoted angiogenesis and alleviated early signs of left ventricular remodelling [113]. Zhao et al. injected lentiviral engineered MSCs into the myocardium for sustained release of SDF-1α to complement transient endogenous cytokine release for enhanced angiogenesis [79]. BM-MSCs overexpressing GATA-4 also promoted angiogenesis in ischemic myocardium through paracrine effects, and several growth factors, including Vascular endothelial growth factor-A (VEGF-A) and IGF-1, may be involved [85]. Glycogen synthase kinase (GSK)-3 was a serine/threonine kinase that regulated a variety of intracellular functions by phosphorylating intracellular substrates such as glycogen synthase and GATA4, and VEGF-A was found to be upregulated in the hearts of mice injected with GSK-3β—bone marrow-derived MSCs in the study by Cho et al. It was therefore hypothesized that it was most likely through this pathway that angiogenesis was promoted [114]. The pro-angiogenic chemokine granulocyte chemotactic protein-2 (GCP-2) played a key role in angiogenesis, Kim et al. transplanted GCP-2 overexpressing human AD-MSCs into myocardial infarcted mice and found that the expression of pro-angiogenic factors, such as VEGF-A and hepatocyte growth factor (HGF), was higher, and that chemokines, such as interleukin (IL)-8 and GCP-2, were also significantly upregulated [19]. Intramyocardial injection of HGF-transfected BM-MSCs into the myocardium of a rat model of MI stimulated angiogenesis and reduced myocardial fibrosis [115]. Growth differentiation factor 11 promoted stem cell activity and leaded to increased survival and retention in infarcted hearts after transfection of MSCs, favoring enhanced angiogenesis [116].

Considering the effect of aging on the pro-angiogenic function of MSCs, Liu et al. ameliorated the aging phenotype of aged MSCs by overexpressing silent mating type information regulation 2 homolog 1 to recapitulated their pro-angiogenic properties [117]. Thioredoxin-1 (Trx1) has been shown to be a potent antioxidant, transcription factor and growth factor regulator, and Suresh et al. found a pro-angiogenic effect in the form of increased capillary density and better maintenance of survival and differentiation after transplantation of MSCs transfected with Trx1 into rats with MI [118]. Overexpression of miR-10a in aged BM-MSCs also increased VEGF and SDF secretion, thereby promoting angiogenesis in ischemic mouse hearts [103]. In contrast, ERBB4-age MSCs promoted angiogenesis by secreting higher levels of angiopoietin, epithelial neutrophil-activating peptide 78, VEGF and fibroblast growth factor 2 [104]. MicroRNA-335 (MiR-335) caused senescence in MSCs through its regulatory effect on mitochondrial dynamics. Hong et al. inhibited miR-335 expression in MSCs by genetic modification and enhanced their angiogenesis in a mouse model of aging MI [69].

Although IGF-1 and HGF had the potential to promote cardiac repair and MSCs overexpressing either IGF-1 or HGF enhanced neointima formation in a porcine model of AMI, simultaneous administration did not appear to exert a synergistic effect [119]. Whereas very small embryonic-like MSCs co-overexpressing hypoxia-inducible factor 2-alpha and Oct4 synergistically enhanced their angiogenesis [120]. Overexpression of β-conjugated protein-treated MSCs optimally upregulated Oct4 in ischemic cardiomyocytes and strongly contributed to the secretion of the pro-angiogenic cytokines Ang1, BFGF, HGF and VEGF [91]. Overexpression of c-Myc favored angiogenesis in cardiac-resident MSCs, but inevitably induced an inflammatory response, which was compensated by its co-expression with Oct4, resulted in a synergistic effect of enhanced angiogenesis [121].

Promoting myocardial regeneration

Previous studies have shown that MSCs can differentiate into cardiomyocytes to participate in the repair of damaged myocardium due to their multidirectional differentiation potential [122,123,124].

Grauss et al. found early on that forced expression of cardiac myosin by genetic modification of hMSCs prior to transplantation could increase their tendency to differentiate into cardiomyocyte-like cells in mice with MI [125]. 5-Bromo-4-chloro-3-indolyl β-D-galactopyranoside-positive cells and upregulation of troponin T were observed after injection of MSCs overexpressing GSK-3β into mice with MI, providing strong evidence that overexpression of GSK-3β promoted the differentiation of MSCs into cardiomyocytes [114]. Compound 56 was an epidermal growth factor receptor inhibitor that promoted cardiogenic differentiation of hMSCs. MiRNA-133a also targeted EGFR to induce cardiogenic differentiation of hMSCs, and the expression of endogenous miRNA-133a was regulated by compound 56. Therefore, EGFR can be targeted by transfection of miRNA-133a to promote the differentiation of hMSCs into cardiomyocytes [126]. Co-overexpression of Akt1 and Wnt11 significantly upregulated the expression of cardiac markers Natural killer type-2 transcription factor related 5, GATA-binding protein 4, α-myosin major histocompatibility complex and brain natriuretic protein, thus contributing to myocardial differentiation in MSCs. Therefore, their promoting effect on myocardial differentiation in MSCs should not be underestimated [81]. T-Box Transcription Factor 20 (Tbx20) belonged to the T-box family of transcription factors involved in the differentiation process that regulated cardiomyocyte differentiation and may played a role in upregulating gene expression in cardiac development and cardiomyocyte homeostasis in conjunction with a variety of other cardiac transcription factors. Human AD-MSCs transduced with Tbx20 lentiviral vectors induced the expression of markers of myocardial differentiation by increasing the expression of cardiomyocyte differentiation markers at both the RNA and protein levels [127]. Pygopus Family PHD Finger 2 (PYGO2) was a core component of the canonical Wnt signaling pathway, and transfection with PYGO2 contributed to canonical Wnt signaling to promote the early differentiation of human umbilical cord-derived mesenchymal stromal cells (hUC-MSCs) into cardiomyocytes, and promoted the mid- to late-stage differentiation of MSCs into cardiomyocytes through activation of the PI3K-Akt signaling pathway [128].

Combining multiple approaches to optimize the biological functions of MSCs to enhance therapeutic efficacy after MI

In recent years, more and more optimization methods have gradually emerged based on genetic engineering to optimize the biological functions of MSCs. This included the combination of adjuvant therapies such as growth factors and anti-inflammatory drugs, which can promote cell growth and reduce the inflammatory response [129]. The use of biomaterials, such as suitable scaffolds and bio-colloids, to provide support and guidance for MSCs can promote cell colonization and differentiation in damaged areas [130]. The use of nanotechnology to achieve the targeted release of therapeutic agents and to improve the spatial and temporal precision of the therapeutic effect.

Medication-assisted treatment

Statins were known to protect endothelial function, inhibit inflammation and stabilize atherosclerotic plaques, among which rosuvastatin may exerted a protective effect on AD-MSCs by modulating FoxO3a induced by the PI3K/Akt and Mitogen-activated extracellular signal-regulated kinase (MEK)/ERK1/2 pathways, so that when combined with AD-MSCs, it enhanced implanted survival and proliferation and reduced myocardial fibrosis [131]. In contrast, combined treatment with atorvastatin increased SDF-1 expression in peri-infarct myocardium while suppressing inflammation [132]. NO production associated with inducible nitric oxide synthase (iNOS) overexpression impaired BMSCs transplanted into infarcted rat myocardium, ameliorated by combined use of selective iNOS inhibitors [133]. Exendin-4 modulated the PI3K/Akt-sfrp 2 pathway to reduce mitochondrial oxidative stress damage, so that AD-MSCs pretreated with exendin-4 can exert better anti-apoptotic effects after transplantation into infarcted myocardium [134]. 5-Diphosphoinositol pentakisphosphate (IP7) physiologically inhibited the production of Akt, which is generated by inositol hexakisphosphate kinases (IP6Ks); Therefore, N2-(m-trifluorobenzyl)-N6-(p-nitrobenzyl) purine (TNP), as a reversible inhibitor of IP6Ks, promoted the implantation of MSCs in the infarcted heart by inhibiting IP6Ks to downregulate the production of IP7 and enhance the activation of Akt promoted the implantation of MSCs in the infarcted heart and paracrine effects that contributed to the protection of myocardial function after MI [135]. Edaravone, a free radical scavenger that inhibited the production of reactive oxygen species (ROS), in combination with BM-MSCs attenuated hypoxia-induced apoptosis by inhibiting the accumulation of intracellular ROS and prolonged the activation of the Akt pathway to promote the paracrine potential of BM-MSCs, which promoted angiogenesis and cardiac stem cell-mediated regeneration of the cardiac muscle [136]. Traditional Chinese medicine promoted stem cell migration and survival by improving the microenvironment of the infarcted myocardium, in which Guanxin Danshen preparation, as the main prescription for clinical treatment of ischemic heart disease, can reduce apoptosis of the transplanted cells when administered in combination with MSCs and reduce angiogenesis in the infarcted area and the periphery, thus improving cardiac function after MI [137].

Biomaterials

CXCR4-overexpressing MSCs promoted neo angiogenesis in infarcted myocardium, Huang et al. designed a stem cell patch using peritoneum as a substrate, which in combination with CXCR4-overexpressing MSCs synergistically created a suitable environment to promote angiogenesis and cardiomyocyte survival, and reduced myocardial remodeling through paracrine effects [138]. Arginyl-glycyl-aspartic acid (RGD)-modified alginate microspheres encapsulate MSCs and, when injected into infarcted rat myocardium, provided myocardial structural support to prevent adverse post-MI remodeling while inducing neovascularization [139]. Collagen I was a major component of the ECM of the heart, and mixing MSCs with type I collagen solution to distribute them in collagen scaffolds and to make three-dimensional (3D) patches, their transplantation improved myocardial function by promoting reverse remodeling of the infarcted area [140]. Poly/gelatin nanofibrous patch can provide adequate mechanical support for MSCs, improved cellular interactions required for infarct repair, attenuates left ventricular remodeling and induced angiogenesis and cardiomyopoiesis in rat model of MI [141]. Hyaluronic acid (HA) was involved in many cellular processes, including proliferation and differentiation, and the use of hyaluronic acid-based hydrogel scaffolds as a vehicle for the delivery of MSCs may improve infarcted myocardial structure and function by promoting cell survival after transplantation, reducing inflammatory responses and increasing neointimal formation [142]. VEGF-loaded gelatin and alginate polyelectrolytes encapsulate MSCs by layer-by-layer self-assembly technology, which was a novel targeted delivery vehicle that can be delivered to myocardial infarcted tissues by virtue of the homing effect of MSCs and release VEGF continuously, thus promoting angiogenesis in infarcted areas and increasing perfusion to improve cardiac function [143]. Chitosan hydrogel was a slow-release carrier that protected cells from the host immune system clearance response and can effectively increase the viability of transplanted cells in the infarcted myocardium. IGF-1 had pro-proliferative, anti-apoptotic and angiogenesis-inducing effects, so chitosan hydrogel modified by the C structural domain of IGF-1 can provide a good microenvironment for the survival of human placenta-derived mesenchymal stromal cells and enhance their ability to promote angiogenesis, anti-apoptosis and inhibit fibrosis [144]. Cell sheet was a bioactive material that can prolong the retention and survival time of transplanted MSCs and improve the transplantation microenvironment. Transplanted hUC-MSCs cell sheets modulated inflammatory responses in infarcted myocardium and inhibited ventricular remodeling through paracrine effects. Additionally, the ECM left behind after UC-MSCs' demise served as a bioactive scaffold for host cells, fostering angiogenesis in infarcted regions. Nerve growth factor primarily facilitated the proliferation and survival of MSCs [145]. Porous microspheres produced based on Polyethylenimine1.8 k and poly (lactic-co-glycolic acid) particles exhibited low toxicity, provided a large inoculum surface area for MSCs, and increased the oxygen and nutrient support required for their growth, thereby greatly enhanced the regenerative potential of MSCs after transplantation into infarcted myocardium [146]. Hydrogel systems based on gelatin methacrylate and oxidized dextran in combination with reductive graphene oxide exhibited conductivity similar to that of natural cardiac tissue, UC-MSCs encapsulated by them were well morphologically shaped and sustained release was achieved, and the efficiency of differentiation of UC-MSCs was improved with the up-regulation of Cardiac Troponin I and Connexin 43 expression [147].

Nanotechnology

Nanocarriers as an siRNA delivery strategy were less toxic and safer than viral vectors, where poly (amidoamine) dendritic polymers with precisely controlled radial symmetry and high cargo capacity in a nanoscale volume combined with the advantage of arginine residues to facilitated cell membrane penetration, and the combination of both of them can provide a highly efficient and biocompatible delivery system for siRNA-mediated silencing of the prolyl hydroxylase structural domain protein 2 in MSCs to improve the survival rate and paracrine effect of MSCs after transplantation [148].

Hollow mesoporous organosilicon nanoparticles (HMONs), after surface optimization with polyethyleneimine (PEI), had achieved enhanced gene loading capacity and protection, avoided the drawbacks of toxicity and adverse immune responses that may be associated with traditional strategies. Using HMONs-PEI as a gene carrier, HGF can be effectively transfected into BM-MSCs without affecting their migration and proliferation, and can induce enhanced expression of infarcted myocardial HGF protein after transplantation. The use of HMONs-PEI as a gene carrier can effectively transfect HGF into BM-MSCs without affecting their migration and proliferation, and can induce enhanced expression of HGF protein in infarcted myocardium after transplantation, which improved therapeutic efficacy by decreasing apoptosis and increasing angiogenesis [149]. Patch made of electro spun cellulose nanofibers can improve the post-infarction myocardial microenvironment by mimicking the heart's natural ECM and can provide a 3D scaffold for implanted AD-MSCs. Combined with the non-toxic, biodegradable, and anti-inflammatory features of chitosan/silk proteins (CS/SF), the biocompatibility of the nanofiber patch made of CS/SF assembled on electro spun cellulose nanofibers by layer-by-layer (LBL) technology was optimized. The biocompatibility of the nanofibrous patch made of CS/SF assembled onto electro spun cellulose nano patches by LBL technology was optimized, thus increasing the survival of AD-MSCs transplanted into infarcted myocardium and effectively improving ventricular remodelling [150]. Self-assembling peptide can form a hydrogel scaffold to provide a favorable microenvironment for stem cell attachment, growth and survival under specific conditions, and after modification with functional motifs glutamine-histidine-arginine-glutamic acid-aspartic acid-glycine-serine (QHREDGS) to obtain the designer self-assembling peptide (DSAP) cellular delivery system, the paracrine effect of MSCs inoculated with DSAP was up-regulated after transplantation into the infarcted myocardium, which resulted in an increase in graft cell survival and angiogenesis [151].

Clinical research and challenges

Previous studies have shown that trans endocardial injections of MSCs were safe (Table 3) [152]. Although treated patients showed transient small increases in myocardial biomarkers (Creatine Kinase Isoenzyme-MB and serum troponin I), there was a modest reduction in the size and mass of myocardial scar tissue, with no serious adverse effects of postoperative pericardial effusion and an improvement in quality of life [153]. Another study also found that scar tissue reduction and functional improvement occurred predominantly in the actual injected area, and provided strong evidence that 20 million injections of autologous MSCs produced the greatest relative gains [154]. In a subsequent clinical trial, the researchers found that the 100 million-dose injections produced superior results, with increased cardiac ejection fraction in addition to reduced scar size, due to the smaller sample size per dose and random allocation of patients to autologous or allogeneic hMSCs transplantation, as well as the effect of disease severity on cell dose [155]. During clinical trials, Gao et al. found that the function and viability of BM-MSCs declined with age and could not meet the therapeutic needs of MI patients in a timely manner, making it necessary to seek an alternative source of stem cells [156]. In a subsequent double-blind randomized controlled trial, Wharton's glial-derived MSCs showed no immune response or tumor formation in allografts and demonstrated enhanced cardiovascular differentiation, effectively improving cardiac function and preventing left ventricular remodelling [157]. Since then, Attar's team conducted a single-blind, randomized, multicenter trial to compare the efficacy of one versus two transplants of Wharton's glial-derived MSCs (WJ-MSCs) after AMI, and found that the left ventricular ejection fraction of the target patients was significantly improved by intracoronary transplantation of WJ-MSCs on days 3–7, and augmented by a booster dose of transplantation 10 days later [158]. Although suitable alternative sources have been identified for the poor efficacy of BM-MSCs after transplantation, in the long term strategies such as combining drugs and engineering were needed to improve the survival and low implantation rates of BM-MSCs in the heart [159].

Table 3 Clinical trial of MSCs for the treatment of myocardial infarction

Current clinical trials of MSCs in the treatment of MI have shown overall safety, but efficacy has not been satisfactory. In addition, there have been no clinical trials of MSC gene modification for specific targets. Therefore, the results of animal experiments that have shown that gene modification of MSCs was more effective in the treatment of MI in basic research should be applied to clinical research in the future to achieve more satisfactory clinical therapeutic effects.

MSCs derivant-exosomes

A growing number of studies have found that MSCs-derived exosomes exert cardioprotective effects similar to those of their parent MSCs, with enhanced safety and therapeutic efficacy [164]. MSCs-derived exosomes were found to exhibit cardioprotective effects similar to those of their parents. By participating in the regulation of intercellular communication, it may play an important role in the treatment of MI as an anti-apoptotic, anti-inflammatory and anti-ventricular remodeling agent [165,166,167]. Exosomes were rich in a variety of biologically active molecules including proteins and miRNAs, among which proteins such as VEGF and MMP stimulated angiogenesis, and factors such as Interleukin-6 (IL-6), Interleukin-10 (IL-10), transforming growth factor-β (TGF-β) and HGF played a favorable role in immunomodulation processes [168]. In addition to this, exosomes attenuated cardiomyocyte damage after MI by inhibiting ferroptosis and mediating polarization processes in macrophages [169, 170]. MicroRNA-136 released from exosomes of young MSCs also downregulated the expression of apoptotic peptidase activating factor in receptor cells as a means to enhance the viability of senescent MSCs and improve their myocardial repair capacity [171].

Various approaches have been used to improve the low yield and low bioavailability of exosomes from MSCs in order to improve their efficacy for post-infarction repair [172]. Pharmacological pretreatment with atorvastatin and Tongxinluo (TXL) improved cardio protection of MSCs exosomes by promoting angiogenesis and other effects [173, 174]. In addition, research on engineered exosomes has also yielded results in that fusion of exosomes with the ischemic myocardium-targeting peptide CSTSMLKAC allowed exosomes to be preferentially targeted to the ischemic myocardium to exert therapeutic effects [175]. Improved stability and sustained release of exosomes using polymer-grafted nanoparticle hydrogels formed by mixing peptide amphiphile-Growth hormone-releasing peptides and naphthalene-conjugated short peptide (NapFF) [176]. Exosomes derived from iron oxide nanoparticles-encapsulated MSCs can be magnetically guided to increase their retention in the infarcted heart and allow the infarcted heart to advance to the repair phase, resulting in a decrease in apoptosis and fibrosis, as well as an enhancement of angiogenesis and recovery of cardiac function [177]. Nanocomplexes constructed by self-assembly of MSCs membranes on the surface of miRNA-containing mesoporous silica nanoparticles mimic exosome function with higher miRNA loading, escaped immunosurveillance, and targeted ischemic myocardium to promote cardiomyocyte proliferation by inhibiting translation of apoptosis-associated proteins [178]. A minimally invasive exosome spray cardiac patch designed by inoculating MSCs-derived exosomes in fibrin scaffolds increased the retention time of exosomes in the infarcted heart and made them more readily available for uptake, effectively reduced apoptosis and facilitates surgical procedures [179]. Injectable conductive hydrogels containing sulfhydryl CP05 peptide can both enhance the retention of hUC-MSCs-derived Exosomes in infarcted myocardium by anchoring it and exert some pro-angiogenic effects [180]. EVs released from MSCs overexpressing hypoxia-inducible factor 1-alpha could promote angiogenesis as well as anti-apoptotic effects by expressing higher levels of microRNA-221-3p, and their survival in the infarcted heart was also improved after wrapping by RGD hydrogel [181]. The short peptide thermosensitive hydrogel developed based on Ang-1 can accelerate the functional recovery of infarcted myocardium by prolonged slow release of ISL1-MSCs-Exosomes allowing it to have a high therapeutic concentration in the injected area [182]. Delivery of exosomes containing microRNA-29b mimics with biocompatible microneedle patches bound to gelatin was a novel attempt to prevent cardiac fibrosis by inhibiting pathological remodeling of the ECM through inhibition of the TGF-β signaling pathway [183]. Electro spun nanofiber cardiac patches made of mixed polycaprolactone and type I collagen can provide both a three-dimensional scaffold for hUC-MSCs exosomes to proliferate and mechanical support for infarcted myocardium, which, combined with the combined effect of TGF-β3 to inhibit myocardial fibrosis, greatly enhanced the therapeutic advantages of exosomes [184]. Bio-conductive polymer hydrogels generated using polypyrrole and chitosan can alleviate arrhythmias by re-synchronizing cardiac electrical transmission, which in combination with the beneficial effects of anti-apoptotic and pro-angiogenic effects of human endometrial mesenchymal stem cell-derived exosomes, provided yet another good method for post-infarction cardiac repair [185]. In conclusion, cell-free therapies based on exosomes of MSCs show broad application prospects in the field of CVD by virtue of their low immunogenicity and regulation of intercellular communication, etc. It was expected that exosomes optimized by different methods will be opened for clinical trials as soon as possible, providing a new option for repair therapy after MI [186].

Conclusion

MSCs played an important role in cardiac repair therapy after MI by virtue of their immunomodulatory ability, vascular and cardiomyocyte differentiation potential, and their efficacy in improving left ventricular ejection fraction, and reducing infarct size. The MSCs-based therapy has been demonstrated to make up for the shortcomings of traditional interventional and pharmacological therapies.

The implantation rate, survival rate, and ability to differentiate into myocardium and vasculature of genetically modified MSCs after transplantation into animal models of MI have been enhanced to a certain extent, and clinical trials have been conducted to screen for appropriate transplantation time, transplantation dose, and source of naive MSCs. The ability of MSCs to be used for cardiac repair after MI was further enhanced with the combination of techniques such as pharmacological intervention, tissue engineering, and nanoengineering. Pharmacological interventions optimized the transplanted myocardial microenvironment for MSCs, tissue processes provided MSCs with a spatial structure conducive to their growth and differentiation, and nano-engineering ameliorated the potential safety concerns of genetic modification by vectors such as viruses.

Future optimization pathways should continue to work on enhancing the survival of MSCs in the harsh microenvironment of infarcted myocardium, and in-depth exploration of the mechanisms of differentiation and migration of transplanted MSCs should be carried out to facilitate clinical translation while ensuring safety, with a view to bringing benefits to MI patients.

Availability of data and materials

Not applicable.

Abbreviations

MI:

Myocardial infarction

MSCs:

Mesenchymal stromal cells

CVD:

Cardiovascular disease

BM-MSCs:

Bone marrow derived mesenchymal stromal cells

AD-MSCs:

Adipose-derived mesenchymal stromal cells

UC-MSCs:

Umbilical cord derived mesenchymal stromal cells

VEGF:

Vascular endothelial growth factor

Ang-1:

Angiopoietin-1

IFN-γ:

Interferon-γ

FGF-2:

Fibroblast growth factor-2

IGF-1:

Insulin-like growth factor-1

BMP-2:

Bone morphogenetic protein-2

CMC:

Cardiomyocyte

TXL:

Tongxinluo

SiRNA:

Small interfering RNA

MiRNA:

MicroRNA

MRNA:

Messenger RNA

Akt1:

Protein kinase Bα

Bax:

BCL2-Associated X

Wnt11:

Wnt Family Member 11

FAIM:

Fas apoptosis inhibitory molecule

LUCAT1: Lung cancer-associated transcript 1;TPP1:

Tripeptidyl peptidase 1

FoxC1:

Forkhead box C1

FNDC5:

Fibronectin type III domain-containing protein 5

IKKβ:

IκB kinase β

MiR-155-5p:

MicroRNA-155-5p

MiR-206:

MicroRNA-206

HO-1:

Haem oxygenase 1

MIF:

Macrophage migration inhibitory factor

AAVs:

Adeno-associated viruses

SFRP2:

Secreted Frizzled-related protein 2

MK:

Midkine

GATA-4:

GATA binding protein 4

Bcl-xL:

B-cell Lymphoma-extra-large

Bcl-2:

B-cell lymphoma-2

ILK:

Integrin-linked kinase

HMSCs:

Human mesenchymal stromal cells

Fstl 1:

Follistatin-like 1

ECM:

Extracellular matrix

Oct4:

Octamer-binding protein 4

BFGF:

Basic fibroblast growth factor

JNK:

C-Jun N-terminal kinase

ASK1:

Apoptosis signal-regulated kinase 1

PI3K/Akt:

Phosphoinositide 3-kinase/ Protein kinase B

SIRT3:

Sirtuin 3

MiR-10a:

MicroRNA-10a

ERBB4:

Erb-B2 receptor tyrosine kinase 4

ERK:

Extracellular signal-regulated kinase

ALKBH5:

ALKB homologue 5

Ang1:

Angiopoietin-1

AMI:

Acute myocardial infarction

SDF-1α:

Stromal cell-derived factor-1α

CXCR4:

C-X-C motif chemokine receptor type 4

VEGF-A:

Vascular endothelial growth factor-A

GSK:

Glycogen synthase kinase

GCP-2:

Granulocyte chemotactic protein-2

HGF:

Hepatocyte growth factor

IL:

Interleukin

Trx1:

Thioredoxin-1

MiR-335:

MicroRNA-335

Tbx20:

T-Box Transcription Factor 20

PYGO2:

Pygopus Family PHD Finger 2

HUC-MSCs:

Human umbilical cord-derived mesenchymal stromal cells

MEK:

Mitogen-activated extracellular signal-regulated kinase

INOS:

Inducible nitric oxide synthase

IP7:

5-Diphosphoinositol pentakisphosphate

IP6Ks:

Inositol hexakisphosphate kinases

TNP:

N2-(m-trifluorobenzyl)-N6-(p-nitrobenzyl) purine

ROS:

Reactive oxygen species

RGD:

Arginyl-glycyl-aspartic acid

HMONs:

Hollow mesoporous organosilicon nanoparticles

PEI:

Polyethyleneimine

3D:

Three-dimensional

CS/SF:

Chitosan/silk proteins

LBL:

Layer-by-layer

QHREDGS:

Glutamine-histidine-arginine-glutamic acid-aspartic acid-glycine-serine

DSAP:

Designer self-assembling peptide

WJ-MSCs:

Wharton's glial-derived MSCs

STEMI:

ST-segment elevation myocardial infarction

IC:

Intradermal injection

IV:

Intravenous injection

HUCS-MSCs:

Human umbilical cord-derived mesenchymal stromal cells

CABG:

Coronary artery bypass grafting

EF:

Ejection fractions

IL-6:

Interleukin-6

IL-10:

Interleukin-10

TGF-β:

Transforming growth factor-β

NapFF:

Naphthalene-conjugated short peptide

References

  1. Mensah GA, Fuster V, Murray CJL, Roth GA. Global burden of cardiovascular diseases and risks, 1990–2022. J Am Coll Cardiol. 2023;82:2350.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Thygesen K, Alpert JS, Jaffe AS, Chaitman BR, Bax JJ, Morrow DA, et al. Fourth universal definition of myocardial infarction (2018). J Am Coll Cardiol. 2018;72:2231.

    Article  PubMed  Google Scholar 

  3. Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ Res. 2016;119:91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhu D, Cheng K. Cardiac cell therapy for heart repair: should the cells be left out? Cells. 2021;10:

  5. Guo QY, Yang JQ, Feng XX, Zhou YJ. Regeneration of the heart: from molecular mechanisms to clinical therapeutics. Mil Med Res. 2023;10:18.

    PubMed  PubMed Central  Google Scholar 

  6. Peng X, Du J, Wang Y. Metabolic signatures in post-myocardial infarction heart failure, including insights into prediction, intervention, and prognosis. Biomed Pharmacother. 2024;170: 116079.

    Article  CAS  PubMed  Google Scholar 

  7. Keykhaei M, Ashraf H, Rashedi S, Farrokhpour H, Heidari B, Zokaei S, et al. Differences in the 2020 ESC versus 2015 ESC and 2014 ACC/AHA guidelines on the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Curr Atheroscler Rep. 2021;23:77.

    Article  PubMed  Google Scholar 

  8. Lawton JS, Tamis-Holland JE, Bangalore S, Bates ER, Beckie TM, Bischoff JM, et al. 2021 ACC/AHA/SCAI guideline for coronary artery revascularization: a report of the American college of cardiology/American heart association joint committee on clinical practice guidelines. J Am Coll Cardiol. 2022;79: e21.

    Article  PubMed  Google Scholar 

  9. Hashimoto H, Olson EN, Bassel-Duby R. Therapeutic approaches for cardiac regeneration and repair. Nat Rev Cardiol. 2018;15:585.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Sun Z, Cai Y, Chen Y, Jin Q, Zhang Z, Zhang L, et al. Ultrasound-targeted microbubble destruction promotes PDGF-primed bone mesenchymal stem cell transplantation for myocardial protection in acute Myocardial Infarction in rats. J Nanobiotechnol. 2023;21:481.

    Article  CAS  Google Scholar 

  11. Shao L, Shen Y, Ren C, Kobayashi S, Asahara T, Yang J. Inflammation in myocardial infarction: roles of mesenchymal stem cells and their secretome. Cell Death Discov. 2022;8:452.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Li Q, Hou H, Li M, Yu X, Zuo H, Gao J, et al. CD73(+) mesenchymal stem cells ameliorate myocardial infarction by promoting angiogenesis. Front Cell Dev Biol. 2021;9: 637239.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Kologrivova I, Shtatolkina M, Suslova T, Ryabov V. Cells of the immune system in cardiac remodeling: main players in resolution of inflammation and repair after myocardial infarction. Front Immunol. 2021;12: 664457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ala M. The beneficial effects of mesenchymal stem cells and their exosomes on myocardial infarction and critical considerations for enhancing their efficacy. Ageing Res Rev. 2023;89: 101980.

    Article  CAS  PubMed  Google Scholar 

  15. Li H, Hu D, Chen G, Zheng D, Li S, Lin Y, et al. Adropin-based dual treatment enhances the therapeutic potential of mesenchymal stem cells in rat myocardial infarction. Cell Death Dis. 2021;12:505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Raziyeva K, Smagulova A, Kim Y, Smagul S, Nurkesh A, Saparov A. Preconditioned and genetically modified stem cells for myocardial infarction treatment. Int J Mol Sci. 2020;21

  17. Wu T, Zhang X, Liu Y, Cui C, Sun Y, Liu W. Wet adhesive hydrogel cardiac patch loaded with anti-oxidative, autophagy-regulating molecule capsules and MSCs for restoring infarcted myocardium. Bioact Mater. 2023;21:20.

    CAS  PubMed  Google Scholar 

  18. Hodgkinson CP, Gomez JA, Mirotsou M, Dzau VJ. Genetic engineering of mesenchymal stem cells and its application in human disease therapy. Hum Gene Ther. 2010;21:1513.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim SW, Lee DW, Yu LH, Zhang HZ, Kim CE, Kim JM, et al. Mesenchymal stem cells overexpressing GCP-2 improve heart function through enhanced angiogenic properties in a myocardial infarction model. Cardiovasc Res. 2012;95:495.

    Article  CAS  PubMed  Google Scholar 

  20. Meng X, Li J, Yu M, Yang J, Zheng M, Zhang J, et al. Transplantation of mesenchymal stem cells overexpressing IL10 attenuates cardiac impairments in rats with myocardial infarction. J Cell Physiol. 2018;233:587.

    Article  CAS  PubMed  Google Scholar 

  21. Yao H, Cottin Y, Chagué F, Maza M, Bichat F, Zeller M, et al. Diagnostic and prognostic impact of new pathophysiology-based categorization of type 1 and type 2 myocardial infarction: data from the French RICO survey. Am Heart J. 2023;266:86.

    Article  PubMed  Google Scholar 

  22. Stefanadis C, Antoniou CK, Tsiachris D,Pietri P. Coronary atherosclerotic vulnerable plaque: current perspectives. J Am Heart Assoc. 2017;6

  23. Ziegler M, Wang X, Peter K. Platelets in cardiac ischaemia/reperfusion injury: a promising therapeutic target. Cardiovasc Res. 2019;115:1178.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Ischemia/reperfusion. Compr Physiol. 2016;7:113.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Heusch G. Myocardial ischaemia-reperfusion injury and cardioprotection in perspective. Nat Rev Cardiol. 2020;17:773.

    Article  PubMed  Google Scholar 

  26. Frangogiannis NG. The inflammatory response in myocardial injury, repair, and remodelling. Nat Rev Cardiol. 2014;11:255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang Y, Li Q, Tao B, Angelini M, Ramadoss S, Sun B, et al. Fibroblasts in heart scar tissue directly regulate cardiac excitability and arrhythmogenesis. Science. 2023;381:1480.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tajabadi M, Goran Orimi H, Ramzgouyan MR, Nemati A, Deravi N, Beheshtizadeh N, et al. Regenerative strategies for the consequences of myocardial infarction: chronological indication and upcoming visions. Biomed Pharmacother. 2022;146: 112584.

    Article  CAS  PubMed  Google Scholar 

  29. Williams AR, Hare JM. Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circ Res. 2011;109:923.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci. 2019;76:3323.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal. 2011;9:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Berebichez-Fridman R, Montero-Olvera PR. Sources and clinical applications of mesenchymal stem cells: state-of-the-art review. Sultan Qaboos Univ Med J. 2018;18: e264.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Cai J, Wu J, Wang J, Li Y, Hu X, Luo S, et al. Extracellular vesicles derived from different sources of mesenchymal stem cells: therapeutic effects and translational potential. Cell Biosci. 2020;10:69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bartolucci J, Verdugo FJ, González PL, Larrea RE, Abarzua E, Goset C, et al. Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: a phase 1/2 randomized controlled trial (RIMECARD trial [randomized clinical trial of intravenous infusion umbilical cord mesenchymal stem cells on cardiopathy]). Circ Res. 2017;121:1192.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wang Y, Qi Z, Yan Z, Ji N, Yang X, Gao D, et al. Mesenchymal stem cell immunomodulation: a novel intervention mechanism in cardiovascular disease. Front Cell Dev Biol. 2021;9: 742088.

    Article  PubMed  Google Scholar 

  36. Miao C, Lei M, Hu W, Han S, Wang Q. A brief review: the therapeutic potential of bone marrow mesenchymal stem cells in myocardial infarction. Stem Cell Res Ther. 2017;8:242.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Shen X, Pan B, Zhou H, Liu L, Lv T, Zhu J, et al. Differentiation of mesenchymal stem cells into cardiomyocytes is regulated by miRNA-1-2 via WNT signaling pathway. J Biomed Sci. 2017;24:29.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Tran T, Cruz C, Chan A, Awad S, Rajasingh J, Deth R, et al. Mesenchymal stem cell-derived long noncoding rnas in cardiac injury and repair. Cells. 2023;12:2268.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dabrowska S, Andrzejewska A, Janowski M, Lukomska B. Immunomodulatory and regenerative effects of mesenchymal stem cells and extracellular vesicles: therapeutic outlook for inflammatory and degenerative diseases. Front Immunol. 2020;11: 591065.

    Article  CAS  PubMed  Google Scholar 

  40. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Prolif. 2020;53: e12712.

    Article  PubMed  Google Scholar 

  41. Figueroa FE, Carrión F, Villanueva S, Khoury M. Mesenchymal stem cell treatment for autoimmune diseases: a critical review. Biol Res. 2012;45:269.

    Article  PubMed  Google Scholar 

  42. Vadivel S, Vincent P, Sekaran S, Visaga Ambi S, Muralidar S, Selvaraj V, et al. Inflammation in myocardial injury- Stem cells as potential immunomodulators for myocardial regeneration and restoration. Life Sci. 2020;250: 117582.

    Article  CAS  PubMed  Google Scholar 

  43. Golpanian S, Wolf A, Hatzistergos KE, Hare JM. Rebuilding the damaged heart: mesenchymal stem cells, cell-based therapy, and engineered heart tissue. Physiol Rev. 2016;96:1127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Pankajakshan D, Agrawal DK. Mesenchymal stem cell paracrine factors in vascular repair and regeneration. J Biomed Technol Res. 2014;1:

  45. Shafei AES, Ali MA, Ghanem HG, Shehata AI, Abdelgawad AA, Handal HR, et al. Mechanistic effects of mesenchymal and hematopoietic stem cells: New therapeutic targets in myocardial infarction. J Cell Biochem. 2018;119:5274.

    Article  CAS  PubMed  Google Scholar 

  46. Abdelwahid E, Kalvelyte A, Stulpinas A, de Carvalho KA, Guarita-Souza LC, Foldes G. Stem cell death and survival in heart regeneration and repair. Apoptosis. 2016;21:252.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Vazir A, Fox K, Westaby J, Evans MJ, Westaby S. Can we remove scar and fibrosis from adult human myocardium? Eur Heart J. 2019;40:960.

    Article  PubMed  Google Scholar 

  48. Tu C, Mezynski R, Wu JC. Improving the engraftment and integration of cell transplantation for cardiac regeneration. Cardiovasc Res. 2020;116:473.

    Article  CAS  PubMed  Google Scholar 

  49. Mahjoor M, Fakouri A, Farokhi S, Nazari H, Afkhami H, Heidari F. Regenerative potential of mesenchymal stromal cells in wound healing: unveiling the influence of normoxic and hypoxic environments. Front Cell Dev Biol. 2023;11:1245872.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Bumroongthai K, Kavanagh DPJ, Genever P, Kalia N. Improving vasculoprotective effects of MSCs in coronary microvessels - benefits of 3D culture, sub-populations and heparin. Front Immunol. 2023;14:1257497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang J, Lu Y, Mao Y, Yu Y, Wu T, Zhao W, et al. IFN-γ enhances the efficacy of mesenchymal stromal cell-derived exosomes via miR-21 in myocardial infarction rats. Stem Cell Res Ther. 2022;13:333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hahn JY, Cho HJ, Kang HJ, Kim TS, Kim MH, Chung JH, et al. Pre-treatment of mesenchymal stem cells with a combination of growth factors enhances gap junction formation, cytoprotective effect on cardiomyocytes, and therapeutic efficacy for myocardial infarction. J Am Coll Cardiol. 2008;51:933.

    Article  CAS  PubMed  Google Scholar 

  53. Xiong Y, Tang R, Xu J, Jiang W, Gong Z, Zhang L, et al. Tongxinluo-pretreated mesenchymal stem cells facilitate cardiac repair via exosomal transfer of miR-146a-5p targeting IRAK1/NF-κB p65 pathway. Stem Cell Res Ther. 2022;13:289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sun J, Shen H, Shao L, Teng X, Chen Y, Liu X, et al. HIF-1α overexpression in mesenchymal stem cell-derived exosomes mediates cardioprotection in myocardial infarction by enhanced angiogenesis. Stem Cell Res Ther. 2020;11:373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Gao X, Liang X, Liu B, Hong Y, He H, Shen Y, et al. Downregulation of ALKBH5 rejuvenates aged human mesenchymal stem cells and enhances their therapeutic efficacy in myocardial infarction. Faseb J. 2023;37: e23294.

    Article  CAS  PubMed  Google Scholar 

  56. Ramesh S, Govarthanan K, Ostrovidov S, Zhang H, Hu Q, Camci-Unal G, et al. Cardiac differentiation of mesenchymal stem cells: impact of biological and chemical inducers. Stem Cell Rev Rep. 2021;17:1343.

    Article  CAS  PubMed  Google Scholar 

  57. Oggu GS, Sasikumar S, Reddy N, Ella KKR, Rao CM, Bokara KK. Gene delivery approaches for mesenchymal stem cell therapy: strategies to increase efficiency and specificity. Stem Cell Rev Rep. 2017;13:725.

    Article  CAS  PubMed  Google Scholar 

  58. Phillips MI, Tang YL. Genetic modification of stem cells for transplantation. Adv Drug Deliv Rev. 2008;60:160.

    Article  CAS  PubMed  Google Scholar 

  59. Chen J, Liu F, Hu W, Qian Y, Xu D, Gao C, et al. FAIM enhances the efficacy of mesenchymal stem cell transplantation by inhibiting JNK-induced c-FLIP ubiquitination and degradation. Stem Cells Int. 2022;2022:3705637.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Xiang Q, Liao Y, Chao H, Huang W, Liu J, Chen H, et al. ISL1 overexpression enhances the survival of transplanted human mesenchymal stem cells in a murine myocardial infarction model. Stem Cell Res Ther. 2018;9:51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Xia Y, Xu X, Guo Y, Lin C, Xu X, Zhang F, et al. Mesenchymal stromal cells overexpressing farnesoid x receptor exert cardioprotective effects against acute ischemic heart injury by binding endogenous bile acids. Adv Sci (Weinh). 2022;9: e2200431.

    Article  PubMed  Google Scholar 

  62. Tao Y, Liu Q, Wu R, Xiao C, Ni C, Wang K, et al. Long noncoding RNA LUCAT1 enhances the survival and therapeutic effects of mesenchymal stromal cells post-myocardial infarction. Mol Ther Nucleic Acids. 2022;27:412.

    Article  CAS  PubMed  Google Scholar 

  63. Zhang H, Zhao C, Jiang G, Hu B, Zheng H, Hong Y, et al. Apelin rejuvenates aged human mesenchymal stem cells by regulating autophagy and improves cardiac protection after infarction. Front Cell Dev Biol. 2021;9: 628463.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Yu K, Zeng Z, Cheng S, Hu W, Gao C, Liu F, et al. TPP1 enhances the therapeutic effects of transplanted aged mesenchymal stem cells in infarcted hearts via the MRE11/AKT pathway. Front Cell Dev Biol. 2020;8: 588023.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Zhao L, Zhang R, Su F, Dai L, Wang J, Cui J, et al. FoxC1-induced vascular niche improves survival and myocardial repair of mesenchymal stem cells in infarcted hearts. Oxid Med Cell Longev. 2020;2020:7865395.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Abu-El-Rub E, Sareen N, Lester Sequiera G, Ammar HI, Yan W, ShamsEldeen AM, et al. Hypoxia-induced increase in Sug1 leads to poor post-transplantation survival of allogeneic mesenchymal stem cells. Faseb j. 2020;34:12860.

    Article  CAS  PubMed  Google Scholar 

  67. Deng J, Zhang N, Wang Y, Yang C, Wang Y, Xin C, et al. FNDC5/irisin improves the therapeutic efficacy of bone marrow-derived mesenchymal stem cells for myocardial infarction. Stem Cell Res Ther. 2020;11:228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kizilay Mancini O, Huynh DN, Menard L, Shum-Tim D, Ong H, Marleau S, et al. Ex vivo Ikkβ ablation rescues the immunopotency of mesenchymal stromal cells from diabetics with advanced atherosclerosis. Cardiovasc Res. 2021;117:756.

    Article  PubMed  Google Scholar 

  69. Hong Y, He H, Jiang G, Zhang H, Tao W, Ding Y, et al. miR-155-5p inhibition rejuvenates aged mesenchymal stem cells and enhances cardioprotection following infarction. Aging Cell. 2020;19: e13128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Liu X, Yang Z, Meng Q, Chen Y, Shao L, Li J, et al. Downregulation of microRNA-206 alleviates the sublethal oxidative stress-induced premature senescence and dysfunction in mesenchymal stem cells via targeting alpl. Oxid Med Cell Longev. 2020;2020:7242836.

    PubMed  PubMed Central  Google Scholar 

  71. Deng R, Liu Y, He H, Zhang H, Zhao C, Cui Z, et al. Haemin pre-treatment augments the cardiac protection of mesenchymal stem cells by inhibiting mitochondrial fission and improving survival. J Cell Mol Med. 2020;24:431.

    Article  CAS  PubMed  Google Scholar 

  72. Zhang Y, Zhu W, He H, Fan B, Deng R, Hong Y, et al. Macrophage migration inhibitory factor rejuvenates aged human mesenchymal stem cells and improves myocardial repair. Aging (Albany NY). 2019;11:12641.

    Article  CAS  PubMed  Google Scholar 

  73. Preda MB, Neculachi CA, Fenyo IM, Vacaru A-M, Publik MA, Simionescu M, et al. Short lifespan of syngeneic transplanted MSC is a consequence of in vivo apoptosis and immune cell recruitment in mice. Cell Death Dis. 2021;12:566.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Xie Y, Liu S, Wang L, Yang H, Tai C, Ling L, et al. Individual heterogeneity screened umbilical cord-derived mesenchymal stromal cells with high Treg promotion demonstrate improved recovery of mouse liver fibrosis. Stem Cell Res Ther. 2021;12:359.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wang Y, Gao T, Wang B. Application of mesenchymal stem cells for anti-senescence and clinical challenges. Stem Cell Res Ther. 2023;14:260.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Xie Y, Liu W, Liu S, Wang L, Mu D, Cui Y, et al. The quality evaluation system establishment of mesenchymal stromal cells for cell-based therapy products. Stem Cell Res Ther. 2020;11:176.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS, et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med. 2003;9:1195.

    Article  CAS  PubMed  Google Scholar 

  78. Noiseux N, Gnecchi M, Lopez-Ilasaca M, Zhang L, Solomon SD, Deb A, et al. Mesenchymal stem cells overexpressing Akt dramatically repair infarcted myocardium and improve cardiac function despite infrequent cellular fusion or differentiation. Mol Ther. 2006;14:840.

    Article  CAS  PubMed  Google Scholar 

  79. Zhao T, Zhang D, Millard RW, Ashraf M, Wang Y. Stem cell homing and angiomyogenesis in transplanted hearts are enhanced by combined intramyocardial SDF-1alpha delivery and endogenous cytokine signaling. Am J Physiol Heart Circ Physiol. 2009;296:H976.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Deng J, Han Y, Yan C, Tian X, Tao J, Kang J, et al. Overexpressing cellular repressor of E1A-stimulated genes protects mesenchymal stem cells against hypoxia- and serum deprivation-induced apoptosis by activation of PI3K/Akt. Apoptosis. 2010;15:463.

    Article  CAS  PubMed  Google Scholar 

  81. Chen B, Chen X, Liu C, Li J, Liu F, Huang Y. Co-expression of Akt1 and Wnt11 promotes the proliferation and cardiac differentiation of mesenchymal stem cells and attenuates hypoxia/reoxygenation-induced cardiomyocyte apoptosis. Biomed Pharmacother. 2018;108:508.

    Article  CAS  PubMed  Google Scholar 

  82. Song H, Kwon K, Lim S, Kang SM, Ko YG, Xu Z, et al. Transfection of mesenchymal stem cells with the FGF-2 gene improves their survival under hypoxic conditions. Mol Cells. 2005;19:402.

    Article  CAS  PubMed  Google Scholar 

  83. Alfaro MP, Vincent A, Saraswati S, Thorne CA, Hong CC, Lee E, et al. sFRP2 suppression of bone morphogenic protein (BMP) and Wnt signaling mediates mesenchymal stem cell (MSC) self-renewal promoting engraftment and myocardial repair. J Biol Chem. 2010;285:35645.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Zhao SL, Zhang YJ, Li MH, Zhang XL, Chen SL. Mesenchymal stem cells with overexpression of midkine enhance cell survival and attenuate cardiac dysfunction in a rat model of myocardial infarction. Stem Cell Res Ther. 2014;5:37.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Li H, Zuo S, He Z, Yang Y, Pasha Z, Wang Y, et al. Paracrine factors released by GATA-4 overexpressed mesenchymal stem cells increase angiogenesis and cell survival. Am J Physiol Heart Circ Physiol. 2010;299:H1772.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Xue X, Liu Y, Zhang J, Liu T, Yang Z, Wang H. Bcl-xL genetic modification enhanced the therapeutic efficacy of mesenchymal stem cell transplantation in the treatment of heart infarction. Stem Cells International. 2015;2015: 176409.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Zeng B, Chen H, Zhu C, Ren X, Lin G, Cao F. Effects of combined mesenchymal stem cells and heme oxygenase-1 therapy on cardiac performance. Eur J Cardiothorac Surg. 2008;34:850.

    Article  PubMed  Google Scholar 

  88. Huang J, Zhang Z, Guo J, Ni A, Deb A, Zhang L, et al. Genetic modification of mesenchymal stem cells overexpressing CCR1 increases cell viability, migration, engraftment, and capillary density in the injured myocardium. Circ Res. 2010;106:1753.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Mu D, Zhang XL, Xie J, Yuan HH, Wang K, Huang W, et al. Intracoronary transplantation of mesenchymal stem cells with overexpressed integrin-linked kinase improves cardiac function in porcine myocardial infarction. Sci Rep. 2016;6:19155.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Shen H, Cui G, Li Y, Ye W, Sun Y, Zhang Z, et al. Follistatin-like 1 protects mesenchymal stem cells from hypoxic damage and enhances their therapeutic efficacy in a mouse myocardial infarction model. Stem Cell Res Ther. 2019;10:17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Wang P, Deng Z, Li A, Li R, Huang W, Cui J, et al. β-Catenin promotes long-term survival and angiogenesis of peripheral blood mesenchymal stem cells via the Oct4 signaling pathway. Exp Mol Med. 2022;54:1434.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Liang Y, Lin Q, Zhu J, Li X, Fu Y, Zou X, et al. The caspase-8 shRNA-modified mesenchymal stem cells improve the function of infarcted heart. Mol Cell Biochem. 2014;397:7.

    Article  CAS  PubMed  Google Scholar 

  93. Lee CY, Shin S, Lee J, Seo HH, Lim KH, Kim H, et al. MicroRNA-mediated down-regulation of apoptosis signal-regulating kinase 1 (ASK1) attenuates the apoptosis of human mesenchymal stem cells (MSCs) transplanted into infarcted heart. Int J Mol Sci. 2016;17

  94. Chen Y, Zhao Y, Chen W, Xie L, Zhao Z-A, Yang J, et al. MicroRNA-133 overexpression promotes the therapeutic efficacy of mesenchymal stem cells on acute myocardial infarction. Stem Cell Res Ther. 2017;8:268.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Yang F, Wu R, Jiang Z, Chen J, Nan J, Su S, et al. Leptin increases mitochondrial OPA1 via GSK3-mediated OMA1 ubiquitination to enhance therapeutic effects of mesenchymal stem cell transplantation. Cell Death Disease. 2018;9:556.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Fu J, Chen X, Liu X, Xu D, Yang H, Zeng C, et al. ELABELA ameliorates hypoxic/ischemic-induced bone mesenchymal stem cell apoptosis via alleviation of mitochondrial dysfunction and activation of PI3K/AKT and ERK1/2 pathways. Stem Cell Res Ther. 2020;11:541.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Banimohamad-Shotorbani B, Kahroba H, Sadeghzadeh H, Wilson DM, Maadi H, Samadi N, et al. DNA damage repair response in mesenchymal stromal cells: from cellular senescence and aging to apoptosis and differentiation ability. Ageing Res Rev. 2020;62: 101125.

    Article  CAS  PubMed  Google Scholar 

  98. Zhang J, Akiyama K, Mun AY, Tagashira R, Zou T, Matsunaga N, et al. Age-related effects on msc immunomodulation, macrophage polarization, apoptosis, and bone regeneration correlate with IL-38 expression. Int J Mol Sci. 2024;25

  99. Turinetto V, Vitale E, Giachino C. Senescence in human mesenchymal stem cells: functional changes and implications in stem cell-based therapy. Int J Mol Sci. 2016;17

  100. Weng Z, Wang Y, Ouchi T, Liu H, Qiao X, Wu C, et al. Mesenchymal stem/stromal cell senescence: hallmarks, mechanisms, and combating strategies. Stem Cells Transl Med. 2022;11:356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Zhao M, Liu S, Wang Y, Lv K, Lou P, Zhou P, et al. The mitochondria-paraspeckle axis regulates the survival of transplanted stem cells under oxidative stress conditions. Theranostics. 2024;14:1517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Zhang DY, Zhang CF, Fu BC, Sun L, Wang XQ, Chen W, et al. Sirtuin3 protects aged human mesenchymal stem cells against oxidative stress and enhances efficacy of cell therapy for ischaemic heart diseases. J Cell Mol Med. 2018;22:5504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Dong J, Zhang Z, Huang H, Mo P, Cheng C, Liu J, et al. miR-10a rejuvenates aged human mesenchymal stem cells and improves heart function after myocardial infarction through KLF4. Stem Cell Res Ther. 2018;9:151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Liang X, Ding Y, Lin F, Zhang Y, Zhou X, Meng Q, et al. Overexpression of ERBB4 rejuvenates aged mesenchymal stem cells and enhances angiogenesis via PI3K/AKT and MAPK/ERK pathways. Faseb j. 2019;33:4559.

    Article  CAS  PubMed  Google Scholar 

  105. Yang M, Wen T, Chen H, Deng J, Yang C, Zhang Z. Knockdown of insulin-like growth factor 1 exerts a protective effect on hypoxic injury of aged BM-MSCs: role of autophagy. Stem Cell Res Ther. 2018;9:284.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res. 2004;95:9.

    Article  CAS  PubMed  Google Scholar 

  107. Nagaya N, Fujii T, Iwase T, Ohgushi H, Itoh T, Uematsu M, et al. Intravenous administration of mesenchymal stem cells improves cardiac function in rats with acute myocardial infarction through angiogenesis and myogenesis. Am J Physiol Heart Circ Physiol. 2004;287:H2670.

    Article  CAS  PubMed  Google Scholar 

  108. Shyu K-G, Wang B-W, Hung H-F, Chang C-C, Shih DT-B. Mesenchymal stem cells are superior to angiogenic growth factor genes for improving myocardial performance in the mouse model of acute myocardial infarction. J Biomed Sci. 2006;13:47.

    Article  CAS  PubMed  Google Scholar 

  109. Matsumoto R, Omura T, Yoshiyama M, Hayashi T, Inamoto S, Koh KR, et al. Vascular endothelial growth factor-expressing mesenchymal stem cell transplantation for the treatment of acute myocardial infarction. Arterioscler Thromb Vasc Biol. 2005;25:1168.

    Article  CAS  PubMed  Google Scholar 

  110. Huang SD, Lu FL, Xu XY, Liu XH, Zhao XX, Zhao BZ, et al. Transplantation of angiogenin-overexpressing mesenchymal stem cells synergistically augments cardiac function in a porcine model of chronic ischemia. J Thorac Cardiovasc Surg. 2006;132:1329.

    Article  CAS  PubMed  Google Scholar 

  111. Jiang S, Haider H, Idris NM, Salim A, Ashraf M. Supportive interaction between cell survival signaling and angiocompetent factors enhances donor cell survival and promotes angiomyogenesis for cardiac repair. Circ Res. 2006;99:776.

    Article  CAS  PubMed  Google Scholar 

  112. Shujia J, Haider HK, Idris NM, Lu G, Ashraf M. Stable therapeutic effects of mesenchymal stem cell-based multiple gene delivery for cardiac repair. Cardiovasc Res. 2008;77:525.

    Article  CAS  PubMed  Google Scholar 

  113. Zhang D, Fan GC, Zhou X, Zhao T, Pasha Z, Xu M, et al. Over-expression of CXCR4 on mesenchymal stem cells augments myoangiogenesis in the infarcted myocardium. J Mol Cell Cardiol. 2008;44:281.

    Article  CAS  PubMed  Google Scholar 

  114. Cho J, Zhai P, Maejima Y, Sadoshima J. Myocardial injection with GSK-3β-overexpressing bone marrow-derived mesenchymal stem cells attenuates cardiac dysfunction after myocardial infarction. Circ Res. 2011;108:478.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Chen H, Xia R, Li Z, Zhang L, Xia C, Ai H, et al. Mesenchymal stem cells combined with hepatocyte growth factor therapy for attenuating ischaemic myocardial fibrosis: assessment using multimodal molecular imaging. Sci Rep. 2016;6:33700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zhao Y, Zhu J, Zhang N, Liu Q, Wang Y, Hu X, et al. GDF11 enhances therapeutic efficacy of mesenchymal stem cells for myocardial infarction via YME1L-mediated OPA1 processing. Stem Cells Transl Med. 2020;9:1257.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Liu X, Chen H, Zhu W, Chen H, Hu X, Jiang Z, et al. Transplantation of SIRT1-engineered aged mesenchymal stem cells improves cardiac function in a rat myocardial infarction model. J Heart Lung Transplant. 2014;33:1083.

    Article  PubMed  Google Scholar 

  118. Suresh SC, Selvaraju V, Thirunavukkarasu M, Goldman JW, Husain A, Alexander Palesty J, et al. Thioredoxin-1 (Trx1) engineered mesenchymal stem cell therapy increased pro-angiogenic factors, reduced fibrosis and improved heart function in the infarcted rat myocardium. Int J Cardiol. 2015;201:517.

    Article  PubMed  Google Scholar 

  119. Gómez-Mauricio G, Moscoso I, Martín-Cancho MF, Crisóstomo V, Prat-Vidal C, Báez-Díaz C, et al. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Zhang S, Zhao L, Wang J, Chen N, Yan J, Pan X. HIF-2α and Oct4 have synergistic effects on survival and myocardial repair of very small embryonic-like mesenchymal stem cells in infarcted hearts. Cell Death Dis. 2017;8: e2548.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Zhao L, Wang J, Wang P, Deng Z, Cui J, Huang W, et al. Oct4 cooperates with c-Myc to improve mesenchymal-to-endothelial transition and myocardial repair of cardiac-resident mesenchymal stem cells. Stem Cell Res Ther. 2022;13:445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Kawada H, Fujita J, Kinjo K, Matsuzaki Y, Tsuma M, Miyatake H, et al. Nonhematopoietic mesenchymal stem cells can be mobilized and differentiate into cardiomyocytes after myocardial infarction. Blood. 2004;104:3581.

    Article  CAS  PubMed  Google Scholar 

  123. Fukuda K, Fujita J. Mesenchymal, but not hematopoietic, stem cells can be mobilized and differentiate into cardiomyocytes after myocardial infarction in mice. Kidney Int. 2005;68:1940.

    Article  CAS  PubMed  Google Scholar 

  124. Tsuji H, Miyoshi S, Ikegami Y, Hida N, Asada H, Togashi I, et al. Xenografted human amniotic membrane-derived mesenchymal stem cells are immunologically tolerated and transdifferentiated into cardiomyocytes. Circ Res. 2010;106:1613.

    Article  CAS  PubMed  Google Scholar 

  125. Grauss RW, van Tuyn J, Steendijk P, Winter EM, Pijnappels DA, Hogers B, et al. Forced myocardin expression enhances the therapeutic effect of human mesenchymal stem cells after transplantation in ischemic mouse hearts. Stem Cells. 2008;26:1083.

    Article  PubMed  Google Scholar 

  126. Lee SY, Ham O, Cha MJ, Song BW, Choi E, Kim IK, et al. The promotion of cardiogenic differentiation of hMSCs by targeting epidermal growth factor receptor using microRNA-133a. Biomaterials. 2013;34:92.

    Article  CAS  PubMed  Google Scholar 

  127. Neshati V, Mollazadeh S, Fazly Bazzaz BS, de Vries AA, Mojarrad M, Naderi-Meshkin H, et al. Cardiomyogenic differentiation of human adipose-derived mesenchymal stem cells transduced with Tbx20-encoding lentiviral vectors. J Cell Biochem. 2018;119:6146.

    Article  CAS  PubMed  Google Scholar 

  128. Shi Y, Qin B, Fan X, Li Y, Wang Y, Yuan W, et al. Novel biphasic mechanism of the canonical Wnt signalling component PYGO2 promotes cardiomyocyte differentiation from hUC-MSCs. Cell Tissue Res. 2023;393:163.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Pasha Z, Wang Y, Sheikh R, Zhang D, Zhao T, Ashraf M. Preconditioning enhances cell survival and differentiation of stem cells during transplantation in infarcted myocardium. Cardiovasc Res. 2008;77:134.

    Article  CAS  PubMed  Google Scholar 

  130. Russo V, Young S, Hamilton A, Amsden BG, Flynn LE. Mesenchymal stem cell delivery strategies to promote cardiac regeneration following ischemic injury. Biomaterials. 2014;35:3956.

    Article  CAS  PubMed  Google Scholar 

  131. Zhang Z, Li S, Cui M, Gao X, Sun D, Qin X, et al. Rosuvastatin enhances the therapeutic efficacy of adipose-derived mesenchymal stem cells for myocardial infarction via PI3K/Akt and MEK/ERK pathways. Basic Res Cardiol. 2013;108:333.

    Article  PubMed  Google Scholar 

  132. Xu J, Xiong YY, Li Q, Hu MJ, Huang PS, Xu JY, et al. Optimization of timing and times for administration of atorvastatin-pretreated mesenchymal stem cells in a preclinical model of acute myocardial infarction. Stem Cells Transl Med. 2019;8:1068.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Li HM, Liu L, Mei X, Chen H, Liu Z, Zhao X. Overexpression of inducible nitric oxide synthase impairs the survival of bone marrow stem cells transplanted into rat infarcted myocardium. Life Sci. 2014;106:50.

    Article  CAS  PubMed  Google Scholar 

  134. Zhou H, Yang J, Xin T, Li D, Guo J, Hu S, et al. Exendin-4 protects adipose-derived mesenchymal stem cells from apoptosis induced by hydrogen peroxide through the PI3K/Akt-Sfrp2 pathways. Free Radic Biol Med. 2014;77:363.

    Article  CAS  PubMed  Google Scholar 

  135. Zhang Z, Liang D, Gao X, Zhao C, Qin X, Xu Y, et al. Selective inhibition of inositol hexakisphosphate kinases (IP6Ks) enhances mesenchymal stem cell engraftment and improves therapeutic efficacy for myocardial infarction. Basic Res Cardiol. 2014;109:417.

    Article  PubMed  Google Scholar 

  136. Zhang GW, Gu TX, Sun XJ, Wang C, Qi X, Wang XB, et al. Edaravone promotes activation of resident cardiac stem cells by transplanted mesenchymal stem cells in a rat myocardial infarction model. J Thorac Cardiovasc Surg. 2016;152:570.

    Article  CAS  PubMed  Google Scholar 

  137. Han XJ, Li H, Liu CB, Luo ZR, Wang QL, Mou FF, et al. Guanxin Danshen Formulation improved the effect of mesenchymal stem cells transplantation for the treatment of myocardial infarction probably via enhancing the engraftment. Life Sci. 2019;233: 116740.

    Article  CAS  PubMed  Google Scholar 

  138. Huang W, Zhang D, Millard RW, Wang T, Zhao T, Fan GC, et al. Gene manipulated peritoneal cell patch repairs infarcted myocardium. J Mol Cell Cardiol. 2010;48:702.

    Article  CAS  PubMed  Google Scholar 

  139. Yu J, Du KT, Fang Q, Gu Y, Mihardja SS, Sievers RE, et al. The use of human mesenchymal stem cells encapsulated in RGD modified alginate microspheres in the repair of myocardial infarction in the rat. Biomaterials. 2010;31:7012.

    Article  CAS  PubMed  Google Scholar 

  140. Maureira P, Marie PY, Yu F, Poussier S, Liu Y, Groubatch F, et al. Repairing chronic myocardial infarction with autologous mesenchymal stem cells engineered tissue in rat promotes angiogenesis and limits ventricular remodeling. J Biomed Sci. 2012;19:93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Kai D, Wang QL, Wang HJ, Prabhakaran MP, Zhang Y, Tan YZ, et al. Stem cell-loaded nanofibrous patch promotes the regeneration of infarcted myocardium with functional improvement in rat model. Acta Biomater. 2014;10:2727.

    Article  CAS  PubMed  Google Scholar 

  142. Bonafè F, Govoni M, Giordano E, Caldarera CM, Guarnieri C, Muscari C. Hyaluronan and cardiac regeneration. J Biomed Sci. 2014;21:100.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Liu G, Li L, Huo D, Li Y, Wu Y, Zeng L, et al. A VEGF delivery system targeting MI improves angiogenesis and cardiac function based on the tropism of MSCs and layer-by-layer self-assembly. Biomaterials. 2017;127:117.

    Article  CAS  PubMed  Google Scholar 

  144. Yao Y, Yang L, Feng LF, Yue ZW, Zhao NH, Li Z, et al. IGF-1C domain-modified hydrogel enhanced the efficacy of stem cells in the treatment of AMI. Stem Cell Res Ther. 2020;11:136.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Guo R, Wan F, Morimatsu M, Xu Q, Feng T, Yang H, et al. Cell sheet formation enhances the therapeutic effects of human umbilical cord mesenchymal stem cells on myocardial infarction as a bioactive material. Bioact Mater. 2021;6:2999.

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Lee YS, Lim KS, Oh JE, Yoon AR, Joo WS, Kim HS, et al. Development of porous PLGA/PEI1.8k biodegradable microspheres for the delivery of mesenchymal stem cells (MSCs). J Control Release. 2015;205:128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Zhu S, Yu C, Liu N, Zhao M, Chen Z, Liu J, et al. Injectable conductive gelatin methacrylate/oxidized dextran hydrogel encapsulating umbilical cord mesenchymal stem cells for myocardial infarction treatment. Bioact Mater. 2022;13:119.

    CAS  PubMed  Google Scholar 

  148. Zhu K, Lai H, Guo C, Li J, Wang Y, Wang L, et al. Nanovector-based prolyl hydroxylase domain 2 silencing system enhances the efficiency of stem cell transplantation for infarcted myocardium repair. Int J Nanomedicine. 2014;9:5203.

    PubMed  PubMed Central  Google Scholar 

  149. Zhu K, Wu M, Lai H, Guo C, Li J, Wang Y, et al. Nanoparticle-enhanced generation of gene-transfected mesenchymal stem cells for in vivo cardiac repair. Biomaterials. 2016;74:188.

    Article  CAS  PubMed  Google Scholar 

  150. Chen J, Zhan Y, Wang Y, Han D, Tao B, Luo Z, et al. Chitosan/silk fibroin modified nanofibrous patches with mesenchymal stem cells prevent heart remodeling post-myocardial infarction in rats. Acta Biomater. 2018;80:154.

    Article  CAS  PubMed  Google Scholar 

  151. Cai H, Wu FY, Wang QL, Xu P, Mou FF, Shao SJ, et al. Self-assembling peptide modified with QHREDGS as a novel delivery system for mesenchymal stem cell transplantation after myocardial infarction. Faseb j. 2019;33:8306.

    Article  CAS  PubMed  Google Scholar 

  152. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009;54:2277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA. 2014;311:62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Suncion VY, Ghersin E, Fishman JE, Zambrano JP, Karantalis V, Mandel N, et al. Does transendocardial injection of mesenchymal stem cells improve myocardial function locally or globally?: An analysis from the Percutaneous Stem Cell Injection Delivery Effects on Neomyogenesis (POSEIDON) randomized trial. Circ Res. 2014;114:1292.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Florea V, Rieger AC, DiFede DL, El-Khorazaty J, Natsumeda M, Banerjee MN, et al. Dose comparison study of allogeneic mesenchymal stem cells in patients with ischemic cardiomyopathy (the TRIDENT study). Circ Res. 2017;121:1279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Gao LR, Pei XT, Ding QA, Chen Y, Zhang NK, Chen HY, et al. A critical challenge: dosage-related efficacy and acute complication intracoronary injection of autologous bone marrow mesenchymal stem cells in acute myocardial infarction. Int J Cardiol. 2013;168:3191.

    Article  PubMed  Google Scholar 

  157. Gao LR, Chen Y, Zhang NK, Yang XL, Liu HL, Wang ZG, et al. Intracoronary infusion of Wharton’s jelly-derived mesenchymal stem cells in acute myocardial infarction: double-blind, randomized controlled trial. BMC Med. 2015;13:162.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Attar A, Farjoud Kouhanjani M, Hessami K, Vosough M, Kojuri J, Ramzi M, et al. Effect of once versus twice intracoronary injection of allogeneic-derived mesenchymal stromal cells after acute myocardial infarction: BOOSTER-TAHA7 randomized clinical trial. Stem Cell Res Ther. 2023;14:264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Park BW, Jung SH, Das S, Lee SM, Park JH, Kim H, et al. In vivo priming of human mesenchymal stem cells with hepatocyte growth factor-engineered mesenchymal stem cells promotes therapeutic potential for cardiac repair. Sci Adv. 2020;6:eaay6994.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Gao LR, Chen Y, Zhang NK, Yang XL, Liu HL, Wang ZG, et al. Intracoronary infusion of Wharton’s jelly-derived mesenchymal stem cells in acute myocardial infarction: double-blind, randomized controlled trial. BMC Med. 2015;13:162.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Hsiao LC, Lin YN, Shyu WC, Ho M, Lu CR, Chang SS, et al. First-in-human pilot trial of combined intracoronary and intravenous mesenchymal stem cell therapy in acute myocardial infarction. Front Cardiovasc Med. 2022;9: 961920.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Can A, Ulus AT, Cinar O, Topal Celikkan F, Simsek E, Akyol M, et al. Human umbilical cord mesenchymal stromal cell transplantation in myocardial ischemia (HUC-HEART Trial). A study protocol of a phase 1/2, controlled and randomized trial in combination with coronary artery bypass grafting. Stem Cell Rev Rep. 2015;11:752.

    Article  PubMed  Google Scholar 

  163. Ulus AT, Mungan C, Kurtoglu M, Celikkan FT, Akyol M, Sucu M, et al. Intramyocardial transplantation of umbilical cord mesenchymal stromal cells in chronic ischemic cardiomyopathy: a controlled, randomized clinical trial (HUC-HEART Trial). Int J Stem Cells. 2020;13:364.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Sun SJ, Wei R, Li F, Liao SY, Tse HF. Mesenchymal stromal cell-derived exosomes in cardiac regeneration and repair. Stem Cell Reports. 2021;16:1662.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Huang L, Ma W, Ma Y, Feng D, Chen H, Cai B. Exosomes in mesenchymal stem cells, a new therapeutic strategy for cardiovascular diseases? Int J Biol Sci. 2015;11:238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Shi Y, Shi H, Nomi A, Lei-Lei Z, Zhang B, Qian H. Mesenchymal stem cell-derived extracellular vesicles: a new impetus of promoting angiogenesis in tissue regeneration. Cytotherapy. 2019;21:497.

    Article  CAS  PubMed  Google Scholar 

  167. Tan SJO, Floriano JF, Nicastro L, Emanueli C,Catapano F. Novel applications of mesenchymal stem cell-derived exosomes for myocardial infarction therapeutics. Biomolecules. 2020;10:

  168. Cheng L, Zhang K, Wu S, Cui M, Xu T. Focus on mesenchymal stem cell-derived exosomes: opportunities and challenges in cell-free therapy. Stem Cells International. 2017;2017:6305295.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Song Y, Wang B, Zhu X, Hu J, Sun J, Xuan J, et al. Human umbilical cord blood–derived MSCs exosome attenuate myocardial injury by inhibiting ferroptosis in acute myocardial infarction mice. Cell Biol Toxicol. 2021;37:51.

    Article  CAS  PubMed  Google Scholar 

  170. Xu R, Zhang F, Chai R, Zhou W, Hu M, Liu B, et al. Exosomes derived from pro-inflammatory bone marrow-derived mesenchymal stem cells reduce inflammation and myocardial injury via mediating macrophage polarization. J Cell Mol Med. 2019;23:7617.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Zhang N, Zhu J, Ma Q, Zhao Y, Wang Y, Hu X, et al. Exosomes derived from human umbilical cord MSCs rejuvenate aged MSCs and enhance their functions for myocardial repair. Stem Cell Res Ther. 2020;11:273.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Yu T, Xu Q, Chen X, Deng X, Chen N, Kou MT, et al. Biomimetic nanomaterials in myocardial infarction treatment: Harnessing bionic strategies for advanced therapeutics. Materials Today Bio. 2024;25: 100957.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Huang P, Wang L, Li Q, Tian X, Xu J, Xu J, et al. Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding RNA H19. Cardiovasc Res. 2020;116:353.

    Article  CAS  PubMed  Google Scholar 

  174. Xiong Y, Tang R, Xu J, Jiang W, Gong Z, Zhang L, et al. Sequential transplantation of exosomes and mesenchymal stem cells pretreated with a combination of hypoxia and Tongxinluo efficiently facilitates cardiac repair. Stem Cell Res Ther. 2022;13:63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Wang X, Chen Y, Zhao Z, Meng Q, Yu Y, Sun J, et al. Engineered exosomes with ischemic myocardium-targeting peptide for targeted therapy in myocardial infarction. J Am Heart Assoc. 2018;7: e008737.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Han C, Zhou J, Liang C, Liu B, Pan X, Zhang Y, et al. Human umbilical cord mesenchymal stem cell derived exosomes encapsulated in functional peptide hydrogels promote cardiac repair. Biomater Sci. 2019;7:2920.

    Article  CAS  PubMed  Google Scholar 

  177. Lee JR, Park BW, Kim J, Choo YW, Kim HY, Yoon JK, et al. Nanovesicles derived from iron oxide nanoparticles-incorporated mesenchymal stem cells for cardiac repair. Sci Adv. 2020;6:eaaz0952.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Yao C, Wu W, Tang H, Jia X, Tang J, Ruan X, et al. Self-assembly of stem cell membrane-camouflaged nanocomplex for microRNA-mediated repair of myocardial infarction injury. Biomaterials. 2020;257: 120256.

    Article  CAS  PubMed  Google Scholar 

  179. Yao J, Huang K, Zhu D, Chen T, Jiang Y, Zhang J, et al. A minimally invasive exosome spray repairs heart after myocardial infarction. ACS Nano. 2021;15:11099.

    Article  CAS  PubMed  Google Scholar 

  180. Zou Y, Li L, Li Y, Chen S, Xie X, Jin X, et al. Restoring cardiac functions after myocardial infarction–ischemia/reperfusion via an exosome anchoring conductive hydrogel. ACS Appl Mater Interfaces. 2021;13:56892.

    Article  CAS  PubMed  Google Scholar 

  181. Wang Q, Zhang L, Sun Z, Chi B, Zou A, Mao L, et al. HIF-1α overexpression in mesenchymal stem cell-derived exosome-encapsulated arginine-glycine-aspartate (RGD) hydrogels boost therapeutic efficacy of cardiac repair after myocardial infarction. Mater Today Bio. 2021;12: 100171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Hu X, Ning X, Zhao Q, Zhang Z, Zhang C, Xie M, et al. Islet-1 mesenchymal stem cells-derived exosome-incorporated angiogenin-1 hydrogel for enhanced acute myocardial infarction therapy. ACS Appl Mater Interfaces. 2022;14:36289.

    Article  CAS  PubMed  Google Scholar 

  183. Yuan J, Yang H, Liu C, Shao L, Zhang H, Lu K, et al. Microneedle patch loaded with exosomes containing microRNA-29b prevents cardiac fibrosis after myocardial infarction. Adv Healthc Mater. 2023;12: e2202959.

    Article  PubMed  Google Scholar 

  184. Ping P, Guan S, Ning C, Yang T, Zhao Y, Zhang P, et al. Fabrication of blended nanofibrous cardiac patch transplanted with TGF-β3 and human umbilical cord MSCs-derived exosomes for potential cardiac regeneration after acute myocardial infarction. Nanomed Nanotechnol Biol Med. 2023;54: 102708.

    Article  CAS  Google Scholar 

  185. Yan C, Wang X, Wang Q, Li H, Song H, Zhou J, et al. A novel conductive polypyrrole-chitosan hydrogel containing human endometrial mesenchymal stem cell-derived exosomes facilitated sustained release for cardiac repair. Adv Healthc Mater. 2024;e2304207.

  186. 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.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Thanks to the support of PubMed, all references are from the website.

Funding

This study was supported by National Natural Science Foundation of China (NSFC) [81571213 and 82070459 (Bin Wang)], Key Project of Jiangsu Province (Grant No. BE2020765) (Bin Wang), Project of Modern Hospital Management and Development Institute, Nanjing University/Aid project of Nanjing Drum Tower Hospital Health, Education & Research Foundation (NDYG2020030) (Bin Wang).

Author information

Authors and Affiliations

Authors

Contributions

BW designed the study. CwX wrote the first draft of the manuscript, YyX and BW revised the manuscript and all authors read, edited, and approved the final manuscript.

Corresponding author

Correspondence to Bin Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, C., Xie, Y. & Wang, B. Genetically modified mesenchymal stromal cells: a cell-based therapy offering more efficient repair after myocardial infarction. Stem Cell Res Ther 15, 323 (2024). https://doi.org/10.1186/s13287-024-03942-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-024-03942-7

Keywords