Application of Stem Cell Technologies to Regenerate Injured Myocardium and Improve Cardiac Function

 

Parisa Mardanpoura,b,c    Karim Nayerniab,c    Saeed Khodayaric,d    Hamid Khodayaric,d    Marek Molcanyia     Jürgen Heschelera,b

 

aInstitute of Neurophysiology, University of Cologne, Cologne, Germany, bInternational Stem Cell Academy, Cologne/Dusseldorf, Germany, cInternational Center for Personalized Medicine, Dusseldorf, Germany, dCancer Research Center, Tehran University of Medical Sciences, Tehran, Iran

 

 

 

 

Key Words

Stem Cell Technologies • Spermatogonial Stem Cells • Injured Myocardium

 

Abstract

In the recent decades, cardiovascular diseases emerged as the major leading cause of human mortality. However, current clinical approaches still do not encompass a thorough therapeutic solution for improving heart function of the patients who suffered an extensive myocardial injury. Based on this status quo, stem cells could become a novel option, as a natural source of the new myocardium lineage cells, being capable of paracrine factors secretion, protection or even regeneration of the damaged heart muscle. Efficient stem cell-based therapy of the heart should lead to repair or/and replacement of the degenerated tissue with functional myocardial and endothelial cells. Hereon, various types of pluripotent and multipotent stem cells have been already studied in the pre-clinical and clinical settings, demonstrating their cardiomyogenic and regenerative potential. In this context, as a type of male adult stem/ progenitors, spermatogonial stem cells feature a remarkable ability for a formation of cardiovascular lineages, based on our own observations. Presented data supports the presumption, that spermatogonial stem cells not only have a suitable capacity to generate functional heart cells but can also potentially improve the function of an injured myocardium. In this review article, we first describe the essential molecular and pathophysiological mechanisms involved in the heart tissue injury. Afterwards, based on our ongoing study, we review the impact of the stem cell technologies on the regeneration therapy in cardiovascular and myocardial diseases. Particular emphasis is being put on the usability of spermatogonial stem cells in cardiac therapy.

 

 

Introduction

 

According to previously published reports, cardiovascular diseases (CVDs), such as coronary artery diseases (CAD) and/or ischemic heart disease (IHD), are known as one of the leading cause of human mortality worldwide [1, 2]. Globally, a number of 422.7 million CVD cases and 17.92 million CVD related deaths have been registered only in 2015 [3].

Extensive variety of the risk factors and pathophysiological variables are involved in development of the CVDs and subsequent heart failure (HF)  [4]. Through the HF development, a reduction in ventricular wall thickness and dilatation, as well as heart dysfunction are accrued into the injured heart areas through degeneration of cardiomyocytes (CMCs), vascular smooth muscle cells (VSMCs), and vascular endothelial cells (VECs), as the main heart lineage cells, in response to the pro-inflammatory cytokines secretion [5, 6]. While several pharmaceutical and interventional therapeutic strategies have been developed to improve CVDs patient’s heart function to some extent, clinically effective CVD treatment still remains  one of the most important challenges in the foreground of the public health [7].

During the past decades, stem cells (SCs) technology has opened a new promising perspective towards treatment of the CVDs patients, particularly IHDs, with ultimate goal to regenerate the damaged myocardium. The SCs generally play a vital role in preserving individual’s hemostasis and development during their entire lifetime [8, 9]. Technically, based on the SCs differentiation potential and origin, they are classified into the two main pluripotent stem cell populations (PSCs, iPSCs) and adult stem/progenitor cell (ASPCs) types. For the purpose of heart regeneration, different kinds of the PSCs such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) [10, 11], as well as the ASPCs such as mesenchymal stem cells (MSCs) [12, 13], CD34+ bone marrow mononuclear stem cells (BMNSCs) [14, 15], cardiac stem/progenitor cells (CSPCs) [16, 17], and spermatogonial stem cells (SSCs) [18, 19] have been already administrated in numerous of experimental and clinical studies. Although all of those administered PSCs and ASPCs have shown a capacity to generate the heart lineage cells, possible teratogenic potential of the PSCs made the direct clinical implementation difficult [20].

From the translational aspect, the concept of utilizing the stem cell technology in CVDs regeneration, consists of two main stages comprising experimental/pre-clinical and clinical phases. It can successfully help us to measure the SCs safety and recovering potential in a standard, a reliable, and a scientific framework (Fig. 1). The safety and efficacy of heart SCs-based therapeutic approaches have been proved through several experimental and clinical trials [21]. Theoretically, the utilized SCs, act first by secreting different types of paracrine/autocrine factors into the injured heart microenvironment, then by stimulating the activation and proliferation of the endogenous (native) CSPCs around the infarcted zone, and finally by potentially substituting the new cardiac lineage cells – altogether inducing protective and regenerative effects in an infarcted myocardium [22-25]. In accordance with the above-mentioned mechanisms, a significant improvement in the animal and patient’s heart functions have been already observed [23, 24]. Given the importance of this novel and challenging therapeutic approach, the present study reviews the potential and strengths of different types of the SCs, particularly SSCs, for treating and regenerating the heart, damaged in the course of different CVDs.

 

Fig. 1. Translational cascade of CVDs stem cell therapy.

 

 

Stem cells

 

Generally, SCs have referred to the parts of individual’s pluripotent, multipotent, and unipotent cellular population with the ability for proliferation, self-renewal, and differentiation into mature cells. The SCs play an essential role in organ formation and development during mammalian embryogenesis and also constitute the major platform of tissue regeneration in the adults [26]. The characteristics, biological behavior and differentiation potential of these cells have been reported in several studies [10-19].

Germline stem cells (GSCs) represent a type of testis-derived PSCs, with a self-renewal and differentiation ability [27, 28]. In males, embryonic germline stem primordial germ cells (PGCs) differentiate into spermatogonial stem cells, from which directly originate the sperm production and male fertility [27, 29]. Previous study has demonstrated that PGCs could be converted into embryonic germ cells (EGCs) under an in vitro condition [30]. A unique characteristic of EGCs germline stem cells is their pluripotent differentiation ability comparable to the ESCs, referring to the fact that germline lineage may retain this potential throughout their differentiation into SSCs. Seandel, et al. (2007) could establish a type of multipotent adult germline stem cells (maGSCs) via using a mouse neonatal testis-derived SSCs [31]. It should be noted that high differentiation capacity of the mammalian maGSCs has been demonstrated by several studies [32-34]. In fact, these stem cells, derived from adult mouse testis (GSCs), could show the characteristics ESC-pluripotency including the expression of the PSCs specific transcription factors and differentiation into three embryonic germ layers including heart lineages and functional CMCs [18, 35, 36]. Similarly, having an equal ESC property in the human GSCs has been reported by Meyer, et al. in 2010 [37]. Adult mouse testis derived SSCs, acquire the ESCs properties and can directly differentiate into derivatives of three embryonic germ layers [8]. These cells have been designated as the maGSCs. This evidence has clearly suggested the maintenance of pluripotency of the GSCs in all stages of development (Fig. 2).

 

Fig. 2. Germline stem cells developmental process and its ability to reprogram to pluripotent stem cells in all stages of development. Abbreviations: PGCs, primordial germ cells; GSCs, germline stem cells; SSCs, spermatogonial stem cells; ESC, emberionic stem cell; EG, embryonic germ cells; mGSC, multipotent germline stem cells; maGSC, multipotent adult germline stem cells.

 

Within the framework of a major discovery, PSCs has been generated in an in vitro condition by transfection of four different genes including octamer-binding protein 3/4 (OCT3/4), SRY (sex determining region Y) -box 2 (SOX2), Krüppel-like factor 4 (KLF4) and MYC (collectively referred to as OSKM) into the adult mature cells by reprogramming them to an ESC-like state, designating them as iPSCs [38, 39]. This methodology enabled the researchers to take a little biopsy and generate different types of the mature cells, ASPCs including the SSCS [40] and heart lineage cells for the regenerative medicine subjects [41]. As an imperative discovery, Guan, Nayernia, and their team (2006) could show that the human SSCs have a similar pluripotent character as the iPSCs [28]. Their transcriptional analysis has shown the SSCs could express the main pluripotency specific transcription factors including the Oct 3/4, Nanog, undifferentiated embryonic cell transcription factor 1 (Utf1), embryonic stem cell-specific gene 1(Esg1), and zinc finger protein 42 (zfi-42, or Rex1). Additionally, they proved the SSCs to encompass cardiomyogenic, myogenic, vasculogenic and also neurogenic differentiation under in vitro condition [28]. Like the iPSCs, this discovery could provide a new source of the pluripotent cell population in adults.

Multipotent cells are able to divide or self-renew successfully even in frequent passages and retain their capacity to generate a range of cell types from originating organ. It is generally thought that the adult SCs have a limited proliferation and differentiation capacity, compared to ESCs. In this manner, HSPCs are multipotent cells residing in the mammalian myocardium that are capable of self-renewing and generating vessels and heart muscle cells [17, 42, 43]. During the heart development, the creation of two different initial cardiac plates including the first heart field (FHF) and the second heart field (SHF) are managed by two primary and separate CSPCs [44, 45]. Expression of LIM-homeodomain transcription factor Islet-1 (Isl1) protein was introduced as the main marker for identification of the SHF [46]. Isl1+ cardiac progenitors constitute a significant contribution to the heart morphogenesis as they are incorporated into the right ventricle, part of the left ventricle, and also the atria development. Important role of the Isl1+ cardiac SCs in the heart development has been demonstrated by the lack of formation of the above mentioned tissue portions in Isl1 homozygous knock-out rodent heart [47]. Tracking and isolating the Isl1+ CSPCs from adult myocardium let scientific community realize the fact that SCs play an impactful character in the heart healing during entire individual's lifetime.

In contrast to the SHF, the FHF is involved exclusively in the left ventricle and portions of the atrial chambers formation. Within a retrospective clonal analysis, it has been obviously conferred that both  FHF and SHF cellular lineages are directly derived from a common primary stem cell before crescent heart formation [48]. The process of cardiac CMs, smooth muscles and VECs accruement underlies a control of several main protected and non-overlapping mechanisms. Furthermore, it should be remarked that evolution and development of the two heart fields is processed through activation of separate signaling [46, 49]. The process of the myocardium development has been clarified to start from a common primary stem cell, which further differentiate into a hierarchy of downstream cellular populations which actively facilitate the cardiogenesis in an embryo [50, 51]. An array of proteins and factors is involved in the process of heart morphogenesis, which can be used as markers at different stages of development (Fig. 3).

 

Fig. 3. A glance into the process of heart progenitor cell lineages development (From Laugwitz et al., 2008) [54] . The expression of lineage markers shown is based on the following studies: Kattman et al., 2006, Moretti et al., 2006 and Wu et al., 2006. The expression of Nkx2.5 in the precardiac mesoderm controls both endothelial and hematopoietic lineages development from the myocardial lineage [46, 54]. Abbreviations: Bry, brachyury T; MLC2a, atrial myosin light chain 2; MLC2v, ventricular myosin light chain 2; cTNT, cardiac troponin T; HCN4, hyperpolarization-activated cation channel 4; SM-MHC, smooth muscle myosin heavy chain; VE-Cadh, VE-Cadherin.

 

 

IHD pathophysiological pathways and its impact on the implanted stem cells

 

Ischemic heart disease, in particular the acute myocardial infarction (MI), emerges as the most prevalent diagnose among the CVD patients [52]. It should be noted that the myocardial injuries resulting from heart ischemia are to great extent developed by a similar pathological mechanism like MI [6, 53]. Thus, the restoration of the previously interrupted blood flow into the heart muscle and preventing further consecutive harmful cascades is known as the central common dogma of MI therapeutic strategies [54].

In the context of MI, several pathophysiological mechanisms and pathways are involved in development of an ischemic myocardial injury. Early after ischemia, changes in the cellular metabolisms, induction of the cellular oxidative stress as well as vascular cell dysfunction and eventually CMCs death are actively initiated during the infarction process. This progressive cascade, through the production of free radicals and flowing them activation of the free radical scavenging enzyme [55, 56], storage of intracellular calcium [6], and also decreasing the ischemic cell’s level of adenosine triphosphate (ATP) [57] can induce tissue and heart lineage cells degeneration. In this regard, generation of high level of reactive oxygen species (ROS) and reactive nitrogen species (RNS) from the stressed cell’s mitochondria, well known as free radicals, directly promotes some of the harmful cascades in CMCs [57]. It has been proven that several apoptosis-related cascades are promoted through the accumulation of ROS and RNS in the heart cells. Functionally, these reactive species directly lead to switch-on of the caspase-3 related apoptosis, increased activation of pro-apoptotic Bcl-2-associated X (Bax) protein, and repression of the anti-apoptotic B-cell lymphoma 2 (Bcl-2) protein activity as well [58].

From an immunological point of view, through promoting the expression of an array of pro-inflammatory factors and cytokines such as tumor necrotic factor-α (TNF- α), interleukin-1 (IL-1) family, and interleukin-6 (IL-6) free radicals also indirectly contribute to the cellular death in the infarcted zone [59]. Moreover, the expression and activation of main death ligand receptors including toll-like receptors (TLR), apoptosis antigen-1 (APO-1 or FAS), and also TNF-related apoptosis-inducing ligand-receptor (TRAIL-R) are the other targets of the free radicals in this pathological cascade [6, 60] (Fig. 4). Following this process, by inducing the expression of different integral membrane proteins such as cysteineXcysteine (CXC) and cysteine-cysteine (CC) chemokine, free radicals production promotes the infiltration of inflammatory leukocytes and thus contributes to the establishment of a harmful inflammatory myocardium microenvironment within the infarcted heart segment [61, 62].

 

Fig. 4. A schematic representation of the cellular and molecular mechanisms of the cardiomyocytes death through the heart injury. Endogenic ROS production acquires early after injuries. Cytoplasmic ROS via changes mPTPs opening on the surface of mitochondria, releases of the cyto c into the cytoplasm, and activation and intra nucleus accumulation of Nf-kB through MAPK signaling stimulation leading to the cardiomyocyte apoptosis and necrosis. Beside, stimulation of the TNF-R1/2, IL-1R, and TRAIL-R as the death ligands by activation of the caspases cascades have a central role in developing the myocardium injuries. Abbreviations: Cyt-C: cytochrome -C, ERK: extracellular signal-regulated kinases, Fas: apoptosis antigen 1 (APO-1 or APT), IL-1R: interleukin-1 receptor, IL-1β: interleukin-1 β , MAPK: mitogen-activated protein kinase, NFκB: nuclear factor kappa-light-chain-enhancer of activated B cells , ROS: reactive oxygen spices, SCs: stem cells, TNF-R: tumor necrosis factor receptor, TNF-α: tumor necrosis factor- α, TRAIL: TNF-related apoptosis-inducing ligand, and TRAIL-R: TNF-related apoptosis-inducing receptor.

 

In a post-infarcted myocardium, circulating blood monocytes which are known to be the primary responder to the chemotactic factors, extensively infiltrate the infarcted zone and immediately differentiate into the mature macrophages under the stimulation of the inflammatory-microenvironment-associated factors [63]. The macrophages have a pleiotropic role within the progress of myocardial pathophysiologic and also regenerative response after the infarction. These cells release various pro-inflammatory and anti-inflammatory cytokines, inducing their complex pleiotropic effects on the healing heart tissue [64-67]. Macrophages, classified into M1 and M2 class, possess a meaningful role in removing the dead cells and additionally interact with cardiac fibrosis [63, 64, 68]. In the course of MI, the macrophage activation takes place first through the M1 pathway and then shifts to the M2 pathway within the differentiation of the infiltrating monocytes [69]. Briefly, under the stimulation and by expression of some of the main chemokine family such as C-C motif chemokine ligand 2 (CCL2) and C-X3-C motif chemokine ligand 1 (CX3CL1) on the infarcted heart cell's surface, Ly-6Chigh monocytes infiltration, polarization, and differentiation takes place in the injured myocardial microenvironment. It seems that M1 macrophages are generated by the Ly-6Chigh monocytes differentiation [70]. It has been shown that the M1 macrophages, unlike the M2 type, play a harmful pro-inflammatory role  through secreting high levels of TNF-α, IL-1β, IL-6, and also interferon gamma (INF-γ) into the injured myocardial environment [63, 64, 71, 72]. Switching the expression of CCL2 to CX3CL1, in the heart healing phases, causes the recruitment and the increase in the number of M2 macrophages through differentiation of the Ly-6CLow monocytes [63, 64, 71, 72]. M2 macrophages can play a vital role in promoting revascularization, regeneration, and remodeling mechanisms in the infarcted healing heart via secretion of several anti-inflammatory cytokines and growth factors such as transforming growth factor- β (TGF-β), IL-10, vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (b-FGF) [63, 64, 71-73].  Previously published experimental studies demonstrate that the macrophages infiltrating the injured myocardium, besides the expression of pro-inflammatory cytokines, can actively release wingless/integrated (Wnt) protein, a family of 19 secreted glycoproteins, into the infarcted zones. In course of this process, activation of Wnt/b-catenin cascade intensifies cardiac damage and disrupts the myocardium hilling [74-76]. Additionally, Palevski et al. (2017) observed that “loss of macrophage Wnt secretion improves remodeling and function after myocardial infarction in mice” [63]. In addition to macrophages, infiltrating CD4+ T cells and myeloid cells can promote cardiac cell apoptosis through the secretion of different pro-apoptotic cytokines [77, 78].

In the progress of the secretion of pro-inflammatory cytokines by infiltrating leukocytes, the myocardial cell death is extensively induced through stimulation of the death ligands and activation of cellular-death specific signaling pathways (Fig. 4). It has been proven that, nuclear factor- kappa B (NF-κB) is a main target of TNF-R (TNF-α receptor), in addition to IL-1βR (IL-1β receptor) and other death ligands such as FAS and TRAIL-R. Activation and intra-nuclear accumulation of the NF-κB plays a critical role in expression of pro-apoptotic genes and has a positive feedback on pro-inflammatory cytokines secretion [79, 80].

Whilst the cytotoxic effects of the TNF- α and IL-1 family, as well as their related mechanisms on the CMCs are carefully proven, the IL-6 affected mechanisms on the CMCs survival are not fully clarified, yet. The IL-6 and IL-6 family members play both pro- and anti-inflammatory roles in a post-infarcted heart [2, 13, 81, 82]. In some studies, it has been observed that the IL-6, as a pro-inflammatory cytokine, can induce CMCs apoptosis and cause the thinning of myocardial wall through stimulation of the NF-κB signaling pathway [5, 8, 10, 11, 17, 19, 30, 33-35, 37, 39, 41, 63, 64, 68, 73-75, 83-87].

It has been recognized that the regeneration potential of implanted SCs is directly affected by the noted inflammatory microenvironment creating early after MI. Based on some observations, into the post-infarcted heart regions, about 90% of all types of injected cells degenerate just during 4 days following transplantation [81, 88]. Through activation of NF-κB signaling pathway, it seems that promotion of the TNF-R and the IL-1βR related cascades play a vital role in the implanted SCs survival [89, 90]. It has been also shown that, into a post-infarcted heart, TNF-α and IFN-γ networks can synergistically enhance the autophagy and apoptosis cell death process through stimulating ROS/ mitogen-activated protein kinase 1/3 (ERK) pathway, inducing Bcl-2-homology (BH)-3 domain only protein (Beclin-1) gene expression, and inhibiting anti-apoptotic B-cell lymphoma 2 (Bcl-2) expression on the SCs [84]. Moreover, other observations have obviously demonstrated that the TNF-α and its mediated signaling pathways inhibit the heart lineage differentiation of the both PSCs and ASPCs. Flowing this mentioned mechanism, generation of neuroaderenergic-like fate can significantly increase through differentiation of the injected stem cells [91, 92]. In agreement with this hypothesis, it is cleared activation of both TNF-R1 and TNF-R2 into the infarcted heart tissue can promote myocardium innate regeneration responses through decreasing the CSPCs differentiation potentials and proliferation through stimulation of the NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways around the infarcted zone [92].

 

 

Cellular therapy approaches for cardiovascular regeneration

 

For a clinically effective regeneration in patients with CVDs, a suitable and a safe source SCs with a high cardiomyogenic potential is greatly needed. Till now, beneficial effects and manifold positive feedback on different types of PSCs and ASPCs implemented in CVDs patients and animal models have been reported in a line-up of preclinical and clinical studies [93-95]. Over the past few decades, main portion of our efforts has been focused on discovery of the SCs physiological and biological behavior, as well as their capacity to generate the functional heart cells. Moreover, we could successfully report stem cell’s outstanding potential for regenerating and recovering the function of an injured heart in the experimental animal models.

In the field of PSCs, our previous studies have carefully outlined the cardiomyogenic and angiogenic potential of ESCs and iPSCs in both in vitro and in vivo conditions [96-103]. Regrettably, the immunogenicity issues and especially the teratogenic potential of undifferentiated PSCs hindered the use of undifferentiated ESCs and iPSCs in the clinical phases [20, 104-106]. However, the application of the ex vivo generated CMCs from the PSCs not only could be a safe strategy but also seemed to be an effective method for replacing lost myocytes in the context of heart SCs therapy.

Within a translational view, pre-clinical studies on the IHD animal models have shown a significant efficacy during intramyocardial implantation of undifferentiated hESCs and hiPSCs [87, 107-109]. The current researches are trying to display the iPSCs as the main PSCs candidate in order to translational and clinical goals. However, there still some challenges to achieve this purpose. Ethically, direct application of the PSCs because of their unpleasant behaviors into the patient's myocardium is too controversial.  Nevertheless, it has been clime that application of the PSCs would be safer in the case of using ex vivo generated mature CMCs.

According to several observations, the iPSCs have been introduced as a cost-effective, reliable, and efficient source of the PSCs in compared with the ESCs although both of them are similar in morphology, phenotype, and specific cell marker. Having a complete histocompatibility of the iPSCs with the patients and lifetime availability to the production of this PSCs, besides their great cardiogenic differentiation potential make them as a powerful SCs for the future heart regeneration goals [110, 111]. In contrast, some of the investigations have already realized that the iPSCs normal behavior and differentiation potential can be affected by an array of genetic and epigenetic alterations during the reprogramming [112]. Method for inducing pluripotency, type of the utilized somatic cell for iPSC preparation, and the preformed material to the cell isolation and culture are the other well-known effectors of the iPSCs normal behavior [113].

Despite the above stated challenges, the potential of the PSCs to the regenerate heart injuries has been proven so far in experimental set-up [114]. Hodgson et al. (2004) observed a significant therapeutic efficacy and the improvement of the rat’s heart function after application of undifferentiated human ESCs into the infarcted areas of laboratory rodents. They also remarked that this notable efficacy was related to the differentiation of implanted SCs  into heart lineage cells [10]. Furthermore, a similar result has been obtained in another study, after administration of undifferentiated human ESCs for improving the heart function in the experimental animal model of myocardial injury [115].  Similarly to ESCs, several observations have proven that using undifferentiated iPSCs would alike be a powerful tool for regenerating the injured myocardium, as well [10, 116-118].

In order to understand the therapeutic benefits of the ex vivo generated CMCs, within the concept of an orthotopic intramyocardial implantation of the murine iPSC-derived CMCs (iPSCs- CMCs) in a syngeneic mice model of cryoinfarcted heart injury, we have tracked implanted long-term surviving cells and importantly observed a significant improvement in the animals heart function after the cell implantation [119]. Our results were supported through another observation in context of intramyocardial injection of human iPSCs-CMCs in non-human primate model of MI [114]. Furthermore, we have been able to improve the iPSCs-CMCs regenerative function based on implementation of a new kind of gelatin microspheres. Notably, we concluded, “intramyocardial transfer of iPSCs-CMCs bound to gelatin microspheres enhances cell retention in the early stage after transplantation significantly” [120].

Because of the main fundamental and developmental difference between the ESCs and iPSCs, it seems there will be some functional discrepancy between CMCs generated out of these two respective cell types. To evaluate and compare the action potentials of the ex vivo generated CMCs from the ESCs and the iPSCs with a mature CMCs in an experimental study, we have first functionally examined and then compared the activity of cardiac-specific voltage-gated Na+, Ca2+, and K+ channels in the ESCs-CMCs and iPSCs-CMCs with the mature CMCs counterparts at the early and late differentiation stages. Our observation has demonstrated that the CMCs generated from the iPSCs had a delayed action against the physiological stimulators compared with ESCs-CMCs and mature harvested CMCs. We also concluded that this notable difference might be related to the incomplete reprogramming of the iPSCs [111, 121, 122].

During the past decades, considerable efforts were undertaken to launch regeneration strategies based on the implementation of autologous and/or identical adult SPCs, in particular MSCs and BMNSCs. Easier access and increased safety, compared to PSCs, in addition to their high proliferative, differentiative and paracrine/ autocrine secretory potential make them the most favorite cells type for the CVDs regeneration concepts in the clinical phases [83, 123-128]. Chen et al. (2004) performed an intracoronary injection of the 8-10 × 109 (cell/ ml) autologous bone marrow-derived MSCs in the acute MI and observed a significant improvement in the patient left ventricular (LV) functions in a time span of several months after the cell therapy [124]. Among all of these interesting reports, including our own observations, some unreliable differentiation processes of the implanted adult SPCs have been observed [121]. Based on our evidence, it should be noted that the bone marrow-derived MSCs implanted into the murine infarcted myocardium may acquire incorrect fates of differentiation such as into the osteocytes, in addition to the heart lineage cells [121]. Our finding clearly presented the fact that the utilized MSCs for the heart regeneration were not as safe as initially considered.

Apart from MSCs, intracoronary implantation of the autologous BMNSCs had a safe  suitable,and remarkable therapeutic response in the patients with heart disorder and infarction [126-128]. It seems that this noted regeneration has largely resulted from an angiogenic response and revascularization of the damaged tissues via implanted SCs stimulation and differentiation [86, 129]. Unfortunately, many of the clinical MSCs and BMNSCs-based therapies have not had a long-term efficacy to improve the patient's heart function, as demonstrated by the lately published meta-analysis [130].

CSPCs were introduced as a convenient adult SCs option for regenerating the injured myocardium [16, 131-133]. In both human and rodent heart, the CSPCs were classified by expression of the cardiac-specific transcription factors such as GATA Binding Protein-4 (GATA-4), NK2 Homeobox 5 (Nkx-2.5), and myocyte enhancer factor-2 (MEF-2). C-kit+ cardiac stem cells (CSCs), Sca-1+ CSCs, cardiosphere-derived SPCs; side population CSCs are some of the well-known populations of the mammalians heart SCs [43, 53]. Differentiation potential into all heart lineage cells, promoting the cardioprotective signaling via secretion of various kinds of paracrine/ autocrine factors besides their high ability to survive in the damaged myocardial microenvironment made them another useful stem cell type for managing the heart regeneration [134, 135]. Practically, regenerative response of the CSPCs is being controlled by regulation of specific mechanisms and signaling pathways. Inside of the infarcted heart microenvironment, increasing expression of stem cell factor (SCF) from the injured CMCs stimulates heart's endogenous c-kit+ CSPCs activity through activation of Wnt/ β-catenin cascade [136]. Furthermore, in response to the activation of phosphoinositide 3-kinase/ protein kinase B (PI3K/ Akt) and Notch pathways, CSPCs can raise their regenerative capacity [137-139]. It seems that the activation of the endogenous innate CSPCs can be an attractive target for developing new CVDs therapeutic agents. Study on the experimental mice model of MI has shown that systemic administration of extracellular high-mobility group box 1 protein (HMGB1),could dramatically induce a regenerative response and a significant improvement of the mice heart function through a stimulation of the C-kit+ CSCs located around the infarction zone [140].

Among various types of the heart SPCs, cardiosphere-derived SPCs are the most trialed cell type in different clinical studies [16, 82, 122, 133]. According to a clinical trial performed by Malliaras, et al. (2014), in response to intracoronary implantation of the autologous cardiosphere-derived SPCs in the patients with acute MI, a significant improvements in the patient's infarcted-segment regional function, the mass of scar size, and also the viable myocardium were observed [133]. Similarly, an improvement in the viable myocardium after the intracoronary implantation of cardiosphere-derived SPCs was proven in other clinical studies [16, 82, 122].

Number of studies have also described a possible potential of testis-derived SPCs to regenerate the injured heart [8, 125-129]. Through one of our studies in 2006, we have successfully identified and isolated a new sort of SCs from the adult mammalian testis with the pluripotent features and high potential to generate different mature cellular lineages. Results of our study suggested the testis as a new source of the PSCs for organ-regenerating strategies [4]. In this regard, our previous research has introduced a fact that the SSCs have a natural potential for creating the functional CMCs and may be able to regenerate lost CMCs [14]. The results of our study have been supported by additional in vitro experiences.

 

 

Spermatogonial stem cells possible impacts for heart regeneration

 

Testis-derived SCs inclosing the maGSCs are a unique and extremely resistant cellular population against numerous types of stresses. A primary study by Meistrich, et al. (1974) has shown an incredible regenerative behavior of the mice SSCs after the animals were exposed to an ionizing radiation [142]. Furthermore, later published studies have also reported that the rate of SSCs survival, proliferation, and migration significantly increased in response to such pathophysiological and physical stress like inflammation and gamma irradiation [85, 142-144]. Interestingly, it has been proven that the testis-derived SCs are resistant to the harmful cellular factors and cascades such as free radicals mediated pathways. In this context, according to an experimental study, Morimoto, et al. (2013) have explained that the creation of ROS and the activation of its related signaling pathways in mice SSCs not only had no apoptosis-inducing effects, but in contrast, proliferative and a self-renewing response in the SSCs were observed to be mediated via activation of NADPH oxidase 1 (Nox1) enzym. The results of these observations determined testis-derived SCs to be potentially highly suitable for regeneration of the damaged tissues with an extensive harmful inflammatory environment.

To verify the hypothesis about maGSCs being beneficial in heart regeneration strategies, we launched one of our current ongoing in vivo trials and carefully evaluated the potential of murine maGSCs in the regeneration of rodent heart. In this study, we have clearly observed that intramyocardial implantation of the mice maGSCs into the healthy and also infarcted myocardium elicited a safe and efficient therapeutic response.  Here, administrated maGSCs into a healthy contracting myocardium have shown an acceptable survival besides a specific differentiation into heart lineage cells (Fig. 5).

 

Fig. 5. Assessing behavior of murine maGSC into the healthy myocardium. Tracing of the I) implanted pre-differentiated maGSC (red labeled cells) and implanted cell nucleus (H & E), II) Stemness-related transcriptional factor (Oct4) and proliferation specific marker (II-B: Ki67), III) Vasculogenic specific markers SMA (III-A) and VWF (III-B), and IV) Cardiomyogenic specific markers βTubulin (IV-A) and Nestin (IV-B) into the heart muscle. Abbreviations: maGSC, multipotent adult germline stem cells; H&E, hematoxylin and eosinOct4, octamer-binding transcription factor 4; Ki67, MKI67; VWF, von Willebrand Factor; and SMA, smooth muscle alpha actin.

 

In case of testicular SCs, we believe that the post-infarcted myocardial inflammatory microenvironment not only lacks a disruptive effect on this utilized maGSCs population but also seems to be a stimulating factor for the testicular SCs survival, proliferation, and differentiation. Accordingly, in a C57BL/6 mice model of MI, murine maGSCs orthotopic implantation into infarcted heart areas showed a significant potential for regeneration of the lost heart tissue through a successful differentiation. Besides a significant survival, our immunohistochemical assessments indicated that the implanted maGSCs could create a large number of CMCs and VECs within 4 weeks after application into the post-infarcted heart (Fig. 6). Moreover, in none of maGSCs-treated mice, any signs of malignancy resulting from aberrant differentiation of implanted SCs could be observed. The results from our pre-clinical study, in line with our other previous experiences, outline the SSCs to be potentially highly suitable for application and translation into future clinical studies for CVD-regeneration in a safe and efficient way.

 

Fig. 6. Assessing behavior of murine maGSC into the infarcted myocardium. Tracing of the I) implanted pre-differentiated maGSC (red labeled cells), II) Stemness-related transcriptional factor (Oct4) and proliferation specific marker (II-B: Ki67), III) Vasculogenic specific markers SMA (III-A) and VWF (III-B), and IV) Cardiomyogenic specific markers βTubulin (IV-A) and Nestin (IV-B) into the infarcted zoon. Abbreviations: maGSC, multipotent adult germline stem cells; Oct4, octamer-binding transcription factor 4; Ki67, MKI67; VWF, von Willebrand Factor; and SMA, smooth muscle alpha actin.

 

 

Conclusion

 

More than two decades have passed since the first scientific efforts for stem cell based regeneration of injured heart were launched. Although, a complete and effective repair in cell-treated patients with myocardial damage could not be unequivocally demonstrated, it is believed that future studies will be able to introduce more efficient technologies for the rehabilitation of the damaged myocardium by means of stem cell therapy. To achieve more effective cardiac regeneration policies, based on application of the stem cell technology, broadening our horizons on pathophysiologic mechanisms of the myocardial damage as well as stem cell behavior and its associated molecular pathways, plays an inevitable role. Accurate and reliable implementation of alternative stem cell sources including the GSCs and SSCs, shown to possess a suitable potential for the generation of the CMCs and endothelial cells (see our own above cited observations), can offer an opportunity with a higher potential to replace the previously lost heart cells.

In addition to undisputable cardiogenic differentiation potential of SSCs, these stem cells may hence encompass a translational capability in heart regeneration strategies, as supported by following propositions: I. SSCs, as a type of PSCs-like cells, have not shown as high tumorigenicity and immunogenicity as their ESCs/iPSCs counterparts in the previous experimental set-ups, II. SSCs feature a low sensitivity and a high resistance to the adverse effects of detrimental local microenvironment of the infarcted heart, III. SSCs present a readily accessible resource, as their isolation and large-scale culturing is possible during the male's whole lifetime. The above stated paradigms suggest that implementation of SSCs might become a part of solution to overcoming barriers of the heart stem cell therapy. Further studies will be essential for optimizing and translating experimental accomplishments into stem cell based regeneration therapy of the heart diseases in clinical routine. Despite previous achievements, further progressive questions have to be addressed on the way towards effective translation of heart stem cell therapy. Can a personalized medicine play an influential role in management of effective stem cell therapy? To which extent does patient’s specific geno/phenotype determine their characteristics of myocardial ischemia and consecutive host’s response to the SC therapeutics? Is there a way to implement personalized medicine in order to optimize the modalities of stem cell therapy, based on patient's unique myocardial specifications? Considering previous experiences from personalized cancer therapy, the implementation of personalized methodologies might be a smart leap towards solving some of the obstacles along the way of stem cell based heart regeneration.

 

 

Disclosure Statement

 

The authors declare to have no competing interests.

 

 

References

 

1 Andersson C, Vasan RS: Epidemiology of cardiovascular disease in young individuals. Nat Rev Cardiol 2018;15:230.
https://doi.org/10.1038/nrcardio.2017.154

 

2 Jagtap S, Meganathan K, Wagh V, Winkler J, Hescheler J, Sachinidis A: Chemoprotective mechanism of the natural compounds, epigallocatechin-3-O-gallate, quercetin and curcumin against cancer and cardiovascular diseases. Curr Med Chem 2009;16:1451-1462.
https://doi.org/10.2174/092986709787909578

 

3 Roth GA, Johnson C, Abajobir A, Abd-Allah F, Abera SF, Abyu G, Ahmed M, Aksut B, Alam T, Alam K: Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015. J Am Coll Cardiol 2017;70:1-25.

 

4 Veerbeek JH, Hermes W, Breimer AY, van Rijn BB, Koenen SV, Mol BW, Franx A, de Groot CJ, Koster MP: Cardiovascular disease risk factors after early-onset preeclampsia, late-onset preeclampsia, and pregnancy-induced hypertension. Hypertension 2015;65:600-606.
https://doi.org/10.1161/HYPERTENSIONAHA.114.04850

 

5 Gong DM, Zhang YL, Chen DY, Hong LJ, Han F, Liu QB, Jiang JJ, Lu YM: Endothelial GPR124 Exaggerates the Pathogenesis of Atherosclerosis by Activating Inflammation. Cell Physiol Biochem 2018;45:547-557.
https://doi.org/10.1159/000487032

 

6 Bagheri F, Khori V, Alizadeh AM, Khalighfard S, Khodayari S, Khodayari H: Reactive oxygen species-mediated cardiac-reperfusion injury: mechanisms and therapies. Life Sci 2016;165:43-55.
https://doi.org/10.1016/j.lfs.2016.09.013

 

7 O'flaherty M, Buchan I, Capewell S: Contributions of treatment and lifestyle to declining CVD mortality: why have CVD mortality rates declined so much since the 1960s?. Heart 2013; 99:159-162.
https://doi.org/10.1136/heartjnl-2012-302300

 

8 Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH, Nolte J, Wolf F, Li M, Engel W: Pluripotency of spermatogonial stem cells from adult mouse testis. Nature 2006;440:1199.
https://doi.org/10.1038/nature04697

 

9 Amidi F, Nejad NA, Hoseini MA, Nayernia K, Mazaheri Z, Yamini N, Saeednia S: In vitro differentiation process of human Wharton's jelly mesenchymal stem cells to male germ cells in the presence of gonadal and non-gonadal conditioned media with retinoic acid. In Vitro Cell Dev Biol Anim 2015;51:1093-1101.
https://doi.org/10.1007/s11626-015-9929-4

 

10 Hodgson DM, Behfar A, Zingman LV, Kane GC, Perez-Terzic C, Alekseev AE, Pucéat M, Terzic A: Stable benefit of embryonic stem cell therapy in myocardial infarction. Am J Physiol Heart Circ Physiol 2004;287:H471-H479.
https://doi.org/10.1152/ajpheart.01247.2003

 

11 Iglesias-García O, Baumgartner S, Macrí-Pellizzeri L, Rodriguez-Madoz JR, Abizanda G, Guruceaga E, Albiasu E, Corbacho D, Benavides-Vallve C, Soriano-Navarro M: Neuregulin-1β induces mature ventricular cardiac differentiation from induced pluripotent stem cells contributing to cardiac tissue repair. Stem Cells Dev 2014;24:484-496.
https://doi.org/10.1089/scd.2014.0211

 

12 Zhang J, Wu Y, Chen A, Zhao Q: Mesenchymal stem cells promote cardiac muscle repair via enhanced neovascularization. Cell Physiol Biochem 2015;35:1219-1229.
https://doi.org/10.1159/000373945

 

13 Miyahara Y, Nagaya N, Kataoka M, Yanagawa B, Tanaka K, Hao H, Ishino K, Ishida H, Shimizu T, Kangawa K: Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat Med 2006;12:459.
https://doi.org/10.1038/nm1391

 

14 Kawamoto A, Iwasaki H, Kusano K, Murayama T, Oyamada A, Silver M, Hulbert C, Gavin M, Hanley A, Ma H: CD34-positive cells exhibit increased potency and safety for therapeutic neovascularization after myocardial infarction compared with total mononuclear cells. Circulation 2006;114:2163-2169.
https://doi.org/10.1161/CIRCULATIONAHA.106.644518

 

15 Strauer BE, Brehm M, Zeus T, Köstering M, Hernandez A, Sorg RdV, Kögler G, Wernet P: Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation 2002;106:1913-1918.
https://doi.org/10.1161/01.CIR.0000034046.87607.1C

 

16 Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, Czer LS, Marbán L, Mendizabal A, Johnston PV: Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet 2012;379:895-904.
https://doi.org/10.1016/S0140-6736(12)60195-0

 

17 Li W, Lu Y, Han R, Yue Q, Song X, Wang F, Wu R, Hou F, Yang L, Xu L: Gremlin2 Regulates the Differentiation and Function of Cardiac Progenitor Cells via the Notch Signaling Pathway. Cell Physiol Biochem 2018;47:579-589.
https://doi.org/10.1159/000490012

 

18 Guan K, Wagner S, Unsöld B, Maier LS, Kaiser D, Hemmerlein B, Nayernia K, Engel W, Hasenfuss G: Generation of functional cardiomyocytes from adult mouse spermatogonial stem cells. Circ Res 2007;100:1615-1625.
https://doi.org/10.1161/01.RES.0000269182.22798.d9

 

19 Fagoonee S, Pellicano R, Silengo L, Altruda F: Potential applications of germline cell-derived pluripotent stem cells in organ regeneration. Organogenesis 2011;7:116-122.
https://doi.org/10.4161/org.7.2.16284

 

20 Ahmed RP, Ashraf M, Buccini S, Shujia J, Haider HK: Cardiac tumorgenic potential of induced pluripotent stem cells in an immunocompetent host with myocardial infarction. Regen Med 2011;6:171-178.
https://doi.org/10.2217/rme.10.103

 

21 Menasché P: Stem cell therapy for heart failure: are arrhythmias a real safety concern? Circulation 2009;119:2735-2740.
https://doi.org/10.1161/CIRCULATIONAHA.108.812693

 

22 Ventura C, Cantoni S, Bianchi F, Lionetti V, Cavallini C, Scarlata I, Foroni L, Maioli M, Bonsi L, Alviano F: Hyaluronan mixed esters of butyric and retinoic acid drive cardiac and endothelial fate in term placenta human mesenchymal stem cells and enhance cardiac repair in infarcted rat hearts. J Biol Chem 2007;282:14243-14252.
https://doi.org/10.1074/jbc.M609350200

 

23 Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, Pocius J, Michael LH, Behringer RR, Garry DJ: Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A 2003;100:12313-12318.
https://doi.org/10.1073/pnas.2132126100

 

24 Gnecchi M, He H, Noiseux N, Liang OD, Zhang L, Morello F, Mu H, Melo LG, Pratt RE, Ingwall JS: Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J 2006;20:661-669.
https://doi.org/10.1096/fj.05-5211com

 

25 Zhang M, Mal N, Kiedrowski M, Chacko M, Askari AT, Popovic ZB, Koc ON, Penn MS: SDF-1 expression by mesenchymal stem cells results in trophic support of cardiac myocytes after myocardial infarction. FASEB J 2007;21:3197-3207.
https://doi.org/10.1096/fj.06-6558com

 

26 Bankaitis ED, Ha A, Kuo CJ, Magness ST: Reserve Stem Cells in Intestinal Homeostasis and Injury. Gastroenterology 2018;155:1348-1361.
https://doi.org/10.1053/j.gastro.2018.08.016

 

27 Spradling A, Fuller MT, Braun RE, Yoshida S: Germline stem cells. Cold Spring Harb Perspect Biol 2011;3:a002642.
https://doi.org/10.1101/cshperspect.a002642

 

28 Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH, Nolte J, Wolf F, Li M, Engel W, Hasenfuss G: Pluripotency of spermatogonial stem cells from adult mouse testis. Nature 2006;440:1199-1203.
https://doi.org/10.1038/nature04697

 

29 Caneguim BH, Beltrame FL, da Luz JS, Valentini SR, Cerri PS, Sasso-Cerri E: Primordial germ cells (spermatogonial stem cells) of bullfrogs express sex hormone-binding globulin and steroid receptors during seasonal spermatogenesis. Cells Tissues Organs 2013;197:136-144.
https://doi.org/10.1159/000341517

 

30 Durcova-Hills G, Surani A: Reprogramming primordial germ cells (PGC) to embryonic germ (EG) cells. Curr Protoc Stem Cell Biol 2008; DOI.10.1002/9780470151808.sc01a03s5.
https://doi.org/10.1002/9780470151808.sc01a03s5

 

31 Seandel M, James D, Shmelkov SV, Falciatori I, Kim J, Chavala S, Scherr DS, Zhang F, Torres R, Gale NW, Yancopoulos GD, Murphy A, Valenzuela DM, Hobbs RM, Pandolfi PP, Rafii S: Generation of functional multipotent adult stem cells from GPR125+ germline progenitors. Nature 2007;449:346-350.
https://doi.org/10.1038/nature06129

 

32 Nikolic A, Volarevic V, Armstrong L, Lako M, Stojkovic M: Primordial Germ Cells: Current Knowledge and Perspectives. Stem Cells Int 2016;2016:1741072.
https://doi.org/10.1155/2016/1741072

 

33 Drusenheimer N, Wulf G, Nolte J, Lee JH, Dev A, Dressel R, Gromoll J, Schmidtke J, Engel W, Nayernia K: Putative human male germ cells from bone marrow stem cells. Soc Reprod Fertil Suppl 2007;63:69-76.

 

34 Mardanpour P, Guan K, Nolte J, Lee JH, Hasenfuss G, Engel W, Nayernia K: Potency of germ cells and its relevance for regenerative medicine. J Anat 2008;213:26-29.
https://doi.org/10.1111/j.1469-7580.2008.00930.x

 

35 Kim S, Izpisua Belmonte JC: Pluripotency of male germline stem cells. Mol Cell 2011;32:113-121.
https://doi.org/10.1007/s10059-011-1024-4

 

36 Niu Z, Wu S, Wu C, Li N, Zhu H, Liu W, Hua J: Multipotent male germline stem cells (mGSCs) from neonate porcine testis. Braz Arch Biol Technol 2016; DOI:10.1590/1678-4324-2016150449.
https://doi.org/10.1590/1678-4324-2016150449

 

37 Meyer S, Nolte J, Opitz L, Salinas-Riester G, Engel W: Pluripotent embryonic stem cells and multipotent adult germline stem cells reveal similar transcriptomes including pluripotency-related genes. Mol Hum Reprod 2010;16:846-855.
https://doi.org/10.1093/molehr/gaq060

 

38 Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin, II, Thomson JA: Induced pluripotent stem cell lines derived from human somatic cells. Science 2007;318:1917-1920.
https://doi.org/10.1126/science.1151526

 

39 Hamalainen RH: Induced pluripotent stem cell-derived models for mtDNA diseases. Methods Enzymol 2014;547:399-415.
https://doi.org/10.1016/B978-0-12-801415-8.00019-9

 

40 Zhu Y, Hu HL, Li P, Yang S, Zhang W, Ding H, Tian RH, Ning Y, Zhang LL, Guo XZ, Shi ZP, Li Z, He Z: Generation of male germ cells from induced pluripotent stem cells (iPS cells): an in vitro and in vivo study. Asian J Androl 2012;14:574-579.
https://doi.org/10.1038/aja.2012.3

 

41 Burridge PW, Li YF: Human induced plurip otent stem cell-derived cardiomyocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat Med 2016;22:547-556.
https://doi.org/10.1038/nm.4087

 

42 Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K: Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003;114:763-776.
https://doi.org/10.1016/S0092-8674(03)00687-1

 

43 Khodayari S, Khodayari H, Alizadeh AM: A glance into the future cardiac stem cells. Tehran Univ Med J 2016;74:223-235.

 

44 Fernandez-Valdivia R, Takeuchi H, Samarghandi A, Lopez M, Leonardi J, Haltiwanger RS, Jafar-Nejad H: Regulation of mammalian Notch signaling and embryonic development by the protein O-glucosyltransferase Rumi. Development 2011;138:1925-1934.
https://doi.org/10.1242/dev.060020

 

45 Cohen ED, Miller MF, Wang Z, Moon RT, Morrisey EE: Wnt5a and Wnt11 are essential for second heart field progenitor development. Development 2012;139:1931-1940.
https://doi.org/10.1242/dev.069377

 

46 Moretti A, Caron L, Nakano A, Lam JT, Bernshausen A, Chen Y, Qyang Y, Bu L, Sasaki M, Martin-Puig S: Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell 2006;127:1151-1165.
https://doi.org/10.1016/j.cell.2006.10.029

 

47 Cai CL, Liang X, Shi Y, Chu PH, Pfaff SL, Chen J, Evans S: Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev Cell 2003;5:877-889.
https://doi.org/10.1016/S1534-5807(03)00363-0

 

48 Haddad F, Hunt SA, Rosenthal DN, Murphy DJ: Right ventricular function in cardiovascular disease, part I: anatomy, physiology, aging, and functional assessment of the right ventricle. Circulation 2008;117:1436-1448.
https://doi.org/10.1161/CIRCULATIONAHA.107.653576

 

49 Kovacic JC, Mercader N, Torres M, Boehm M, Fuster V: Epithelial-to-mesenchymal and endothelial-to-mesenchymal transition: from cardiovascular development to disease. Circulation 2012;125:1795-1808.
https://doi.org/10.1161/CIRCULATIONAHA.111.040352

 

50 Meilhac SM, Lescroart F, Blanpain C, Buckingham ME: Cardiac cell lineages that form the heart. Cold Spring Harb Perspect Med 2014;4:a013888.
https://doi.org/10.1101/cshperspect.a013888

 

51 Laugwitz KL, Moretti A, Caron L, Nakano A, Chien KR: Islet1 cardiovascular progenitors: a single source for heart lineages? Development 2008;135:193-205.
https://doi.org/10.1242/dev.001883

 

52 Al-Shaar L, Li Y, Rimm E, Manson JE, Hu F, Willett W: Abstract MP27: Weight Change, BMI, and Mortality Among Survivors of Myocardial Infarction: Analysis of Two Prospective US Cohort Studies. Circulation 2018;137:AMP27.

 

53 Faghihi M, Alizadeh AM, Khori V, Latifpour M, Khodayari S: The role of nitric oxide, reactive oxygen species, and protein kinase C in oxytocin-induced cardioprotection in ischemic rat heart. Peptides 2012;37:314-319.
https://doi.org/10.1016/j.peptides.2012.08.001

 

54 Engstrøm T, Kelbæk H, Helqvist S, Høfsten DE, Kløvgaard L, Holmvang L, Jørgensen E, Pedersen F, Saunamäki K, Clemmensen P: Complete revascularisation versus treatment of the culprit lesion only in patients with ST-segment elevation myocardial infarction and multivessel disease (DANAMI-3-PRIMULTI): an open-label, randomised controlled trial. Lancet 2015;386:665-671.
https://doi.org/10.1016/S0140-6736(15)60648-1

 

55 Chen X, Gu M, Zhao X, Zheng X, Qin Y, You X: Deterioration of cardiac function after acute myocardial infarction is prevented by transplantation of modified endothelial progenitor cells overexpressing endothelial NO synthases. Cell Physiol Biochem 2013;31:355-365.
https://doi.org/10.1159/000343373

 

56 Chi RF, Wang JP, Wang K, Zhang XL, Zhang YA, Kang YM, Han XB, Li B, Qin FZ, Fan BA: Progressive Reduction in Myocyte Autophagy After Myocardial Infarction in Rabbits: Association with Oxidative Stress and Left Ventricular Remodeling. Cell Physiol Biochem 2017;44:2439-2454.
https://doi.org/10.1159/000486167

 

57 Jiang YQ, Chang Gl, Wang Y, Zhang DY, Cao L, Liu J: Geniposide prevents hypoxia/reoxygenation-induced apoptosis in H9c2 cells: improvement of mitochondrial dysfunction and activation of GLP-1R and the PI3K/AKT signaling pathway. Cell Physiol Biochem 2016;39:407-421.
https://doi.org/10.1159/000445634

 

58 Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X, Choi AM: Mechanisms of cell death in oxidative stress. Antioxid Redox Signal 2007;9:49-89.
https://doi.org/10.1089/ars.2007.9.49

 

59 Neri M, Fineschi V, Di Paolo M, Pomara C, Riezzo I, Turillazzi E, Cerretani D: Cardiac oxidative stress and inflammatory cytokines response after myocardial infarction. Curr Vasc Pharmacol 2015;13:26-36.
https://doi.org/10.2174/15701611113119990003

 

60 Chen Q, Kang J, Fu C: The independence of and associations among apoptosis, autophagy, and necrosis. Signal Transduct Target Ther 2018;3:18.
https://doi.org/10.1038/s41392-018-0018-5

 

61 Bujak M, Dobaczewski M, Gonzalez-Quesada C, Xia Y, Leucker T, Zymek P, Veeranna V, Tager AM, Luster AD, Frangogiannis NG: Induction of the CXC chemokine interferon-γ-inducible protein 10 regulates the reparative response following myocardial infarction. Circ Res 2009;105:973-983.
https://doi.org/10.1161/CIRCRESAHA.109.199471

 

62 Mirabelli-Badenier M, Braunersreuther V, Viviani GL, Dallegri F, Quercioli A, Veneselli E, Mach F, Montecucco F: CC and CXC chemokines are pivotal mediators of cerebral injury in ischaemic stroke. Thromb Haemost 2011;106:409-420.
https://doi.org/10.1160/TH10-10-0662

 

63 Palevski D, Levin-Kotler LP, Kain D, Naftali-Shani N, Landa N, Ben-Mordechai T, Konfino T, Holbova R, Molotski N, Rosin-Arbesfeld R: Loss of macrophage Wnt secretion improves remodeling and function after myocardial infarction in mice. J Am Heart Assoc 2017;6:e004387.
https://doi.org/10.1161/JAHA.116.004387

 

64 Lambert JM, Lopez EF, Lindsey ML: Macrophage roles following myocardial infarction. Int J Cardiol 2008;130:147-158.
https://doi.org/10.1016/j.ijcard.2008.04.059

 

65 Ben-Mordechai T, Palevski D, Glucksam-Galnoy Y, Elron-Gross I, Margalit R, Leor J: Targeting macrophage subsets for infarct repair. J Cardiovasc Pharmacol Ther 2015;20:36-51.
https://doi.org/10.1177/1074248414534916

 

66 Weinberger T, Schulz C: Myocardial infarction: a critical role of macrophages in cardiac remodeling. Front Physiol 2015;6:107.
https://doi.org/10.3389/fphys.2015.00107

 

67 Lin S-L, Li B, Rao S, Yeo E-J, Hudson TE, Nowlin BT, Pei H, Chen L, Zheng JJ, Carroll TJ: Macrophage Wnt7b is critical for kidney repair and regeneration. Proc Natl Acad Sci U S A 2010;107:4194-4199.
https://doi.org/10.1073/pnas.0912228107

 

68 Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo J-L, Libby P, Weissleder R, Pittet MJ: The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med 2007;204:3037-3047.
https://doi.org/10.1084/jem.20070885

 

69 Porcheray F, Viaud S, Rimaniol AC, Leone C, Samah B, Dereuddre-Bosquet N, Dormont D, Gras G: Macrophage activation switching: an asset for the resolution of inflammation. Clin Exp Immunol 2005;142:481-489.
https://doi.org/10.1111/j.1365-2249.2005.02934.x

 

70 Boufenzer A, Lemarié J, Simon T, Derive M, Bouazza Y, Tran N, Maskali F, Groubatch F, Bonnin P, Bastien C: TREM-1 mediates inflammatory injury and cardiac remodeling following myocardial infarction. Circ Res 2015;116:1772-1782.
https://doi.org/10.1161/CIRCRESAHA.116.305628

 

71 Jung M, Ma Y, Padmanabhan Iyer R, Yabluchiansky A, Garrett MR, Lindsey ML: IL-10 Regulates Inflammation to Improve LV Physiology After Myocardial Infarction by Stimulating M2 Macrophage Polarization and Fibroblast Activation. Circ Res 2017;121:A142.
https://doi.org/10.1007/s00395-017-0622-5

 

72 Shiri S, Alizadeh AM, Baradaran B, Farhanghi B, Shanehbandi D, Khodayari S, Khodayari H, Tavassoli A: Dendrosomal curcumin suppresses metastatic breast cancer in mice by changing m1/m2 macrophage balance in the tumor microenvironment. Asian Pac J Cancer Prev 2015;16:3917-3922.
https://doi.org/10.7314/APJCP.2015.16.9.3917

 

73 Gombozhapova A, Rogovskaya Y, Shurupov V, Rebenkova M, Kzhyshkowska J, Popov SV, Karpov RS, Ryabov V: Macrophage activation and polarization in post-infarction cardiac remodeling. Int J Biomed Sci 2017;24:13.
https://doi.org/10.1186/s12929-017-0322-3

 

74 Hermans KC, Daskalopoulos EP, Blankesteijn WM: Interventions in Wnt signaling as a novel therapeutic approach to improve myocardial infarct healing. Fibrogenesis Tissue Repair 2012;5:16.
https://doi.org/10.1186/1755-1536-5-16

 

75 Mizutani M, Wu JC, Nusse R: Fibrosis of the Neonatal Mouse Heart After Cryoinjury Is Accompanied by Wnt Signaling Activation and Epicardial-to-Mesenchymal Transition. J Am Heart Assoc 2016;5:e002457.
https://doi.org/10.1161/JAHA.115.002457

 

76 Bao MW, Cai Z, Zhang XJ, Li L, Liu X, Wan N, Hu G, Wan F, Zhang R, Zhu X, Xia H, Li H: Dickkopf-3 protects against cardiac dysfunction and ventricular remodelling following myocardial infarction. Basic Res Cardiol 2015;110:25.
https://doi.org/10.1007/s00395-015-0481-x

 

77 Yilmaz G, Arumugam TV, Stokes KY, Granger DN: Role of T lymphocytes and interferon-γ in ischemic stroke. Circulation 2006;113:2105-2112.
https://doi.org/10.1161/CIRCULATIONAHA.105.593046

 

78 Wan E, Yeap XY, Dehn S, Terry R, Novak M, Zhang S, Iwata S, Han X, Homma S, Drosatos K: Enhanced efferocytosis of apoptotic cardiomyocytes through myeloid-epithelial-reproductive tyrosine kinase links acute inflammation resolution to cardiac repair after infarction. Circ Res 2013;113:1004-1012.
https://doi.org/10.1161/CIRCRESAHA.113.301198

 

79 Liu G, Zhang H, Hao F, Hao J, Pan L, Zhao Q, Wo J: Clusterin Reduces Cold Ischemia-Reperfusion Injury in Heart Transplantation Through Regulation of NF-kB Signaling and Bax/Bcl-xL Expression. Cell Physiol Biochem 2018;45:1003-1012.
https://doi.org/10.1159/000487295

 

80 Frangogiannis NG: Interleukin-1 in cardiac injury, repair, and remodeling: pathophysiologic and translational concepts. Discoveries (Craiova) 2015;3:pii:e41.
https://doi.org/10.15190/d.2015.33

 

81 Laflamme MA, Murry CE: Regenerating the heart. Nat Biotechnol 2005;23:845.
https://doi.org/10.1038/nbt1117

 

82 Lee ST, White AJ, Matsushita S, Malliaras K, Steenbergen C, Zhang Y, Li TS, Terrovitis J, Yee K, Simsir S: Intramyocardial injection of autologous cardiospheres or cardiosphere-derived cells preserves function and minimizes adverse ventricular remodeling in pigs with heart failure post-myocardial infarction. J Am Coll Cardiol 2011;57:455-465.
https://doi.org/10.1016/j.jacc.2010.07.049

 

83 Amado LC, Saliaris AP, Schuleri KH, John MS, Xie JS, Cattaneo S, Durand DJ, Fitton T, Kuang JQ, Stewart G: Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A 2005;102:11474-11479.
https://doi.org/10.1073/pnas.0504388102

 

84 Dang S, Yu ZM, Zhang CY, Zheng J, Li KL, Wu Y, Qian LL, Yang ZY, Li XR, Zhang Y: Autophagy promotes apoptosis of mesenchymal stem cells under inflammatory microenvironment. Stem Cell Res Ther 2015;6:247.
https://doi.org/10.1186/s13287-015-0245-4

 

85 Huleihel M, Fadlon E, Abuelhija A, Haber EP, Lunenfeld E: Glial cell line-derived neurotrophic factor (GDNF) induced migration of spermatogonial cells in vitro via MEK and NF-kB pathways. Differentiation 2013;86:38-47.
https://doi.org/10.1016/j.diff.2013.06.005

 

86 Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP: Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003;361:47-49.
https://doi.org/10.1016/S0140-6736(03)12111-3

 

87 Xie CQ, Zhang J, Xiao Y, Zhang L, Mou Y, Liu X, Akinbami M, Cui T, Chen YE: Transplantation of human undifferentiated embryonic stem cells into a myocardial infarction rat model. Stem Cells Dev 2007;16:25-30.
https://doi.org/10.1089/scd.2006.110206

 

88 Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD: Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 2002;105:93-98.
https://doi.org/10.1161/hc0102.101442

 

89 Chiong M, Wang Z, Pedrozo Z, Cao D, Troncoso R, Ibacache M, Criollo A, Nemchenko A, Hill J, Lavandero S: Cardiomyocyte death: mechanisms and translational implications. Cell Death Dis 2011;2:e244.
https://doi.org/10.1038/cddis.2011.130

 

90 Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L: Mesenchymal stem cell-natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood 2006;107:1484-1490.
https://doi.org/10.1182/blood-2005-07-2775

 

91 Wei R, Yang J, Gao M, Wang H, Hou W, Mu Y, Chen G, Hong T: Infarcted cardiac microenvironment may hinder cardiac lineage differentiation of human embryonic stem cells. Cell Biol Int 2016;40:1235-1246.
https://doi.org/10.1002/cbin.10679

 

92 Hamid T, Xu Y, Ismahil MA, Li Q, Jones SP, Bhatnagar A, Bolli R, Prabhu SD: TNF receptor signaling inhibits cardiomyogenic differentiation of cardiac stem cells and promotes a neuroadrenergic-like fate. Am J Physiol Heart Circ Physiol 2016;311:H1189-H1201.
https://doi.org/10.1152/ajpheart.00904.2015

 

93 Lang CI, Wolfien M, Langenbach A, Müller P, Wolkenhauer O, Yavari A, Ince H, Steinhoff G, Krause BJ, David R: Cardiac cell therapies for the treatment of acute myocardial infarction: a meta-analysis from mouse studies. Cell Physiol Biochem 2017;42:254-268.
https://doi.org/10.1159/000477324

 

94 Bittle GJ, Morales D, Deatrick KB, Parchment N, Saha P, Mishra R, Sharma S, Pietris N, Vasilenko A, Bor C: Stem Cell Therapy for Hypoplastic Left Heart Syndrome: Mechanism, Clinical Application, and Future Directions. Circ Res 2018;123:288-300.
https://doi.org/10.1161/CIRCRESAHA.117.311206

 

95 Menasché P: Cell therapy trials for heart regeneration - lessons learned and future directions. Nat Rev Cardiol 2018:15:659-671.
https://doi.org/10.1038/s41569-018-0013-0

 

96 Mauritz C, Schwanke K, Reppel M, Neef S, Katsirntaki K, Maier LS, Nguemo F, Menke S, Haustein M, Hescheler J: Generation of functional murine cardiac myocytes from induced pluripotent stem cells. Circulation 2008;118:507-517.
https://doi.org/10.1161/CIRCULATIONAHA.108.778795

 

97 Wobus AM, Kaomei G, Shan J, Wellner MC, Rohwedel J, Guanju J, Fleischmann B, Katus HA, Hescheler J, Franz WM: Retinoic acid accelerates embryonic stem cell-derived cardiac differentiation and enhances development of ventricular cardiomyocytes. J Mol Cell Cardiol 1997;29:1525-1539.
https://doi.org/10.1006/jmcc.1997.0433

 

98 Mummery C, Ward-van Oostwaard D, Doevendans P, Spijker R, van den Brink S, Hassink R, van der Heyden M, Opthof T, Pera M, de la Riviere AB: Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation 2003;107:2733-2740.
https://doi.org/10.1161/01.CIR.0000068356.38592.68

 

99 Ivanyuk D, Budash G, Zheng Y, Gaspar JA, Chaudhari U, Fatima A, Bahmanpour S, Grin VK, Popandopulo AG, Sachinidis A: Ascorbic acid-induced cardiac differentiation of murine pluripotent stem cells: transcriptional profiling and effect of a small molecule synergist of Wnt/β-catenin signaling pathway. Cell Physiol Biochem 2015;36:810-830.
https://doi.org/10.1159/000430140

 

100 Sauer H, Bekhite MM, Hescheler J, Wartenberg M: Redox control of angiogenic factors and CD31-positive vessel-like structures in mouse embryonic stem cells after direct current electrical field stimulation. Exp Cell Res 2005;304:380-390.
https://doi.org/10.1016/j.yexcr.2004.11.026

 

101 Sauer H, Günther J, Hescheler J, Wartenberg M: Thalidomide inhibits angiogenesis in embryoid bodies by the generation of hydroxyl radicals. Am J Pathol 2000;156:151-158.
https://doi.org/10.1016/S0002-9440(10)64714-1

 

102 Halbach M, Peinkofer G, Baumgartner S, Maass M, Wiedey M, Neef K, Krausgrill B, Ladage D, Fatima A, Saric T: Electrophysiological integration and action potential properties of transplanted cardiomyocytes derived from induced pluripotent stem cells. Cardiovasc Res 2013;100:432-440.
https://doi.org/10.1093/cvr/cvt213

 

103 Xi J, Khalil M, Shishechian N, Hannes T, Pfannkuche K, Liang H, Fatima A, Haustein M, Suhr F, Bloch W: Comparison of contractile behavior of native murine ventricular tissue and cardiomyocytes derived from embryonic or induced pluripotent stem cells. FASEB J 2010;24:2739-2751.
https://doi.org/10.1096/fj.09-145177

 

104 Shinde V, Srinivasan SP, Henry M, Rotshteyn T, Hescheler J, Rahnenführer J, Grinberg M, Meisig J, Blüthgen N, Waldmann T: Comparison of a teratogenic transcriptome-based predictive test based on human embryonic versus inducible pluripotent stem cells. Stem Cell Res Ther 2016;7:190.
https://doi.org/10.1186/s13287-016-0449-2

 

105 Postovit LM, Margaryan NV, Seftor EA, Kirschmann DA, Lipavsky A, Wheaton WW, Abbott DE, Seftor RE, Hendrix MJ: Human embryonic stem cell microenvironment suppresses the tumorigenic phenotype of aggressive cancer cells. Proc Natl Acad Sci U S A 2008;105:4329-4334.
https://doi.org/10.1073/pnas.0800467105

 

106 Tan Y, Ooi S, Wang L: Immunogenicity and tumorigenicity of pluripotent stem cells and their derivatives: genetic and epigenetic perspectives. Curr Stem Cell Res Ther 2014;9:63-72.
https://doi.org/10.2174/1574888X113086660068

 

107 Nussbaum J, Minami E, Laflamme MA, Virag JA, Ware CB, Masino A, Muskheli V, Pabon L, Reinecke H, Murry CE: Transplantation of undifferentiated murine embryonic stem cells in the heart: teratoma formation and immune response. FASEB J 2007;21:1345-1357.
https://doi.org/10.1096/fj.06-6769com

 

108 Zhang Y, Wang D, Chen M, Yang B, Zhang F, Cao K: Intramyocardial transplantation of undifferentiated rat induced pluripotent stem cells causes tumorigenesis in the heart. PloS One 2011;6:e19012.
https://doi.org/10.1371/journal.pone.0019012

 

109 Ikeda Y, Terzic A, Nelson TJ, Mael AA, Martinez Fernandez AJ, Yamada S: Method of treating heart tissue using induced pluripotent stem cells. US Patent, Patent No. 10,047,346.

 

110 Rikhtegar R, Pezeshkian M, Dolati S, Safaie N, Afrasiabi Rad A, Mahdipour M, Nouri M, Jodati AR, Yousefi M: Stem cells as therapy for heart disease: iPSCs, ESCs, CSCs, and skeletal myoblasts. Biomed Pharmacother 2019;109:304-313.
https://doi.org/10.1016/j.biopha.2018.10.065

 

111 Yoshida Y, Yamanaka S: Induced Pluripotent Stem Cells 10 Years Later: For Cardiac Applications. Circ Res 2017;120:1958-1968.
https://doi.org/10.1161/CIRCRESAHA.117.311080

 

112 Liang G, Zhang Y: Genetic and epigenetic variations in iPSCs: potential causes and implications for application. Cell Stem Cell 2013;13:149-159.
https://doi.org/10.1016/j.stem.2013.07.001

 

113 Lin Y, Liu H, Klein M, Ostrominski J, Hong SG, Yada RC, Chen G, Navarengom K, Schwartzbeck R, San H, Yu ZX, Liu C, Linask K, Beers J, Qiu L, Dunbar CE, Boehm M, Zou J: Efficient differentiation of cardiomyocytes and generation of calcium-sensor reporter lines from nonhuman primate iPSCs. Sci Rep 2018;8:5907.
https://doi.org/10.1038/s41598-018-24074-y

 

114 Abou-Saleh H, Zouein FA, El-Yazbi A, Sanoudou D, Raynaud C, Rao C, Pintus G, Dehaini H, Eid AH: The march of pluripotent stem cells in cardiovascular regenerative medicine. Stem Cell Res Ther 2018;9:201.
https://doi.org/10.1186/s13287-018-0947-5

 

115 Carvalho E, Verma P, Hourigan K, Banerjee R: Myocardial infarction: stem cell transplantation for cardiac regeneration. Regen Med 2015;10:1025-1043.
https://doi.org/10.2217/rme.15.63

 

116 Templin C, Zweigerdt R, Schwanke K, Olmer R, Ghadri JR, Emmert MY, Müller E, Küest SM, Cohrs S, Schibli R: Transplantation and tracking of human induced pluripotent stem cells in a pig model of myocardial infarction: assessment of cell survival, engraftment and distribution by hybrid SPECT-CT imaging of sodium iodide symporter trangene expression. Circulation 2012;126:430-439.
https://doi.org/10.1161/CIRCULATIONAHA.111.087684

 

117 Xiong Q, Ye L, Zhang P, Lepley M, Tian J, Li J, Zhang L, Swingen C, Vaughan JT, Kaufman DS: Functional consequences of human induced pluripotent stem cells therapy: myocardial ATP turnover rate in the in vivo swine hearts with post-infarction remodeling. Circulation 2013;127:997-1008.
https://doi.org/10.1161/CIRCULATIONAHA.112.000641

 

118 Nelson TJ, Martinez-Fernandez A, Yamada S, Perez-Terzic C, Ikeda Y, Terzic A: Repair of acute myocardial infarction by human stemness factors induced pluripotent stem cells. Circulation 2009;120:408-416.
https://doi.org/10.1161/CIRCULATIONAHA.109.865154

 

119 Lepperhof V, Polchynski O, Kruttwig K, Bruggemann C, Neef K, Drey F, Zheng Y, Ackermann JP, Choi YH, Wunderlich TF, Hoehn M, Hescheler J, Saric T: Bioluminescent imaging of genetically selected induced pluripotent stem cell-derived cardiomyocytes after transplantation into infarcted heart of syngeneic recipients. PloS One 2014;9:e107363.
https://doi.org/10.1371/journal.pone.0107363

 

120 Sahito R, Heras-Bautista C, Krausgrill B, Maass M, Baumgartner S: Gelatine Microspheres Support Direct Intramyocardial Delivery of Induced Pluripotent Stem Cell-Derived Cardiomyocytes. J Stem Cell Res Ther 2015;5:2.

 

121 Breitbach M, Bostani T, Roell W, Xia Y, Dewald O, Nygren JM, Fries JW, Tiemann K, Bohlen H, Hescheler J: Potential risks of bone marrow cell transplantation into infarcted hearts. Blood 2007;110:1362-1369.
https://doi.org/10.1182/blood-2006-12-063412

 

122 Johnston PV, Sasano T, Mills K, Evers R, Lee ST, Smith RR, Lardo AC, Lai S, Steenbergen C, Gerstenblith G: Engraftment, differentiation, and functional benefits of autologous cardiosphere-derived cells in porcine ischemic cardiomyopathy. Circulation 2009;120:1075-1083.
https://doi.org/10.1161/CIRCULATIONAHA.108.816058

 

123 Hatzistergos KE, Quevedo H, Oskouei BN, Hu Q, Feigenbaum GS, Margitich IS, Mazhari R, Boyle AJ, Zambrano JP, Rodriguez JE: Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circ Res 2010;107:913-922.
https://doi.org/10.1161/CIRCRESAHA.110.222703

 

124 Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ, Zhang JJ, Chunhua RZ, Liao LM, Lin S: Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. Am J Cardiol 2004;94:92-95.
https://doi.org/10.1016/j.amjcard.2004.03.034

 

125 Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS: A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol 2009;54:2277-2286.
https://doi.org/10.1016/j.jacc.2009.06.055

 

126 Wojakowski W, Tendera M, Michałowska A, Majka M, Kucia M, Maslankiewicz K, Wyderka R, Ochała A, Ratajczak MZ: Mobilization of CD34/CXCR4+, CD34/CD117+, c-met+ stem cells, and mononuclear cells expressing early cardiac, muscle, and endothelial markers into peripheral blood in patients with acute myocardial infarction. Circulation 2004;110:3213-3220.
https://doi.org/10.1161/01.CIR.0000147609.39780.02

 

127 Kang HJ, Kim HS, Zhang SY, Park KW, Cho HJ, Koo BK, Kim YJ, Lee DS, Sohn DW, Han KS: Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomised clinical trial. Lancet 2004;363:751-756.

 

128 Bartunek J, Vanderheyden M, Vandekerckhove B, Mansour S, De Bruyne B, De Bondt P, Van Haute I, Lootens N, Heyndrickx G, Wijns W: Intracoronary injection of CD133-positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction: feasibility and safety. Circulation 2005;112:I178-I183.

 

129 Kamihata H, Matsubara H, Nishiue T, Fujiyama S, Tsutsumi Y, Ozono R, Masaki H, Mori Y, Iba O, Tateishi E: Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation 2001;104:1046-1052.
https://doi.org/10.1161/hc3501.093817

 

130 Lee SH, Hong JH, Cho KH, Noh JW, Cho HJ: Discrepancy between short-term and long-term effects of bone marrow-derived cell therapy in acute myocardial infarction: a systematic review and meta-analysis. Stem Cell Res Ther 2016;7:153.
https://doi.org/10.1186/s13287-016-0415-z

 

131 Sanz-Ruiz R, Casado Plasencia A, Borlado LR, Fernández-Santos ME, Al-Daccak R, Claus P, Palacios I, Sádaba R, Charron D, Bogaert J: Rationale and design of a clinical trial to evaluate the safety and efficacy of intracoronary infusion of allogeneic human cardiac stem cells in patients with acute myocardial infarction and left ventricular dysfunction: the randomized multicenter double-blind controlled CAREMI trial (cardiac stem cells in patients with acute myocardial infarction). Circ Res 2017;121:71-80.
https://doi.org/10.1161/CIRCRESAHA.117.310651

 

132 Penicka M, Horak J, Kobylka P, Pytlik R, Kozak T, Belohlavek O, Lang O, Skalicka H, Simek S, Palecek T: Intracoronary injection of autologous bone marrow-derived mononuclear cells in patients with large anterior acute myocardial infarction: a prematurely terminated randomized study. J Am Coll Cardiol 2007;49:2373-2374.
https://doi.org/10.1016/j.jacc.2007.04.009

 

133 Malliaras K, Makkar RR, Smith RR, Cheng K, Wu E, Bonow RO, Marbán L, Mendizabal A, Cingolani E, Johnston PV: Intracoronary cardiosphere-derived cells after myocardial infarction: evidence of therapeutic regeneration in the final 1-year results of the CADUCEUS trial (CArdiosphere-Derived aUtologous stem CElls to reverse ventricUlar dySfunction). J Am Coll Cardiol 2014;63:110-122.
https://doi.org/10.1016/j.jacc.2013.08.724

 

134 Burchfield JS, Dimmeler S: Role of paracrine factors in stem and progenitor cell mediated cardiac repair and tissue fibrosis. Fibrogenesis Tissue Repair 2008;1:4.
https://doi.org/10.1186/1755-1536-1-4

 

135 Segers VF, Tokunou T, Higgins LJ, MacGillivray C, Gannon J, Lee RT: Local delivery of protease-resistant stromal cell derived factor-1 for stem cell recruitment after myocardial infarction. Circulation 2007;116:1683-1692.
https://doi.org/10.1161/CIRCULATIONAHA.107.718718

 

136 Buikema JW, Zwetsloot PPM, Doevendans PA, Domian IJ, Sluijter JP: Wnt/β-catenin signaling during cardiac development and repair. J Cardiovasc Dev Dis 2014;1:98-110.
https://doi.org/10.3390/jcdd1010098

 

137 Tang J, Wang J, Kong X, Yang J, Guo L, Zheng F, Zhang L, Huang Y, Wan Y: Vascular endothelial growth factor promotes cardiac stem cell migration via the PI3K/Akt pathway. Exp Cell Res 2009;315:3521-3531.
https://doi.org/10.1016/j.yexcr.2009.09.026

 

138 Xiao N, Qi XY, Tang LN, Tan LL, Chen YQ, Zhao HM: VEGF promotes cardiac stem cells differentiation into vascular endothelial cells via the PI3K/Akt signaling pathway. Artif Cells Nanomed Biotechnol 2014;42:400-405.
https://doi.org/10.3109/21691401.2013.837473

 

139 Boni A, Urbanek K, Nascimbene A, Hosoda T, Zheng H, Delucchi F, Amano K, Gonzalez A, Vitale S, Ojaimi C: Notch1 regulates the fate of cardiac progenitor cells. Proc Natl Acad Sci U S A 2008;105:15529-15534.
https://doi.org/10.1073/pnas.0808357105

 

140 Limana F, Germani A, Zacheo A, Kajstura J, Di Carlo A, Borsellino G, Leoni O, Palumbo R, Battistini L, Rastaldo R: Exogenous high-mobility group box 1 protein induces myocardial regeneration after infarction via enhanced cardiac C-kit+ cell proliferation and differentiation. Circ Res 2005;97:e73-e83.
https://doi.org/10.1161/01.RES.0000186276.06104.04

 

141 Kattman SJ, Huber TL, Keller GM: Multipotent flk-1+ cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages. Dev Cell 2006;11:723-732.
https://doi.org/10.1016/j.devcel.2006.10.002

 

142 Meistrich ML, Hunter NR, Suzuki N, Trostle PK, Withers HR: Gradual regeneration of mouse testicular stem cells after exposure to ionizing radiation. Radiat Res 1978;74:349-362.
https://doi.org/10.2307/3574894

 

143 Morimoto H, Iwata K, Ogonuki N, Inoue K, Atsuo O, Kanatsu-Shinohara M, Morimoto T, Yabe-Nishimura C, Shinohara T: ROS are required for mouse spermatogonial stem cell self-renewal. Cell Stem cell 2013;12:774-786.
https://doi.org/10.1016/j.stem.2013.04.001

 

144 Withers H, Hunter N, Barkley Jr H, Reid B: Radiation survival and regeneration characteristics of spermatogenic stem cells of mouse testis. Radiat Res 1974;57:88-103.
https://doi.org/10.2307/3573759

 

145 Wu SM, Fujiwara Y, Cibulsky SM, Clapham DE, Lien CL, Schultheiss TM, Orkin SH: Developmental origin of a bipotential myocardial and smooth muscle cell precursor in the mammalian heart. Cell 2006;127:1137-1150.
https://doi.org/10.1016/j.cell.2006.10.028