2024 Stem Cell Therapy For Cardiac Regeneration Past Present and Future

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

OPEN ACCESS | Review

Stem cell therapy for cardiac regeneration: past, present,


and future
Jaideep Kaur Gill, Sargun Kaur Rehsia, Elika Verma , Niketa Sareen, and Sanjiv Dhingra
Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre Regenerative Medicine Program,
Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Biomedical Engineering Program, University of
Manitoba, Winnipeg MB, R2H2A6, Canada
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

Corresponding authors: Niketa Sareen (email: nsareen@sbrc.ca); Sanjiv Dhingra (email: sdhingra@sbrc.ca)

Abstract
Cardiac disorders remain the leading cause of mortality worldwide. Current clinical strategies, including drug therapy, surgi-
cal interventions, and organ transplantation offer limited benefits to patients without regenerating the damaged myocardium.
Over the past decade, stem cell therapy has generated a keen interest owing to its unique self-renewal and immune privileged
characteristics. Furthermore, the ability of stem cells to differentiate into specialized cell types, has made them a popular
therapeutic tool against various diseases. This comprehensive review provides an overview of therapeutic potential of differ-
ent types of stem cells in reference to cardiovascular diseases. Furthermore, it sheds light on the advantages and limitations
associated with each cell type. An in-depth analysis of the challenges associated with stem cell research and the hurdles for
its clinical translation and their possible solutions have also been elaborated upon. It examines the controversies surround-
ing embryonic stem cells and the emergence of alternative approaches, such as the use of induced pluripotent stem cells for
cardiac therapeutic applications. Overall, this review serves as a valuable resource for researchers, clinicians, and policymak-
ers involved in the field of regenerative medicine, guiding the development of safe and effective stem cell-based therapies to
revolutionize patient care.
Key words: induced pluripotent stem cells, mesenchymal stem cells, cardiac regeneration, myocardial ischemia

Introduction
tients with coronary heart disease and help patients pre-
Heart failure continues to be a leading cause of mor- vent any severe cardiac effects after an initial episode of
tality around the world. In 2019 alone, around 17.9 mil- cardiac arrest (Rossignol et al. 2019). However, these ther-
lion people died of cardiovascular diseases, globally (CVDs apies provide only marginal benefits in terms of long-term
2021). It is anticipated that the number of patients suf- recovery from disease and their continuous use might be
fering from the disease will soar as the elderly popula- associated with severe side-effects masking the positive im-
tion is growing (Groenewegen et al. 2020). Besides the el- pact they have on the patients. Nevertheless, these thera-
derly, younger adults too are predisposed to cardiac disor- pies are insufficient to regenerate or repair the cardiac mi-
ders due to risk factors such as obesity, high cholesterol, lieu, so their role in cardiac remodeling is limited (Tamargo
high blood pressure, and sedentary lifestyle (Kelishadi and and López-Sendón 2011). In addition to this, for patients
Poursafa 2014). It is evident that the primary etiology of with organ transplantation as the only option, surgeries are
heart failure is coronary artery disease and myocardial is- limited by the number of organs available for transplanta-
chemia. These aetiologies commonly exist together and aug- tion. Therefore, new therapeutic options that outperform
ment the risk of developing a heart failure (Fox et al. 2001). the conventional approaches for treatment of cardiac disor-
Conventional therapies to treat cardiovascular diseases in- ders are being investigated. Tissue regeneration using cell
volve catheter-based interventions, including angioplasty for therapy is an active area of research due to limited regen-
unclogging the arteries, and surgical interventions, includ- erative capacity of the heart following an ischemic injury.
ing coronary artery bypass surgery or organ transplant in Cell therapy provides a reliable solution for replacing the
case of end-stage heart failure (Freystaetter and Akowuah terminally differentiated cardiomyocytes (CMs) lost during
2021). These treatments might also include administration myocardial ischemia. Stem cell therapy is a novel treatment
of cardioprotective drugs like beta blockers, calcium chan- approach involving the use of undifferentiated stem cells to
nel blockers, or oral diuretics. These cardioprotective treat- improve a given cardiac condition (Wei et al. 2013; Padda et
ments have been proven to enhance cardiac function in pa- al. 2015).

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 161


Canadian Science Publishing

Fig. 1. Types of stem cells for cardiac regeneration and source of stem cells. Different types of stem cells are employed for
cardiac therapy, including mesenchymal stem cells (MSCs) and pluripotent stem cells. MSCs can be derived from marrows of
iliac crest and femur (BM-MSCs); umbilical cord (UC-MSCs); adipose tissue (AD-MSCs); or from amniotic fluid. Pluripotent stem
cells can be isolated from blastocyst stage embryo (ESCs) or by de-differentiation of somatic cells from skin or blood, in the
presence of growth factors (iPSCs). Created using BioRender (BioRender.com).

Bone marrow Umbilical cord


(for BM- (for UC-SCs)
MSCs)
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

Adipocytes
Blastocyst
Stem cell (for AD-MSCs)
(for ESCs) culture Heart

Somatic cells Amniotic fluid


+
Reprogramming factors (for MSCs)
SKIN BLOOD
(Oct3/4, Sox2, Klf4, c-Myc)
(for iPSCs)

Stem cells are the undifferentiated cells in our body that Different types of stem cells for heart
possess the ability of self-renewal and differentiation into
multiple cell types of our body (Zakrzewski et al. 2019).
repair
Based on their differentiation abilities, stem cells can be
Pluripotent stem cells
either totipotent, pluripotent, or multipotent in nature.
Totipotent cells possess the ability to generate all cell types Embryonic stem cells
in the human body, including the embryonic and extra
embryonic tissues. Pluripotent cells are generally derived ESCs are pluripotent stem cells obtained from the inner
from inner cell mass of the embryo. They have the capac- cell mass of day 4 to 7 blastocyst stage embryo (Zakrzewski
ity to divide into cell types from all three lineages (endo- et al. 2019). ESCs are promising candidates for the treat-
dermal, mesodermal, or ectodermal), except the extraem- ment of cardiac diseases due to their ability to proliferate
bryonic tissues. These cells are referred to as embryonic indefinitely and differentiate into cells of all three germ
stem cells (ESCs). Additionally, pluripotent stem cells can layers (Menasché 2020; Zhu et al. 2009). Following their first
also be induced from adult somatic cells like skin tissue or isolation in 1994, human ESCs (hESCs) have been widely
blood cells by de-differentiating them in the presence of re- used for the generation of different cell types for multiple
quired growth factors known as Yamanaka factors (Rikhtegar therapeutic applications (Hentze et al. 2009; Pré et al. 2013).
et al. 2019). These induced pluripotent stem cells (iPSCs) Directed differentiation of hESCs into CMs can be achieved
have gained a lot of attention lately, for their flexibility under appropriate culture conditions and CM-specific factors
in being re-programmed from any somatic cell and dif- (Dixon et al. 2011). However, a small percentage of hESCs
ferentiating them into cells of interest (Yamanaka 2020). (5%–15%) spontaneously differentiate into CMs under the
Multipotent mesenchymal stem cells (MSCs) have a lim- required conditions (Wong and Bernstein 2010). Current re-
ited differentiation potential and can generate only cer- search is focused on controlling the differentiation of hESCs
tain cell types. These stem cells can be isolated from into the cardiac lineage to increase the yield of hESC-derived
adult tissues, including bone marrow (BM), blood, adi- CMs for cell therapy. The techniques for enrichment, purifi-
pose tissue (AD), or neonatal sources like umbilical cord cation, and selection have been developed to direct cardiac
(UC), placenta, or amniotic fluid (Guo et al. 2020) (Fig. differentiation toward comparatively pure homogeneity.
1). Based on the donor, the transplantation of stem cells The strategies such as long-term culture, three-dimensional
can be either autologous (cells from the patient are in- (3D) tissue engineering, mechanical loading, electrical stim-
jected back in the same person for treatment) or allo- ulation, modification of substrate stiffness, and therapy
geneic (where the cells from a healthy donor are in- with neurohormonal substances have been tested in hESC-
jected into the diseased, non-self host) (Zakrzewski et al. derived CMs to improve their differentiation potential and
2019). maturation status (Yang et al. 2014).

162 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

ESCs have shown the potential to promote cardiac re- Yamanaka factors (Oct3/4, Sox2, Klf4, and c-Myc) (Rikhtegar
modeling in pre-clinical trials by differentiating into car- et al. 2019). Exhibiting unique characteristics of pluripotency
diac progenitors, which engraft into the host myocardium and self renewal, iPSCs are becoming an ideal choice for the
and reduce scar formation. A study reported that extracel- field of regenerative medicine. Additionally, there are sev-
lular vesicles (EVs) secreted by human pluripotent stem cells eral modifications in cell culture methods and protocols that
(hPSCs)-derived cardiovascular progenitor cells (hPSCs–CPCs) have been optimized to promote the chamber-specific differ-
exhibited cardioprotective effects under hypoxic conditions, entiation of iPSCs to generate ventricular, nodal, and atrial
which in turn increased the expression levels of metasta- CMs, smooth muscle cells, and cardiac fibroblasts (Batalov
sis associated lung adenocarcinoma transcript 1 (MALAT-1). and Feinberg 2015; Batalov et al. 2021; Wang et al. 2021).
The MALAT-1 was found to promote tube formation in hu- Unlike their embryonic counterparts, iPSCs are being highly
man umbilical vein endothelial cells by targeting miR-497 considered for disease modeling and drug screening owing
(Wu et al. 2020). Further, Chong et al. (2014) demonstrated to their accessibility and ethical acceptability (Cong et al.
the ability of hESC-derived CMs to remuscularize infarcted 2019; Rikhtegar et al. 2019). A study demonstrated that iPSC-
monkey hearts through intramyocardial (IM) administration. derived CM transplantation improved functional recovery in
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

They found that the grafted cells formed electromechanical macaque monkey hearts via reduction in the scar size. Fur-
connections with host myocytes, indicating significant poten- thermore, after 1 month of transplantation, the CM-treated
tial for regeneration of infarcted heart muscle in humans. hearts exhibited significant recovery in global LVEF of around
Although sustained engraftment of ESCs is rare, ESC-derived 10.6% ± 0.9% (Liu et al. 2018). In a study by Ye et al., the
progenitors have been observed to positively influence the re- efficacy of iPSCs in acute MI was investigated in a porcine
modeling of infarcted heart tissue through paracrine mecha- model. Three types of cells derived from iPSCs were intramy-
nisms (Chong et al. 2014). In the first clinical case report on ocardially administered, namely CMs, endothelial cells, and
a 68-year old patient suffering from severe HF, hPSCs–CPCs smooth muscle cells. Additionally, a 3D fibrin patch was ap-
displayed promising results with no complications pertain- plied, which released insulin-like growth factor-1 (IFG-1) to
ing to arrhythmias, tumor formation, or immunosuppres- increase survival. Significant improvements were observed in
sion along with an increased left ventricular ejection frac- many parameters of cardiac function, such as LV function and
tion (LVEF) (36%) following transplant (Menasché et al. 2015). arteriole density. Additionally, myocardial metabolism and
Menasche et al. evaluated the safety of hESCs in patients with ATP turnover rate were significantly improved, while apop-
severe ischemic LV failure and explored the feasibility of pro- tosis and infarct size were considerably reduced without any
ducing clinical-grade CPCs. In this ESCORT trial, around 8.2 ventricular arrhythmias (Ye et al. 2014). Another approach
million hESC-derived cardiovascular progenitors embedded to using iPSCs in heart disease is through iPSC-derived EVs
in a fibrin patch were delivered epicardially during a coro- (iPSC-EVs). These vesicles contain molecular cargo such as
nary artery bypass procedure to six patients with a median iPSC-released miRNAs and additional factors that aid in cel-
LVEF of 26%. The patient safety was evaluated after a year of lular differentiation and angiogenesis (Rikhtegar et al. 2019).
treatment based on the imaging for cardiac or off-target tu- Adamiak et al. compared the efficacy of iPSCs and iPSC-EVs
mors and serial interrogations of cardioverter–defibrillators in a mice model of acute MI. The mice were divided into
placed in every patient to screen for arrhythmias. The study three groups, namely IM injection of vehicle, iPSCs, or iPSC-
also checked for the presence of donor-specific antibodies in EVs obtained from murine iPSC supernatants. While both iP-
recipients to assess alloimmune response. It was observed SCs and iPSC-EVs showed similar improvements in functional
that few patients acquired clinically silent alloimmunization. myocardium, iPSC-EVs showed a more significant decrease in
Additionally, most of the patients were symptomatically re- apoptosis. Additionally, teratoma formation was observed in
lieved, with cell-treated segments displaying an increased sys- the iPSC group, but not in the iPSC-EV group (Adamiak et al.
tolic motion. This trial helped to establish a protocol to ob- 2018). One of the most fruitful applications of iPSCs that is be-
tain highly purified (97.55%) cardiovascular progenitor popu- ing widely exploited is the development of disease models for
lation from hESCs (Menasché et al. 2018). Although there are different pathological conditions. Several researchers have
well-established in vitro cell differentiation techniques and reported isolation and application of patient-specific iPSCs as
encouraging results from in vivo preclinical research, several disease models, since these cells offer patient-specific geno-
obstacles must be overcome before wide acceptance of ESCs type that can be studied for its genetic abnormalities without
as clinical candidates for patients with myocardial infarction epigenetic effects on cells (Musunuru et al. 2018). These mod-
(MI). For instance, due to their embryonic origin, isolation els have been reported for valvular defects, rhythm disorders,
of hESCs is associated with ethical issues in many countries metabolic disorders, and patient-specific iPSCs for dilated car-
(Volarevic et al. 2018). Furthermore, some studies also express diomyopathy (Lin et al. 2015; Blinova et al. 2019; Sequiera et
the concern of teratoma formation following ESCs injection al. 2022).
(Hentze et al. 2009; Yamanaka 2020). Several clinical trials are currently investigating the ap-
plication of ESCs and iPSCs for cardiac therapy (Okano et
al. 2013; Deinsberger et al. 2020). An ongoing phase I trial
Induced pluripotent stem cells (NCT03763136) is evaluating the safety and efficacy of al-
Since 2006, iPSCs have emerged as a revolutionary devel- logeneic iPSC-derived CM transplantation in patients with
opment in the field of regenerative medicine. These cells are severe heart failure. With around 20 patients, this study
generated by reprogramming adult somatic cells using four is currently recruiting and is estimated to complete this

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 163


Canadian Science Publishing

year (Help Therapeutics 2022). Another phase I clinical trial of pelvis or from marrow in the femur (Deans and Moseley
(NCT04696328) is investigating the safety and efficacy of al- 2000). These cells are easy to isolate and offer easy expan-
logeneic iPSC-derived CM sheets for treating ischemic car- sion in vitro without loss of function. In addition to this, they
diomyopathy. This trial will focus on improvement of LVEF can be obtained from young healthy donors and transplanted
alongside other safety-related evaluations (Toda 2021). into allogeneic, diseased, and aged hosts (Da Silva and Hare
These clinical trials are evaluating the safety and efficacy 2013). Numerous experimental studies have reported sup-
of stem cell therapy for heart disease, and their results may porting evidence for the use of BM-MSCs in improving the
provide important insights into the potential of these ther- function of damaged myocardium by secretion of soluble
apies for clinical use in cardiac patients (Shiba et al. 2016). factors and promoting angiogenesis, reducing immune re-
However, to ensure a successful clinical translation, it is im- sponse, and preventing fibrosis (Ammar et al. 2015; Stephen
portant to address the challenges associated with the use of et al. 2016; Shafei et al. 2017). Boomsma et al. demonstrated
iPSCs, such as the risk of teratoma formation and the high that after injection of murine BM-MSCs into the infarcted
cost of production (Ye et al. 2014). Recently, in a study, in- myocardium, systolic function was preserved post injection.
ducible caspase-9 (iC9) was introduced into iPSCs, and the Also, the MSCs were able to migrate to the site of damage
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

exposure of these iPSCs to chemical inducer of dimerization and restore contractility of the heart by secreting paracrine
(CID) lead to initiation of a caspase cascade, which helped in factors (Boomsma et al. 2007). In a rabbit model of dilated
elimination of tumor-forming cells, thereby creating an effi- ischemic cardiomyopathy, Mu et al. demonstrated that trans-
cient iC9/CID safeguard system (Ando et al. 2015). The trans- plantation of autologous BM-MSCs ameliorated cardiac func-
plantation of iPSCs in the heart is also reported to cause tion through paracrine interaction with endogenous CMs.
arrhythmogenic response; in large animal models, there is The BM-MSCs also resulted in upregulation of VEGF recep-
a greater risk of developing temporary ventricular arrhyth- tors, thus suggesting their pro-angiogenic potential (Mu et
mia compared to small animal models (Kadota et al. 2020). al. 2011). In swine model of MI-induced injury, IM injection
Okawa et al. compared the transcriptomic profile of fetal of BM-MSCs improved cardiac function and ventricular wall
heart and found that iPSC-derived CMs (iPSC-CMs) had similar thickness while reducing the scar size (Schuleri et al. 2009).
immature phenotype and electrophysiology, which may con- Cai et al. reported similar findings in acute MI models where
tribute to arrhythmia (van den Berg et al. 2015). To overcome injection of BM-MSCs in swine hearts resulted in improved
this challenge and achieve a mature phenotype, Parikh et al. cardiac function by upregulation of mTOR and improved glu-
(2017) used a combination of triiodo-l-thyronine and dexam- cose metabolism in the heart by increasing the expression
ethasone to increase the formation of T-tubules in iPSC-CMs, of GLUT1 and 4 transporters (Cai et al. 2016). In CHART-1
which resulted in subsequent calcium release and excitation– study, the efficacy of autologous cardiopoietic MSCs was as-
contraction coupling that were similar to adult myocardium. sessed in 351 participants, the MSC-treated groups showed
progressive reduction in LV end-diastolic and end-systolic vol-
MSCs for cardiac therapy umes within 1 year of stem cell injection (Teerlink et al. 2017).
MSCs are multipotent cells with the ability to differenti- Despite the promising results obtained in several studies,
ate into cells of specific lineages, including osteogenic, chon- some of the reports have demonstrated increased expression
drogenic, and adipogenic (Poomani et al. 2022). MSCs can of several apoptotic and senescence genes with age, which
be isolated from several adult or neonatal tissues, including might affect the functions and proliferation of autologous
BM, AD, endometrium, dental pulp, and UC along with sev- stem cells isolated from older patients, limiting the use of
eral other sources (Guo et al. 2020). Their ease of isolation autologous MSCs in clinical trials (Choudhery et al. 2012).
and maintenance, as well as least teratoma formation ten- However, aging itself does not affect the positive outcomes
dency makes MSCs favorable candidates for clinical studies. of MSCs therapy, which was shown in a study by Golpanian
Currently, MSCs from BM and UC are most widely used in et al. (2015) where the patients receiving MSCs were segre-
clinics for cardiac therapy, followed by AD derivatives (Kabat gated into different age groups and the results of improved
et al. 2020). MSCs mediate their action by secreting various cardiac function and scar size did not vary based on the re-
paracrine factors involved in tissue regeneration pathways, cipient’s age. In addition to improving cardiac function in
including vascular endothelial growth factor (VEGF), hepa- vivo, BM-MSCs have also been reported to stimulate the dif-
tocyte growth factor (HGF), and transforming growth factor ferentiation as well as proliferation of ckit+ cells in the my-
(TGF)-beta, which aid in angiogenesis and wound repair. They ocardium. In pig MI models, treatment with BM-MSCs re-
also release immunosuppressive molecules such as IL-10, sulted in increase in ckit+ and GATA4+ cardiac stem cells
prostaglandin E2 (PGE2), macrophage colony-stimulating fac- compared to control animals. In this study, MSCs stimulated
tor, and tumor necrosis factor (TNF), which assist in prevent- generation of Nkx and troponin positive adult cardioblasts
ing adverse immune response in the ischemic myocardium, in vitro (Hatzistergos et al. 2010). Furthermore, Huang et al.
thus repairing the tissue (Dhingra et al. 2013; Yan et al. 2019; (2013) showed that transplantation of sheet fragments of au-
Jiang and Xu 2020; Sareen et al. 2020). tologous BM-MSCs post MI into the infarcted myocardium
of a porcine model significantly improved ventricular dila-
tion and maintained cardiac function after surgically induced
Bone marrow-derived MSCs
MI. Hare et al. (2009) tested the efficacy of allogeneic MSCs
Bone marrow-derived MSCs (BM-MSCs) are commonly iso- in acute MI patients in Prochymal trial and found improved
lated from blood obtained from the marrow in iliac crest LVEF and reverse remodeling in BM-MSC-treated patients

164 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

compared to untreated controls. To address the effect of MSC trial, assessed the efficacy and safety of UC-MSCs in patients
dose in modulating cardiac function, Florea et al. conducted with chronic HF. The patients who received UC-MSCs showed
a TRIDENT trial where 30 ischemia patients received differ- significant improvement in LVEF with no adverse events
ent doses (20 million or 100 million) of MSCs. Patients receiv- till 12 months follow-up period. Additionally, there were no
ing higher dose of MSCs showed improved EF compared to alloantibodies generated against the transplanted cells till
the patients treated with lower numbers of MSCs; however, 90 days of injection (Bartolucci et al. 2017). Currently, a ran-
both groups showed reduction in the scar size (Florea et al. domized clinical trial is underway, involving 20 MI patients
2017). undergoing coronary artery bypass grafting (CABG), to study
the effect of hUC-MSCs in improving the heart function of
these patients (Shanghai East Hospital 2021).
UC-derived MSCs
UC-derived MSCs (UC-MSCs) are formed during fifth week
AD-derived MSCs
of embryogenesis. The UC is a rich source of MSCs that can
be isolated from its different parts, including blood, UC AD was first identified as a source of MSCs for cell therapy
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

endothelium, and wharton’s jelly (Mushahary et al. 2018). by Zuk et al. (2001). The AD-derived MSCs (AD-MSCs) are
The UC-MSCs express both adult stem cell markers and ESC isolated from stroma of the AD using collagenase-mediated
markers like Tra 1-60, Tra 1-81, and stage-specific embryonic degradation, yielding progenitor cells that can differentiate
antigen 1 and 4 along with alkaline phosphatase (Carlin et into osteogenic, adipogenic, or chondrogenic cell types (Zuk
al. 2006). These MSCs exhibit higher multipotency compared et al. 2002). Due to easier access to autologous AD compared
to adipose or BM derivatives (Fong et al. 2011). Similar to BM- to BM and UC, as well as the abundance of fat tissue for their
MSCs, UC-MSCs are known to suppress T cell proliferation, isolation, AD-MSCs are sought after candidates for multiple
but their rejection time is slower compared to BM-MSCs; pathologies (Zuk 2013; Frese et al. 2016). In addition to this,
therefore, they are favored for allogeneic transplantation these cells are also advantageous in terms of shorter dou-
(Weiss et al. 2008; Deuse et al. 2011). Additionally, UC-MSCs bling time and generation of significantly higher stem cell
exhibit longer telomeres and higher telomerase activity, progenitors compared to BM-MSCs (Zhu et al. 2012). AD-MSCs
higher proliferation rates, and ease of isolation through least can differentiate into CMs and display a later onset of senes-
invasiveness, compared to other multipotent stem cell types, cence, signifying their potential for use in treating chronic
making them a promising source of MSCs for cardiac stem cardiac conditions (Tambrchi et al. 2022). Some studies report
cell therapy (Yannarelli et al. 2013b; Fazzina et al. 2016). In a the differentiation of AD-MSCs into cells of different vascular
study using mouse MI models, animals receiving human UC- lineages, aiding in cardiovascular remodeling (Ni et al. 2019).
MSCs (hUC-MSCs) sheets showed significant improvement In addition to this, generation of CMs has also been reported
in EF and improved infarct size, compared to untreated by several researchers through spontaneous or chemical-
controls. The authors also reported low oncogenicity as well induced differentiation in vitro, indicating the potential of
as reduced TNF-α secretion (Gao et al. 2022). Another group AD-MSCs for cardiac regeneration (Planat-Bénard et al. 2004;
of researchers showed antiapoptotic and proangiogenic role Choi et al. 2010; Jiang et al. 2018). In one such study, Chang et
of UC-MSCs in murine MI models. They also found enhanced al. demonstrated that rats receiving AD-MSCs after MI showed
activation of resident cardiac progenitors cells in the UC- improved heart function. After 12 weeks of transplantation,
MSC-treated animals (Nascimento et al. 2014). Additionally, the authors also observed expression of CM markers, includ-
UC-MSCs-conditioned media have also been reported to ing GATA1 and myosin heavy chain in transplanted AD-MSCs
reduce inflammation in porcine myocardium by prevent- (Chang et al. 2011). Ammar et al. showed that AD-MSCs were
ing the activation of NF-κB pathway. Treated animals also able to alleviate the symptoms of cardiotoxicity in doxoru-
exhibited improved cardiac function and reduced fibrosis bicin (Dox)-treated diabetic rats. AD-MSC-treated diabetic
as seen by TGF-β and IL-6 levels (Liu et al. 2016). A similar rats showed reduced collagen deposition and smooth muscle
study in rats with dilated cardiomyopathy showed reduced actin (SMA) expression as well as reduced infiltration of im-
expression of TNFα, TGF-β1, and ERK1/2 in UC-MSC-treated mune cells in Dox-treated myocardium (Ammar et al. 2015).
animals, highlighting its antifibrotic role (Zhang et al. 2018). In porcine models of acute MI, intracoronary (IC) injection of
In a study by Yannarelli et al. (2013a), it was shown that cul- autologous AD-MSCs was associated with increased cardio-
ture of embryonic CMs with UC-MSCs resulted in significant protection and better cardiac perfusion with no significant
upregulation of cardiac-specific genetic markers compared to changes in EF or cardiac volumes (Bobi et al. 2017). Besides
CMs that were co-cultured with BM-MSCs. Some studies have the use of AD-MSCs alone, several studies showed positive
also shown the differentiation of UC-MSCs into myotubes, effects following the transplantation of AD sheets as such
smooth muscle cells, and beating CMs by culturing them un- in the ischemic myocardium (Hamdi et al. 2011). Kim et al.
der defined conditions (Hollweck et al. 2011; Tompkins et al. (2017) showed that the use of AD-MSC sheets in acute MI mod-
2018; Chen et al. 2021). Qui et al. show that cardiac allograft els resulted in better cell engraftment and higher growth
transplantation that was accompanied by UC-MSC injection factor and cytokine secretion to prevent adverse cardiac
had lesser major histocompatibility complex class-II (MHC-II) remodeling compared to hearts that received AD-MSCs in-
expression and survived longer compared to the animals jections. Given the positive outcomes of pre-clinical studies,
without UC-MSCs (Liu et al. 2016). Of the few clinical trials several clinical trials have been performed using AD-MSCs to
conducted yet in cardiac patients using UC-MSCs, RIMECARD assess changes in heart function following transplantation.

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 165


Canadian Science Publishing

In a randomized, double-blinded APOLLO trial, the safety 2022). In a study involving porcine MI model, the efficacy of
of AD-MSCs was tested for the first time in ST-elevated MI different delivery approaches was tested. Amongst endocar-
(STEMI) patients. IC infusion of AD-MSCs was deemed safe dial, IM and IC routes, IC injection showed the largest average
without coronary flow impediment. The authors reported number of engrafted cells compared to other ways of MSC
improved cardiac function and 50% reduction in scar forma- administration (Moscoso et al. 2009). One such study states
tion (Cytori Therapeutics 2013b). PRECISE clinical trial was that only 3.5% of the injected MSCs were confined in the
performed in 27 patients with ischemic cardiomyopathy. heart after 6 weeks of injection. However, it was also found
Patients receiving AD-MSCs showed better maximal oxygen that highest number of these cells were retained when MSCs
consumption at 18 months follow-up period, compared to were injected a day after MI (Kurtz 2008). Some of the stud-
untreated controls. The patients also showed improved LV ies have also reported the use of biomaterial-based patches to
mass and wall motion score index while preserving exercise deliver stem cells. There was improvement in the retention
capacity in AD-MSC-treated patients (Perin et al. 2014). The and engraftment of stem cells when the cells were delivered
Athena trial program conducted double-blinded study in 31 through patches (Park et al. 2019; Tabei et al. 2019). However,
ischemic cardiomyopathy patients. Although there was no given the variations in the cell type and characteristics, one
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

difference in LV function in AD-MSCs and placebo patients, mode of delivery may be preferred over the other based on
the AD-MSC-treated patients showed improved cardiac per- the target organ and patient condition to achieve the best
fusion and reduced 12 month hospitalization rate (Henry et possible outcomes (Campbell and Suzuki 2012; Li et al. 2021).
al. 2017). In a recent phase II clinical trial involving patients
with heart failure with reduced EF (HFrEF), the patients who
Challenges involved in clinical translation of
received AD-MSCs showed improved quality of life; however,
the ventricular function and dimensions did not change
stem cells
Stem cell technology has certainly shown progress towards
compared to placebo group, after a 3-year follow-up period
clinical use in treatment of heart diseases (Table 1). However,
(Qayyum et al. 2023). Given the mixed results from different
there are several challenges and shortcomings that need to be
trials, there is a need to perform more extensive studies to
abridged before stem cell therapy becomes a norm in clinical
understand the role of AD-MSCs in improving heart function.
settings (Fig. 2). Following are some of the hurdles for the
translation of stem cell therapy.
Delivery of stem cells to the heart
The route and technique of stem cell delivery to the heart
Heterogeneity of stem cells
are crucial factors in contributing towards the clinical out-
come of cell therapy (Labusca et al. 2018). Stem cells can To begin with, it is very challenging to characterize MSCs,
be delivered to the injured myocardium via IM, intravenous since these cells lack the expression of unique surface mark-
(IV), and IC routes, or using 3D scaffolds (Rheault-Henry et al. ers and express markers that are present on other cell types
2021). IM injection involves direct delivery of stem cells in the as well. Therefore, even though there are standards defined
ischemic myocardial tissue. It is considered to be most precise by the International Society for Cellular Therapy, these are
approach to deliver cells and can be carried out with the help quite ambiguous and can be applied to other cell types
of catheters in patients with coronary heart disease (Sheng et (Nesselmann et al. 2008). For instance, MSCs show marked
al. 2013). Though IM injection offers targeted delivery of stem expression of CD29 (integrin ß1), which is also expressed
cells, this mode is invasive and can result in severe negative by epithelial and myoepithelial cells (Togarrati et al. 2018).
effects, including patient deaths. For similar reasons, it is not Similarly, CD90 (Thy-1) is co-expressed by hematopoeitc stem
feasible to inject multiple doses of cells in the same patient cells and keratinocytic stem cells (Kisselbach et al. 2009). In
(Sheng et al. 2013). On the other hand, IV approach involves addition to this, CD105 (endoglin), is expressed by endothe-
infusion of stem cells in the blood stream using IV drip. It is lial and certain hematopoietic stem cells as well (Nesselmann
generally preferred in patients after an acute MI, and due to et al. 2008; Rossi et al. 2019). Besides MSCs, pluripotent stem
its least invasive nature, it is a viable route to deliver multiple cells have also been reported for heterogeneity at different
cell infusions. However, despite being amongst the common levels. Since ESCs and iPSCs are known for their indefinite
methods of cell delivery, not many of the cells transplanted multiplication and pluripotent nature, it is challenging to
via IV mode survive and might end up being trapped in the control the differentiated cell population generated by these
blood vessels or untargeted organs (Yukawa et al. 2012). Addi- cells. Each ESC and iPSC cell line exhibits differences in dif-
tionally, this technique relies on homing of stem cells in the ferentiation potential favoring variable cell types (Yamanaka
injured myocardium using paracrine signaling (Sheng et al. 2020). Amongst other factors, major contributors towards
2013). IC infusion allows the cells to be delivered in the my- interculture variability include the time they have been in
ocardium via one of the coronary arteries. It is the most preva- culture, as well as genetic factors (Hayashi et al. 2019). Lately,
lent mode of cell delivery since it can be accompanied by the analysis of single cell cultures has revealed the presence of
surgical intervention in patients with MI. This method, how- different cell types in a single population of pluripotent stem
ever, might not be feasible in patients with less perfused ves- cells, adding to the complexity of stem cell hetrogeneity
sels. In addition to this, it might be important to consider the (Yang et al. 2021). In a study by Yang et al., exome sequencing
size of cells and the arteries before delivery to prevent cells revealed the presence of almost 400 variants of iPSCs in
from being obstructed in smaller capillaries (Bilewska et al. mouse fibroblast-derived 24 iPSC clones (C. Li et al. 2015).

166 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

Table 1. Summary of clinical trials completed in the past decade using stem cells from different sources, including bone
marrow, adipose tissue, and umbilical cord.
Route of National clinical trial
Study, phase (Year) Phase Cell type administration Major findings number (reference)
Safety and efficacy of 2 Autologous CD34+ stem Intracoronary –Improved LVEF NCT00629018
autologous stem cells Increased 6 min walk (Vrtovec 2015)
cells for dilated distance
cardiomyopathy –Reduced levels of
(2006–2013) NT——proBNP
APOLLO Trial 1 Adipose tissue-derived Intracoronary –Improved cardiac NCT00442806 (Cytori
(2007–2012) stem and regenerative function and perfusion Therapeutics
cells (ADRCs) defect 2013b)
–Decreased scar size
–Evidence on
pro-angiogenic,
anti-apoptotic, and
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

immunomodulatory
effects of ADRCs
PRECISE Trial 1 ADRCs Direct injection in –Maximal oxygen NCT00426868 (Cytori
(2007–2012) the left ventricle consumption and Therapeutics
metabolic equivalents 2013a)
preserved
–Improved left ventricular
mass and wall motion
score index
TAC–HFT (2008–2014) 1/2 Autologous MSCs and Injections with an –No adverse events after a NCT00768066 (Hare
bone marrow infusion catheter 1-month follow-up 2015a)
mononuclear cells Minnesota Living with
(BMCs) Heart Failure score
improved at 1 year
follow-up for both
treatment groups
PROCHYMAL 2 Allogeneic MSCs Intravenous –Reduction in ventricular NCT00877903 (Hare
(2009–2016) tachycardia et al. 2009)
POSEIDON–pilot 1 Autologous and Transendocardial –Reduction in infarct size NCT01087996 (Hare
study (2010–2013) allogeneic BM-MSCs –Improved ventricular 2015b)
remodeling
MESAMI (2010–2014) 1/2 Autologous MSCs Transendocardial –Improved EF and LVESV NCT01076920
(University
Hospital, Toulouse
2014)
POSEIDON–DCM 1/2 Autologous and Transendocardial –Improved endocardial NCT01392625 (Hare
(2011–2017) allogeneic BM-MSCs function and increased 2018)
EF
–TNFα suppression
RIMECARD Trial 1/2 Umbilical cord-derived Intravenous –Improved LVEF NCT01739777
(2012–2015) MSCs (UB-MSCs) –Improved functional (Bartolucci et al.
capacity 2017)
–Improved quality of life
TRIDENT (2013–2017) 2 Allogeneic MSCs Transendocardial –Increases EF NCT02013674 (Florea
–Reduced scar size et al. 2017)
To assess the safety of 2 Allogeneic BM-MSCs Intravenous –Improved functional NCT02467387
ischemia tolerant capacity (CardioCell LLC
MSCs (2014–2017) –Improved 2020)
immunomodulatory
markers
SCIENCE Trial 2 Allogeneic AD-MSCs Intramyocardial –Slight improvement in NCT02673164
(2016–2021) New York Heart (JKastrup 2021)
Association (NYHA) class
To assess the safety of 1/2 Hydrogel-packed Intramyocardial –No adverse effects 1 year NCT02635464 (Dai
UC-MSCs with UC-MSCs after therapy 2020)
injectable collagen Enhanced cardiac function
scaffold
(2015–2019)

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 167


Canadian Science Publishing

Table 1. (concluded).
Route of National clinical trial
Study, phase (Year) Phase Cell type administration Major findings number (reference)
Chronic myocardial 2 Allogeneic cardiology Intramyocardial –No significant changes NCT03092284
ischemia stem cell centre observed between (JKastrup 2022)
(2015–2022) adipose tissue-derived groups with respect to
mesenchymal stromal myocardial
cell product function/structure; no
(CSCC_ASC) changes in 6 min walk
test

Fig. 2. Challenges in clinical translation of stem cell therapy for heart repair. (i) Heterogeneity of stem cells: the differentiation
potential of stem cells varies between different stem cell types as well as within same stem cell type, which contributes to
the variability in the effects seen following treatment. (ii) Dose optimization: it is important to optimise the dose of stem
cells for transplantation. Given different cell numbers can contribute to different end results based on cell type and organ
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

of transplantation, cell numbers being transplanted should be optimized before clinical adaptation of stem cell therapy. (iii)
Migration of stem cells to untargeted organs can result in cell loss soon after transplantation or even obstruction of smaller
capillaries. (iv) Loss of stem cell immunoprivilege post transplantation: it results in the recognition of stem cell by host immune
response resulting in eventual loss from the site of transplantation. Immune suppression of stem cells varies with cell type
and source, which should be considered before transplantation for better survival in donor heart. Part of this figure was created
using the software bioicons (https://bioicons.com/).

Heterogeneity/
Uncontrolled differentiation

Cardiac
Patient

Stem cells
cultured in vitro

Dose optimization
tion
n
Loss of stem cell immune privilege

Migration

Using single cell RNA sequencing, another study revealed the Some of the major contributors towards cell death in the is-
variation in human PSC-derived limbal stem cells, in a time- chemic myocardium include reduced blood supply, increased
dependent manner (Sun et al. 2021). Furthermore, lack of oxidative stress, and enhanced inflammation in the my-
availability of standard protocols contributes to generation ocardium (Abdelwahid et al. 2016). Therefore, to overcome
of variable degrees of pluripotent stem cells in different labs these challenges, enhancing stem cell survival in injured
(Pattison et al. 2018). All these factors contribute significantly heart muscle by improving cell engraftment, inhibiting cell
towards results with low reproducibility and more variability death pathways, and increasing the expression of cell sur-
following transplantation of ESCs and iPSCs. vival proteins might be important for their clinical transla-
tion (Abdelwahid et al. 2016). Consequently, genetic modifi-
cation of MSCs to increase the secretion of pro-survival genes
Survival of transplanted stem cells in vivo
has been practiced to improve stem cell survival in the heart.
Besides the struggles associated with characterization of Noiseux et al. demonstrated that MSCs overexpressing Akt
stem cells, there is still a long way to go to ensure longer sur- show marked reduction in apoptosis and increased secretion
vival and engraftment of MSCs in the ischemic myocardium. of VEGF and insulin growth factor (IGF), thereby showing im-

168 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

proved heart function post MI (Kang et al. 2015; Shan et al. The size of coronary capillaries is limited to about 7–10 μm.
2018). Similarly, another study showed the effect of increased Therefore, to prevent microinfarctions due to microvascular
stretching on MSCs in improving their angiogenic potential plugging, it is important to consider the size of bigger stem
and decreasing apoptosis by targeting VEGF signaling (Zhu cells before injection (Gallet et al. 2015; Ishikawa 2015). Gao
et al. 2015). Several other studies have shown positive effects et al. conducted a randomized controlled trial in 18–80-year
induced by hypoxic pre-conditioning of stem cells prior to old patients with STEMI. According to this study, IC injec-
transplantation (Yan et al. 2012; Hao et al. 2019). Induction of tion of MSCs can easily result in adherence of these cells to
chemical or physically established hypoxia had similar effects the capillaries and affect the blood flow in the heart (Gao
on improving stem cell survival in vivo. It is also shown that et al. 2013). Furthermore, another study reported the trap-
MSCs pre-conditioned with caspase inhibitors in the presence ping of MSCs in lungs soon after IV injection, due to larger
of hyperoxia improved cell survival in vivo (Saini et al. 2013). size of MSCs (Eggenhofer et al. 2014; Masterson et al. 2021).
Hu et al. compared the improvement in cell survival and rate In another study comparing multipotent stem cells, neural
of apoptosis between normally cultured MSCs and hypoxia progenitor cells, BM mononuclear cells, and MSCs, the au-
preconditioned MSCs in primate model of MI. After 3 months, thors found that smaller sized cells were able to bypass the
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

a significant improvement in scar size and LV function was pulmonary microvascular barrier compared to larger MSCs
noted due to the secretion of pro-angiogenic factors such as (Fischer et al. 2009). Similar findings were presented by an-
HGF, erythropoietin, and angiopoietin-I (Hu et al. 2016). Many other study where the patients showed pulmonary embolism
researchers propose that due to hypoxic microenvironment after IV injection of AD-MSCs (Jung et al. 2013).
in the body, 2%–5% O2 levels is helpful for enhanced thera-
peutic potential of stem cells as well as preventing sponta-
neous differentiation of ESCs (Ezashi et al. 2005; Rodrigues Immunogenicity of stem cells after
et al. 2010; Antebi et al. 2018). Furthermore, poor survival transplantation
of iPSCs due to their inability to adapt to the environment
MSCs are considered to be immunoprivileged; therefore,
has been reported following transplantation, due to insuffi-
these cells should be able to evade host immune system and
cient vascularization and the incidence of necrosis (Cho et
avoid immune rejection. However, several studies have re-
al. 2021). To address this challenge, the co-administration of
ported that MSCs become immunogenic after transplanta-
a fibrin patch containing IGF-encapsulated microspheres has
tion in the ischemic heart (Dhingra et al. 2013; Abu-El-Rub
been proposed to provide this growth factor and increase sur-
et al. 2020; Sareen et al. 2020). Therefore, another major
vival and cardiac function. Ye et al. (2014) found that the en-
challenge in clinical translation of MSC-based therapies is
graftment of iPSCs was improved when combined with a fib-
to maintain the immunoprivilege of stem cells after trans-
rin patch, resulting in around 20-fold higher engraftment.
plantation. Huang et al. investigated whether MSCs maintain
their immunoprivilege and functional capacity after trans-
Migration and homing of stem cells in vivo plantation in a rat MI model. Assessment of cardiac environ-
ment at 6 months showed that allogeneic MSCs switch their
Another major challenge associated with stem cell therapy immune-tolerant state to immune-reactive state (Huang et al.
is the migration of cells to the site of injury as well as their 2010). Alteration in immune state has been reported due to
lower retention at the transplantation site (Guo et al. 2020). multiple reasons, including change in the expression of cell
According to some reports, expression of surface markers, surface immune antigen, MHC-II molecules on the surface
cell size as well as culture conditions of stem cells, plays an of MSCs, or reduced secretion of immunosuppressive soluble
important role in their engraftment and migration to the tar- factors such as PGE2 (Abu-El-Rub et al. 2020; Liu et al. 2020;
get site. In one such study, stem cells over expressing CXCR4 Sareen et al. 2020). Immune activity of stem cells can also be
were able to migrate and engraft better in the ischemic my- attributed to age of donor as well as the source of stem cells
ocardium promoting angiogenesis (Zhang et al. 2008). Sim- (Hass et al. 2011; Gao et al. 2016). Wu et al. (2014) showed that
ilarly, Becker et al. showed that migration of MSCs was as- both BM-MSCs and ADSCs from older animals show lesser
sociated with the expression of matrix metalloprotienase- suppression of CD4+ , CD8+ T cells compared to the cell iso-
2 (MMP-2). They also reported that higher confluence of lated from younger animals, highlighting the important role
MSCs in cultures resulted in increased synthesis of MMP- of donor age in regulating stem cell immunoprivilege. Simi-
2 inhibitor, therefore lesser cell migration (De Becker and larly, Ribeiro et al. (2013) compared the immunosuppressive
Riet 2016). Furthermore, to achieve better migration of trans- abilities of MSCs sourced from adult and neonatal tissues and
planted cells to the target organ, stem cells can be pre-treated found that these cells differ in their ability to suppress the
with certain chemokines and factors, including TNFα and in- CD4+ and CD8+ T cell activation, emphasizing further on the
tegrin β1 (Segers et al. 2006; Ip et al. 2007; Wu and Zhao importance of source of stem cells for transplantation.
2012). Inhibition of adhesion molecules including VCAM, on
the surface of MSCs has also been reported to positively affect
their translocation to the target organ (Chen et al. 2019). In Uncontrolled differentiation of stem cells in vivo
addition to this, higher culture temperatures negatively af- resulting in undesired phenotype
fect stem cell migration through NF-κβ pathway (Sen and Ta
2020). Stem cell size is another important factor, which needs Some other studies have reported concerns over differenti-
to be considered before the injection of stem cells in heart. ation of stem cells into cells of undesired phenotype. Follow-

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 169


Canadian Science Publishing

ing transplantation, when cells are exposed to host microen- absence of contaminants or genetic abnormalities (Guadix et
vironment, they might respond to the available factors and al. 2019; Henn et al. 2023). This will also be a key step in
undergo spontaneous differentiation (Fowler et al. 2020). In unlocking the value of demonstrating process comparabil-
addition to this, the CMs generated following differentiation ity, allowing for any necessary process changes and multisite
of iPSCs or ESCs might be of immature phenotype and there- manufacturing models (Martin et al. 2019). Fourth, scaling
fore exhibit differences in calcium handling and sarcomere up the production of stem cells will significantly increase the
organization (van den Berg et al. 2015; Rheault-Henry et al. cost. Optimizing production methods, including automation
2021; Campostrini et al. 2022). This might result in increased and bioreactor systems, can help improve efficiency and re-
arrhythmias following transplantation of these cells in the duce labor and material costs (Lee et al. 2022). Lastly, ethi-
myocardium. Furthermore, in some of the older studies, un- cal and regulatory considerations must be addressed while
fractionated BM cells have been shown to differentiate into scaling up the stem cells production. Meeting regulatory
osteoblasts in some cases resulting in ossification of the heart requirements and obtaining necessary approvals can be a
tissue. Therefore, it is advisable to use sufficient quantities of complex and time-consuming process, adding to the chal-
MSCs in the perfusate while ensuring their purity simultane- lenges of large-scale production (Bauer 2004; Lo and Parham
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

ously (Breitbach et al. 2007). To overcome these hurdles, it 2009).


might be better to use differentiated CMs derived from stem Addressing these challenges requires multidisciplinary
cells for cardiac repair and regeneration (Hasan et al. 2016). collaboration between scientists, engineers, clinicians, and
Transplantation of cells treated with factors for directed dif- regulatory experts. Continued research and technological
ferentiation might be another approach to consider. Boland advancements will play a crucial role in overcoming these
et al. suggested that adding Wnt-3 in the culture media of hurdles and enabling the large-scale production of stem cells
MSCs represses the osteogenic capacity of MSCs (Narcisi et for clinical applications.
al. 2015). On the other hand, it might be beneficial to rule
out the use of cells altogether and consider cell-derived prod-
ucts instead. Cell-free systems are gaining popularity since
Future directions: stem cell therapy for cardiac
they combine best of both the worlds. They are able to escape
regeneration
the immune response since they lack the expression of anti-
gen presenting cell surface proteins. Moreover, they exhibit Stem cell therapy has shown promise for cardiac regenera-
similar positive effects as seen by whole cell transplantation tion and repair. However, for clinical translation of stem cell-
(Keshtkar et al. 2018; Jafarinia et al. 2020). based approaches, the above-described challenges need to be
addressed. The heterogeneity of stem cell isolation and cul-
ture procedures can be addressed by standardizing the me-
Challenges with large-scale production of stem dia components being used for each stem cell type. Optimiz-
cells for clinical application ing the media components for clinical use is a daunting task
To replace the CMs lost during an ischemic event, treat- at present. However, reports suggest that standardizing cul-
ment using exogenous stem cells has shown a great promise. ture conditions is beneficial in preventing interculture vari-
However, to achieve expected results, these cells need to ability. It has been reported that iPSCs cultured under stan-
be expanded to higher numbers before transplantation in dard conditions had lesser variation in their genome (Molina-
patients. These large stem cell numbers are obtained us- Ruiz et al. 2022). According to many researchers fetal bovine
ing bioreactors. Nevertheless, large-scale production of stem serum (FBS) is the most widely used component with unde-
cells faces major challenges. There are a lot of aspects that fined composition in the culture medium, and it might pose
need to be considered when executing these steps, beginning some health risks associated with the transfer of xenogeneic
from donor selection to the final purified product ready to components into human body (Dessels et al. 2016). However,
be administered in vivo. First, there is a lack of standard- it has also been associated with reduced expression of p53-
ized/optimized protocol for sourcing, isolating, and main- related genes along with increased reprogramming efficiency
taining sufficient number of high-quality stem cells (Lappin of stem cells (Kwon et al. 2016). Therefore, it is inevitable for
and Cheng 2021). Heathman et al. (2016) highlighted the sig- cell culture media and requires better standardization. Des-
nificance of understanding the impact of donor features on sels et al. highlighted various alternates for stem cell culture
expanding autologous and allogeneic cell-based treatment replacing the serum component either through growth fac-
bioprocesses in developing manufacturing processes from tors or through serum albumin, replacing fetal calf serum
numerous BM-hMSC donors. Second, stem cells possess lim- completely. For human cell cultures, different options such
ited proliferative capability affecting the scope of their scala- as plasma, platelet-rich plasma, and plasma lysate can be con-
bility and expansion. To achieve large-scale production, stem sidered (Dessels et al. 2016). In one such study, Kocaoemer et
cells need to be expanded in vitro while maintaining their al. examined the effect of human AB-serum and thrombin-
stemness and avoiding genetic abnormalities or other alter- activated platelet-rich plasma as substitutes for FBS. The au-
ations that could affect their safety and efficacy (Brown et al. thors reported that differentiation of ADSCs into osteogenic
2013; Neri 2019). Third, it is important to undergo quality and adipogenic lineages was significantly lower in case of
control and characterization to ensure the safety and efficacy cells cultured in bovine serum compared to the serum ob-
of stem cells for clinical use. This involves comprehensive tained from human donors (Kocaoemer et al. 2007). Similarly,
characterization to confirm the identity, purity, potency, and Atashi et al. (2015) showed higher cell proliferation following

170 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

culture of stem cells in the presence of autologous platelet- vasculopathy. The MQDs were found to exhibit immunomod-
rich plasma. A meta-analysis by Palombella et al. compared ulatory effects by specifically decreasing the activation of
the variations in different parameters between FBS-based me- human CD4+ IFN-gamma+ T-lymphocytes and enhancing the
dia versus human platelet lysate (HPL)-based culture media. proliferation of immunosuppressive CD4+ CD25+ FoxP3+ reg-
They measured doubling time, expression of stem cell mark- ulatory T-cells (Rafieerad et al. 2021). Use of quantum dots to
ers, paracrine activity of MSCs, immunosuppressive abilities, trace the route of injected MSCs in vivo has been reported in
and cell morphology. The authors found that the doubling some recent studies. These materials can provide a substitute
time of MSCs was higher in HPL-supplemented media. How- for fluorescent molecules and organic dyes owing to their
ever, no differences in immunosuppressive abilities were re- photostability (Muller-Borer et al. 2007).
ported in both the conditions (Palombella et al. 2022). On the
other hand, Copland et al. (2013) suggested that the presence
of fibrinogen in HPL could negatively impact the immuno-
Exosomes as cell-free systems for cardiac
suppressive ability of MSCs due to its ability to trigger the
regeneration
pro-inflammatory phenotype of MSCs by activation of TLR4
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

receptor. One of the potential solutions could be to reduce Lately, EVs have emerged as promising cell-free systems
the fibrinogen content in HPL. However, this issue still needs for cardiac regeneration due to their advantages including
to be addressed in future studies to ensure safety of the stem lack of immune response caused by cell surface markers
cell procedures since most of the manufacturing facilities and no limitation to injection doses due to smaller sizes (Ha
have begun to use HPL to culture stem cells (Kinzebach and et al. 2016). Additionally, they can be targeted to specific
Bieback 2013). tissues or organs by modification of their cargos (Minghua
et al. 2018). Based on their size, EVs can be divided into
microvesicles (100–1000 nm), apoptotic bodies (50 nm–5 μm)
Cell-free systems for cardiac regeneration or exosomes (30–150 nm). Of these, exosomes are one of the
most studied EVs (Sanz-Ros et al. 2023). Stem cell-derived
Biomaterials for cardiac regeneration
exosomes have been reported to yield positive results similar
Cell retention and survival after transplantation are the to cell therapy through paracrine mechanisms (Gallet et al.
major barriers to achieving the full therapeutic efficacy of 2017). Exosomes contain nucleic acids, proteins, lipids, and
stem cells. Therefore, investigating newer avenues using several other factors, which have been reported to play a
biomaterials with or without stem cells for regeneration of positive role in cardiac disease models, including atrial fib-
the myocardium is becoming an active area of research. The rillation, MI, and cardiac hypertrophy (Fu et al. 2020; Meng
biomaterials offer a conducive 3D microenvironment to in- et al. 2020). Moreover, by acting as carriers of important
jected stem cells while masking the surface antigens that are miRNAs, exosomes can regulate inflammation and apoptosis
responsible for triggering an immune response. In addition resulting in reduced expression of fibrotic and inflammatory
to this, the biomaterials also offer efficient environment for markers and improving heart function (Sareen et al. 2023).
growth and support of endogenous or injected cells aiding In a study by Shao et al. (2017), it was demonstrated that the
in cardiac regeneration (Majid et al. 2020). Some of the most miRNA sequence profile for MSC-derived exosomes (MSCs-Ex)
commonly used biomaterials for cardiac regeneration in- was similar to MSCs, indicating exosomes to be the probable
clude organic compounds like polyethylene glycol; inorganic reason for positive effects of MSCs. Zhang et al. (2019) demon-
compounds, including silica nanoparticles, gold nanopar- strated that exosomes secreted by hypoxia-preconditioned
ticles, carbon nanoparticles as well as biological materials, BM-MSC overexpressing miRNA-24 suppressed CM death in
including viral like particles, protein nano systems as well as rats. A study conducted by Cheng et al. illustrated the effect of
EVs (Rafieerad et al. 2019, 2020; Han et al. 2022). Proteins such EVs overexpressing HIF1α on CMs in rats. Through paracrine
as laminins, fibronectins, collagen, and polysaccharides have signaling, these exosomes reduced scarring and increased
also been employed for use as scaffolds mimicking natural ex- the survival of the CMs under hypoxic conditions (Cheng et
tra cellular matrix. These semi-scaffolds can serve as binding al. 2020). Particularly for pluripotent stem cells, exosomes
sites for growth factors such as fibroblast growth factor (FGF), can prevent the risk of teratoma formation as demonstrated
VEGF, and HGF (Rosellini et al. 2018). In a recently concluded by Adamiak et al. The authors compared the effect of iPSCs
clinical trial to demonstrate the safety and efficacy of colla- and iPSC-EVs in MI models. Though both the groups showed
gen hydrogels for stem cell delivery to the heart, the patients similar positive effects on heart tissue, iPSC group showed
receiving CABG were additionally subjected to either a “cells cardiac tumors that were absent in iPSC-EV-treated animals
with collagen” treatment or “cells” by itself. The patients (Adamiak et al. 2018). Similar to MSCs, according to a study
who received hydrogel-packed MSCs demonstrated improved in pluripotent cells, exosomes from iPSC-derived CMs (iPSC-
LVEF with no adverse effects 1 year after the therapy (He et CMs Ex) were reported to have miRNA profile similar to
al. 2020). Jin et al. (2009) administered MSCs embedded in iPSC-CMs. The iPSC-CM Ex group showed reduced apoptosis
poly(lactide-co-epsilon-caprolactone) into a rat model of MI and cardiac fibrosis along with improved cardiac function
and noted that it considerably improved LVEF and reduced post treatment (Santoso et al. 2020). This study serves as
infarct size. Rafieerad et al. examined the impact of using an important source of potential target transcripts of the
0D titanium carbide MXene quantum dots (MQDs) on im- exosomes and thereby determine suitable cargoes that may
mune response regulation with the aim of treating allograft enhance their therapeutic benefits. It is also noteworthy that

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 171


Canadian Science Publishing

iPSC-CMs-EVs have also been shown to improve wall thick- investigated whether the upregulation of ERBB4 in MSCs has
ness, reduce cardiac hypertrophy, and promote angiogenesis a role in increasing cardio protection in a mouse model of MI.
in porcine models without any safety concerns related to fi- The ERBB4 overexpression reduced apoptosis, accelerated CM
brous tissues formation and unwanted remodeling (Gao et al. division, and decreased the infarct size. Moreover, it also pro-
2020). Currently, a clinical trial (NCT03384433) is underway moted cell migration and rendered the MSCs resistant to hy-
that aims to examine the effect of miR-124 containing allo- poxic and low glucose conditions of the cardiac milieu (Liang
geneic MSCs-Ex in patients with acute ischemic stroke. The et al. 2015). Hence, it is reasonable to profess that MSCs mod-
study will assess the safety of these exosomes over a 1-year ified to overexpress ERBB4 have significant cardioprotective
period using a modified ranking scale (Dehghani 2021). benefits.
Another promising approach includes the application of
nanoparticles imitating EVs to target MI. A study conducted Conclusion
by Yao et al. demonstrates the possibility of using a nano
complex that imitates an exosome, specifically mesoporous Stem cell therapy has the potential to offer a long-term
silica nanoparticles that are covered with MSC membranes. promise to improve the condition of patients with heart dis-
ease. The stem cells exhibit paracrine effects to suppress the
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

The membrane coating is a camouflaging strategy for the


immune system, as the nanoparticles can efficiently reach immune response in the ischemic heart favoring reverse re-
the target area and decrease interactions with other envi- modeling. Findings from multiple studies have supported
ronments along the way. This nanocomplex also included their role in improving heart function, reducing cardiac fi-
miRNA, specifically miR-21, which is an essential part of the brosis, preventing CM death, and promoting vascular regen-
exosome. It has been shown to promote regeneration in in- eration of the myocardium post ischemia. However, the field
farcted cardiac muscle and decrease CM apoptosis (Yao et al. of stem cells lacks standardization in terms of cell culture
2020). The potential use of nanoparticles in cardiac repair is techniques and cell characterization, which might be the con-
critical because this technique is less invasive and can im- tributing factor towards the variable results seen in clinics.
prove delivery. These can be addressed by defining standard operating pro-
tocols for all the laboratories to follow. Stem cells also face is-
sues like host immune rejection and migration to undesired
Genetic manipulation of stem cells tissues post transplantation. However, use of synthetic bio-
Innumerable gene modification therapies have been inves- materials for their direct injection in the myocardium and
tigated so far with the aim of bringing stem cell therapy EVs as cargos for their paracrine factors is a viable alternate
to patient-care settings. These genetic modifications can en- to cell therapy. Though these strategies are feasible, more in-
hance cell survival, improve cardiac-specific differentiation depth studies are required to assess their efficacy and safety
of stem cells, improve secretion of paracrine factors, or re- for translational application in cardiac patients.
duce immune response generated by transplanted stem cells
(Korshunova et al. 2020). AD-MSCs treated with lentiviral par- Acknowledgements
ticles for enhanced secretion of IFG-1 showed improved heart This work was supported by funding from the Canadian In-
function compared to non-transduced AD-MSCs. IGF1+ AD- stitutes of Health Research to SD. NS was supported by “Go
MSCs also showed enhanced secretion of VEGF and HGF for it” Project of Fondazione CRUI (Italy).
(Bagno et al. 2016). Moreover, Li et al. showed that when
MSCs are genetically manipulated for increased expression
of IGF-1 and treated with 5-azacytidine (5-AZA) in a porcine Article information
model, they triggered to differentiate into CM-like cells to
a higher degree compared to cells treated with 5-AZA alone History dates
(J. Li et al. 2015). Another strategy used to increase the mi- Received: 2 June 2023
gration and survival of MSCs is genetically engineering them Accepted: 2 August 2023
to overexpress stromal-derived factor-1, which is one of the Version of record online: 16 January 2024
most common chemokines playing a role in cell migration
(Nowakowski et al. 2016). Additionally, genetic modification Notes
of MSCs to overexpress immunosuppressive factors like cy- This paper is a part of a collection entitled "Physiology of
clooxygenase 2 has been reported to enhance their survival Adaptation and Medicine." It comprises papers from the ISAM
rate in ischemic rat hearts (Sareen et al. 2020). The MHC-II Conference held in Orlando, Florida, October 25-28, 2022.
and human leukocyte antigen (HLA) are major contributors https://cdnsciencepub.com/topic/cjpp-physiology-adaptation-
towards immune response generation following stem cell medicine.
transplantation. Therefore, some studies focus on preventing
the upregulation of MHC-II to prevent immune response gen- Copyright
eration in MSCs (Abu-El-Rub et al. 2019, 2020). Another group © 2024 The Author(s). This work is licensed under a Creative
of researchers engineered PSCs to express HLA-E genes at B2M Commons Attribution 4.0 International License (CC BY 4.0),
locus, without surface expression of HLA-A, B, or C, making which permits unrestricted use, distribution, and reproduc-
them resistant to natural killer cell lysis and were unrecog- tion in any medium, provided the original author(s) and
nizable by CD8+ T cells (Gornalusse et al. 2017). Liang et al. source are credited.

172 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

Data availability enchymal stem cells on cardiopathy]). Circ. Res. 121(10): 1192–1204.
No data were generated. doi:10.1161/CIRCRESAHA.117.310712.
Batalov, I., and Feinberg, A.W. 2015. Differentiation of cardiomy-
ocytes from human pluripotent stem cells using monolayer culture.
Author information Biomark. Insights, 10(Suppl 1): 71–76. doi:10.4137/BMI.S20050.
Batalov, I., Jallerat, Q., Kim, S., Bliley, J., and Feinberg, A.W. 2021. En-
Author ORCIDs gineering aligned human cardiac muscle using developmentally in-
spired fibronectin micropatterns. Sci. Rep. 11(1): 11502. doi:10.1038/
Elika Verma https://orcid.org/0000-0002-6642-7280 s41598-021-87550-y.
Sanjiv Dhingra https://orcid.org/0000-0002-2664-7633 Bauer, S.R. 2004. Stem cell-based products in medicine: FDA regulatory
considerations. In Handbook of stem cells. pp. 805–814. doi:10.1016/
Author contributions B978-012436643-5/50163-2.
Bilewska, A., Abdullah, M., Mishra, R., Musialek, P., Gunasekaran, M.,
Conceptualization: JKG, SKR, NS, SD Saha, P., et al. 2022. Safety and efficacy of transcoronary transfer of
Data curation: NS human neonatal stem cells to ischemic myocardium using a novel
Funding acquisition: SD cell-delivery system (CIRCULATE catheter) in swine model of acute
Methodology: JKG, SKR, EV, NS, SD myocardial infarction. Postepy Kardiol. Interwencyjnej, 18(4): 431–
438. doi:10.5114/aic.2022.121697.
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

Project administration: SD Blinova, K., Schocken, D., Patel, D., Daluwatte, C., Vicente, J., Wu, J.C.,
Resources: SD and Strauss, D.G. 2019. Clinical trial in a dish: personalized stem
Supervision: SD cell-derived cardiomyocyte assay compared with clinical trial results
Writing – original draft: JKG, SKR, EV, NS, SD for two QT-prolonging drugs. Clin. Transl. Sci. 12(6): 687–697. doi:10.
1111/cts.12674.
Writing – review & editing: JKG, EV, NS, SD Bobi, J., Solanes, N., Fernández-Jiménez, R., Galán-Arriola, C., Dantas,
A.P., Fernández-Friera, L., et al. 2017. Intracoronary administration of
Competing interests allogeneic adipose tissue-derived mesenchymal stem cells improves
The authors declare no conflict of interest to this work. myocardial perfusion but not left ventricle function, in a transla-
tional model of acute myocardial infarction. J. Am. Heart Assoc. 6(5):
e005771. doi:10.1161/JAHA.117.005771.
References Boomsma, R.A., Swaminathan, P.D., and Geenen, D.L. 2007. Intra-
Abdelwahid, E., Kalvelyte, A., Stulpinas, A., de Carvalho, K.A.T., Guarita- venously injected mesenchymal stem cells home to viable my-
Souza, L.C., and Foldes, G. 2016. Stem cell death and survival in ocardium after coronary occlusion and preserve systolic function
heart regeneration and repair. Apoptosis, 21(3): 252–268. doi:10.1007/ without altering infarct size. Int. J. Cardiol. 122(1): 17–28. doi:10.
s10495-015-1203-4. 1016/j.ijcard.2006.11.022.
Abu-El-Rub, E., Sequiera, G.L., Sareen, N., Yan, W., Moudgil, M., Sabbir, Breitbach, M., Bostani, T., Roell, W., Xia, Y., Dewald, O., Nygren,
M.G., and Dhingra, S. 2019. Hypoxia-induced 26S proteasome dys- J.M., et al. 2007. Potential risks of bone marrow cell transplanta-
function increases immunogenicity of mesenchymal stem cells. Cell tion into infarcted hearts. Blood, 110(4): 1362–1369. doi:10.1182/
Death Dis. 10(2): Article 2. doi:10.1038/s41419-019-1359-x. blood-2006-12-063412.
Abu-El-Rub, E., Sareen, N., Sequiera, G.L., Ammar, H.I., Yan, W., Sham- Brown, P.T., Handorf, A.M., Jeon, W.B., and Li, W.-J. 2013. Stem cell-
sEldeen, A.M., et al. 2020. Hypoxia-induced increase in Sug1 leads to based tissue engineering approaches for musculoskeletal regener-
poor post-transplantation survival of allogeneic mesenchymal stem ation. Curr. Pharmaceut. Design, 19(19): 3429–3445. doi:10.2174/
cells. FASEB J. 34(9): 12860–12876. doi:10.1096/fj.202000454R. 13816128113199990350.
Adamiak, M., Cheng, G., Bobis-Wozowicz, S., Zhao, L., Kedracka-Krok, S., Cai, M., Shen, R., Song, L., Lu, M., Wang, J., Zhao, S., et al. 2016. Bone
Samanta, A., et al. 2018. Induced pluripotent stem cell (iPSC)–derived marrow mesenchymal stem cells (BM-MSCs) improve heart function
extracellular vesicles are safer and more effective for cardiac repair in swine myocardial infarction model through paracrine effects. Sci.
than iPSCs. Circ. Res. 122(2): 296–309. doi:10.1161/CIRCRESAHA.117. Rep. 6(1): 1–12. doi:10.1038/srep28250.
311769. Campbell, N.G., and Suzuki, K. 2012. Cell delivery routes for stem cell
Ammar, H.I., Sequiera, G.L., Nashed, M.B., Ammar, R.I., Gabr, H.M., El- therapy to the heart: current and future approaches. J. Cardiovasc.
sayed, H.E., et al. 2015. Comparison of adipose tissue- and bone Transl. Res. 5(5): 713–726. doi:10.1007/s12265-012-9378-3.
marrow-derived mesenchymal stem cells for alleviating doxorubicin- Campostrini, G., Kosmidis, G., Ward-van Oostwaard, D., Davis, R.P., Yian-
induced cardiac dysfunction in diabetic rats. Stem Cell Res. Ther. 6: gou, L., Ottaviani, D., et al. 2022. Maturation of hiPSC-derived car-
148. doi:10.1186/s13287-015-0142-x. diomyocytes promotes adult alternative splicing of SCN5A and re-
Ando, M., Nishimura, T., Yamazaki, S., Yamaguchi, T., Kawana-Tachikawa, veals changes in sodium current associated with cardiac arrhythmia.
A., Hayama, T., et al. 2015. A safeguard system for induced pluripotent Cardiovasc. Res. cvac059. doi:10.1093/cvr/cvac059.
stem cell-derived rejuvenated T cell therapy. Stem Cell Rep. 5(4): 597– CardioCell LLC. 2020. A Phase IIa randomized study to assess the safety,
608. doi:10.1016/j.stemcr.2015.07.011. tolerability, and preliminary efficacy of a single intravenous dose
Antebi, B., Rodriguez, L.A., Walker, K.P., Asher, A.M., Kamucheka, R.M., of ischemia-tolerant allogeneic mesenchymal bone marrow cells to
Alvarado, L., et al. 2018. Short-term physiological hypoxia potentiates subjects with non-ischemic heart Failure (Clinical Trial Registration
the therapeutic function of mesenchymal stem cells. Stem Cell Res. NCT02467387). Available from https://classic.clinicaltrials.gov/ct2/s
Ther. 9(1): 265. doi:10.1186/s13287-018-1007-x. how/NCT02467387.
Atashi, F., Jaconi, M.E.E., Pittet-Cuénod, B., and Modarressi, A. 2015. Au- Cardiovascular diseases (CVDs). 2021. Available from https://www.wh
tologous platelet-rich plasma: a biological supplement to enhance o.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) [ac-
adipose-derived mesenchymal stem cell expansion. Tissue Eng. C cessed 14 April 2022].
Methods, 21(3): 253–262. doi:10.1089/ten.tec.2014.0206. Carlin, R., Davis, D., Weiss, M., Schultz, B., and Troyer, D. 2006. Expres-
Bagno, L.L., Carvalho, D., Mesquita, F., Louzada, R.A., Andrade, B., Kasai- sion of early transcription factors Oct-4, Sox-2 and Nanog by porcine
Brunswick, T.H., et al. 2016. Sustained IGF-1 secretion by adipose- umbilical cord (PUC) matrix cells. Reprod. Biol. Endocrinol. 4(1): 8.
derived stem cells improves infarcted heart function. Cell Transplant. doi:10.1186/1477-7827-4-8.
25(9): 1609–1622. doi:10.3727/096368915X690215. Chang, J.-C., Lee, P.-C., Lin, Y.-C., Lee, K.-W., and Hsu, S. 2011. Primary
Bartolucci, J., Verdugo, F.J., González, P.L., Larrea, R.E., Abarzua, E., Goset, adipose-derived stem cells enriched by growth factor treatment im-
C., et al. 2017. Safety and efficacy of the intravenous infusion of proves cell adaptability toward cardiovascular differentiation in a ro-
umbilical cord mesenchymal stem cells in patients with heart fail- dent model of acute myocardial infarction. J. Stem Cells, 6(1): 21–37.
ure: a phase 1/2 randomized controlled trial (RIMECARD trial [ran- Chen, Q., Li, Y., Chen, Z., Du, H., and Wan, J. 2019. Anti-VCAM 1 antibody-
domized clinical trial of intravenous infusion umbilical cord mes- coated mesenchymal stromal cells attenuate experimental colitis via

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 173


Canadian Science Publishing

immunomodulation. Med. Sci. Monit. 25: 4457–4468. doi:10.12659/ Dhingra, S., Li, P., Huang, X.-P., Guo, J., Wu, J., Mihic, A., et al. 2013.
MSM.914238. Preserving prostaglandin E2 level prevents rejection of implanted
Chen, Y., Shen, H., Ding, Y., Yu, Y., Shao, L., and Shen, Z. 2021. The ap- allogeneic mesenchymal stem cells and restores postinfarction ven-
plication of umbilical cord-derived MSCs in cardiovascular diseases. tricular function. Circulation, 128(11_suppl_1): S69–S78. doi:10.1161/
J. Cell. Mol. Med. 25(17): 8103–8114. doi:10.1111/jcmm.16830. CIRCULATIONAHA.112.000324.
Cheng, H., Chang, S., Xu, R., Chen, L., Song, X., Wu, J., et al. 2020. Hypoxia- Dixon, J.E., Dick, E., Rajamohan, D., Shakesheff, K.M., and Denning, C.
challenged MSC-derived exosomes deliver miR-210 to attenuate post- 2011. Directed differentiation of human embryonic stem cells to in-
infarction cardiac apoptosis. Stem Cell Res. Ther. 11(1): 224. doi:10. terrogate the cardiac gene regulatory network. Mol. Ther. 19(9): 1695–
1186/s13287-020-01737-0. 1703. doi:10.1038/mt.2011.125.
Cho, S., Lee, C., Skylar-Scott, M.A., Heilshorn, S.C., and Wu, J.C. 2021. du Pré, B.C., Doevendans, P.A., and van Laake, L.W. 2013. Stem cells for
Reconstructing the heart using iPSCs: engineering strategies and ap- cardiac repair: an introduction. J. Geriatr. Cardiol. 10(2): 186. doi:10.
plications. J. Mol. Cell. Cardiol. 157: 56–65. doi:10.1016/j.yjmcc.2021. 3969/j.issn.1671-5411.2013.02.003.
04.006. Eggenhofer, E., Luk, F., Dahlke, M.H., and Hoogduijn, M.J. 2014. The life
Choi, Y.S., Dusting, G.J., Stubbs, S., Arunothayaraj, S., Han, X.L., Collas, and fate of mesenchymal stem cells. Front. Immunol. 5: 148. doi:10.
P., et al. 2010. Differentiation of human adipose-derived stem cells 3389/fimmu.2014.00148.
into beating cardiomyocytes. J. Cell. Mol. Med. 14(4): 878–889. doi:10. Ezashi, T., Das, P., and Roberts, R.M. 2005. Low O2 tensions and the pre-
1111/j.1582-4934.2010.01009.x. vention of differentiation of hES cells. Proc. Natl. Acad. Sci. U.S.A.
Chong, J.J.H., Yang, X., Don, C.W., Minami, E., Liu, Y.-W., Weyers, J.J., 102(13): 4783–4788. doi:10.1073/pnas.0501283102.
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

et al. 2014. Human embryonic-stem-cell-derived cardiomyocytes re- Fazzina, R., Iudicone, P., Fioravanti, D., Bonanno, G., Totta, P., Zizzari,
generate non-human primate hearts. Nature, 510(7504): 273–277. I.G., and Pierelli, L. 2016. Potency testing of mesenchymal stromal
doi:10.1038/nature13233. cell growth expanded in human platelet lysate from different human
Choudhery, M.S., Khan, M., Mahmood, R., Mehmood, A., Khan, S.N., and tissues. Stem Cell Res. Ther. 7(1): 122. doi:10.1186/s13287-016-0383-3.
Riazuddin, S. 2012. Bone marrow derived mesenchymal stem cells Fischer, U.M., Harting, M.T., Jimenez, F., Monzon-Posadas, W.O., Xue, H.,
from aged mice have reduced wound healing, angiogenesis, prolif- Savitz, S.I., et al. 2009. Pulmonary passage is a major obstacle for in-
eration and anti-apoptosis capabilities. Cell Biol. Int. 36(8): 747–753. travenous stem cell delivery: the pulmonary first-pass effect. Stem
doi:10.1042/CBI20110183. Cells Dev. 18(5): 683–691. doi:10.1089/scd.2008.0253.
Cong, X., Zhang, S.-M., Ellis, M.W., and Luo, J. 2019. Large animal models Florea, V., Rieger, A.C., DiFede, D.L., El-Khorazaty, J., Natsumeda, M.,
for the clinical application of human induced pluripotent stem cells. Banerjee, M.N., et al. 2017. Dose comparison study of allogeneic
Stem Cells Dev. 28(19): 1288–1298. doi:10.1089/scd.2019.0136. mesenchymal stem cells in patients with ischemic cardiomyopa-
Copland, I.B., Garcia, M.A., Waller, E.K., Roback, J.D., and Galipeau, J. thy (the TRIDENT study). Circ. Res. 121(11): 1279–1290. doi:10.1161/
2013. The effect of platelet lysate fibrinogen on the functionality CIRCRESAHA.117.311827.
of MSCs in immunotherapy. Biomaterials, 34(32): 7840–7850. doi:10. Fong, C.-Y., Chak, L.-L., Biswas, A., Tan, J.-H., Gauthaman, K., Chan, W.-K.,
1016/j.biomaterials.2013.06.050. and Bongso, A. 2011. Human Wharton’s jelly stem cells have unique
Cytori Therapeutics. 2013a. A randomized clinical trial of adiPose- transcriptome profiles compared to human embryonic stem cells and
deRived stEm & regenerative cells In the treatment of patients with other mesenchymal stem cells. Stem Cell Rev. Rep. 7(1): 1–16. doi:10.
non revaScularizable ischEmic myocardium— —the PRECISE Trial (Clin- 1007/s12015-010-9166-x.
ical Trial Registration NCT00426868). Available from https://clinicaltr Fowler, J.L., Ang, L.T., and Loh, K.M. 2020. A critical look: challenges in
ials.gov/study/NCT00426868. differentiating human pluripotent stem cells into desired cell types
Cytori Therapeutics. 2013b. A randomized clinical trial of adiPOse- and organoids. Wiley Interdiscip. Rev. Dev. Biol. 9(3): e368. doi:10.
derived stem ceLLs In the treatment of patients with ST-elevation 1002/wdev.368.
myOcardial infarction— —The APOLLO trial (Clinical Trial Registration Fox, K.F., Cowie, M.R., Wood, D.A., Coats, A.J., Gibbs, J.S., Underwood,
NCT00442806). Available from https://clinicaltrials.gov/study/NCT00 S.R., et al. 2001. Coronary artery disease as the cause of incident heart
442806. failure in the population. Eur. Heart J. 22(3): 228–236. doi:10.1053/
Da Silva, J.S., and Hare, J.M. 2013. Cell-based therapies for myocardial euhj.2000.2289.
repair: emerging role for bone marrow-derived mesenchymal stem Frese, L., Dijkman, P.E., and Hoerstrup, S.P. 2016. Adipose tissue-derived
cells (MSCs) in the treatment of the chronically injured heart. Meth- stem cells in regenerative medicine. Transfus. Med. Hemother. 43(4):
ods Mol. Biol. 1037: 145–163. doi:10.1007/978-1-62703-505-7_8. 268–274. doi:10.1159/000448180.
Dai, J. 2020. The safety and efficacy assessment of human umbilical cord- Freystaetter, K., and Akowuah, E. 2021. Common cardiac surgical proce-
derived mesenchymal stem cells (hUC-MSCs) with injectable collagen dures: bypass, valve repair and replacement. Surgery (Oxford), 39(3):
scaffold transplantation for chronic ischemic cardiomyopathy (Clini- 156–163. doi:10.1016/j.mpsur.2021.01.005.
cal Trial Registration NCT02635464). Available from https://clinicaltr Fu, S., Zhang, Y., Li, Y., Luo, L., Zhao, Y., and Yao, Y. 2020. Extracellular
ials.gov/study/NCT02635464. vesicles in cardiovascular diseases. Cell Death Discov. 6: 68. doi:10.
De Becker, A., and Riet, I.V. 2016. Homing and migration of mesenchymal 1038/s41420-020-00305-y.
stromal cells: how to improve the efficacy of cell therapy? World J. Gallet, R., Dawkins, J., Valle, J., Simsolo, E., de Couto, G., Middleton, R.,
Stem Cells, 8(3): 73–87. doi:10.4252/wjsc.v8.i3.73. et al. 2017. Exosomes secreted by cardiosphere-derived cells reduce
Deans, R.J., and Moseley, A.B. 2000. Mesenchymal stem cells: biology scarring, attenuate adverse remodelling, and improve function in
and potential clinical uses. Exp. Hematol. 28(8): 875–884. doi:10.1016/ acute and chronic porcine myocardial infarction. Eur. Heart J. 38(3):
S0301-472X(00)00482-3. 201–211. doi:10.1093/eurheartj/ehw240.
Dehghani, L. 2021. Safety and efficacy of allogenic mesenchymal stem Gallet, R., Tseliou, E., Dawkins, J., Middleton, R., Valle, J., Angert, D.,
cells derived exosome on disability of patients with acute ischemic et al. 2015. Intracoronary delivery of self-assembling heart-derived
stroke: a randomized, single-blind, placebo-controlled, phase 1, 2 microtissues (cardiospheres) for prevention of adverse remodel-
trial (Clinical Trial Registration NCT03384433). Available from https: ing in a pig model of convalescent myocardial infarction. Circ.
//clinicaltrials.gov/study/NCT03384433. Cardiovasc. Interv. 8(5): e002391. doi:10.1161/CIRCINTERVENTIONS.
Deinsberger, J., Reisinger, D., and Weber, B. 2020. Global trends in clinical 115.002391.
trials involving pluripotent stem cells: a systematic multi-database Gao, F., Chiu, S.M., Motan, D.a.L., Zhang, Z., Chen, L., Ji, H.-L., et al. 2016.
analysis. NPJ Regen. Med. 5: 15. doi:10.1038/s41536-020-00100-4. Mesenchymal stem cells and immunomodulation: current status and
Dessels, C., Potgieter, M., and Pepper, M.S. 2016. Making the switch: al- future prospects. Cell Death Dis. 7(1): e2062. doi:10.1038/cddis.2015.
ternatives to fetal bovine serum for adipose-derived stromal cell ex- 327.
pansion. Front. Cell Dev. Biol. 4. doi:10.3389/fcell.2016.00115. Gao, L.R., Pei, X.T., Ding, Q.A., Chen, Y., Zhang, N.K., Chen, H.Y., et al.
Deuse, T., Stubbendorff, M., Tang-Quan, K., Phillips, N., Kay, M.A., Eier- 2013. A critical challenge: dosage-related efficacy and acute compli-
mann, T., et al. 2011. Immunogenicity and immunomodulatory prop- cation intracoronary injection of autologous bone marrow mesenchy-
erties of umbilical cord lining mesenchymal stem cells. Cell Trans- mal stem cells in acute myocardial infarction. Int. J. Cardiol. 168(4):
plant. 20(5): 655–667. doi:10.3727/096368910X536473. 3191–3199. doi:10.1016/j.ijcard.2013.04.112.

174 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

Gao, L., Wang, L., Wei, Y., Krishnamurthy, P., Walcott, G.P., Menasché, P., Hass, R., Kasper, C., Böhm, S., and Jacobs, R. 2011. Different populations
and Zhang, J. 2020. Exosomes secreted by hiPSC-derived cardiac cells and sources of human mesenchymal stem cells (MSC): a comparison
improve recovery from myocardial infarction in swine. Sci. Transl. of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 9:
Med. 12(561): eaay1318. doi:10.1126/scitranslmed.aay1318. 12. doi:10.1186/1478-811X-9-12.
Gao, S., Jin, Y., Ma, J., Wang, J., Wang, J., Shao, Z., et al. 2022. Preclini- Hatzistergos, K.E., Quevedo, H., Oskouei, B.N., Hu, Q., Feigenbaum, G.S.,
cal study of human umbilical cord mesenchymal stem cell sheets for Margitich, I.S., et al. 2010. Bone marrow mesenchymal stem cells
the recovery of ischemic heart tissue. Stem Cell Res. Ther. 13(1): 252. stimulate cardiac stem cell proliferation and differentiation. Circ.
doi:10.1186/s13287-022-02919-8. Res. 107(7): 913–922. doi:10.1161/CIRCRESAHA.110.222703.
Golpanian, S., El-Khorazaty, J., Mendizabal, A., DiFede, D.L., Suncion, V.Y., Hayashi, Y., Ohnuma, K., and Furue, M.K. 2019. Pluripotent stem
Karantalis, V., et al. 2015. Effect of aging on human mesenchymal cell heterogeneity. Adv. Exp. Med. Biol. 1123: 71–94. doi:10.1007/
stem cell therapy in ischemic cardiomyopathy patients. J. Am. Coll. 978-3-030-11096-3_6.
Cardiol. 65(2): 125–132. doi:10.1016/j.jacc.2014.10.040. He, X., Wang, Q., Zhao, Y., Zhang, H., Wang, B., Pan, J., et al. 2020. Ef-
Gornalusse, G.G., Hirata, R.K., Funk, S.E., Riolobos, L., Lopes, V.S., fect of intramyocardial grafting collagen scaffold with mesenchy-
Manske, G., et al. 2017. HLA-E-expressing pluripotent stem cells es- mal stromal cells in patients with chronic ischemic heart disease:
cape allogeneic responses and lysis by NK cells. Nat. Biotechnol. 35(8): a randomized clinical trial. JAMA Network Open, 3(9): e2016236.
765–772. doi:10.1038/nbt.3860. doi:10.1001/jamanetworkopen.2020.16236.
Groenewegen, A., Rutten, F.H., Mosterd, A., and Hoes, A.W. 2020. Epi- Heathman, T.R.J., Rafiq, Q.A., Chan, A.K.C., Coopman, K., Nienow, A.W.,
demiology of heart failure. Eur. J. Heart Fail. 22(8): 1342–1356. doi:10. Kara, B., and Hewitt, C.J. 2016. Characterization of human mesenchy-
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

1002/ejhf.1858. mal stem cells from multiple donors and the implications for large
Guadix, J.A., López-Beas, J., Clares, B., Soriano-Ruiz, J.L., Zugaza, J.L., and scale bioprocess development. Biochem. Eng. J. 108: 14–23. doi:10.
Gálvez-Martín, P. 2019. Principal criteria for evaluating the quality, 1016/j.bej.2015.06.018.
safety and efficacy of hMSC-based products in clinical practice: cur- Help Therapeutics. 2022. Epicardial injection of allogeneic human
rent approaches and challenges. Pharmaceutics, 11(11): 552. doi:10. pluripotent stem cell-derived cardiomyocytes to treat severe chronic
3390/pharmaceutics11110552. heart failure (Clinical Trial Registration NCT03763136). Available
Guo, Y., Yu, Y., Hu, S., Chen, Y., and Shen, Z. 2020. The therapeutic po- from https://clinicaltrials.gov/ct2/show/NCT03763136.
tential of mesenchymal stem cells for cardiovascular diseases. Cell Henn, A.D., Pereira, T., Hunsberger, J., Mitra, K., Izadifar, Z., Somara,
Death Dis. 11(5): Article 5. doi:10.1038/s41419-020-2542-9. S., et al. 2023. Cytocentric measurement for regenerative medicine.
Ha, D., Yang, N., and Nadithe, V. 2016. Exosomes as therapeutic drug car- Front. Med. Technol. 5. doi:10.3389/fmedt.2023.1154653.
riers and delivery vehicles across biological membranes: current per- Henry, T.D., Pepine, C.J., Lambert, C.R., Traverse, J.H., Schatz, R., Costa,
spectives and future challenges. Acta Pharmaceut. Sin. B, 6(4): 287– M., et al. 2017. The Athena trials: autologous adipose-derived regen-
296. doi:10.1016/j.apsb.2016.02.001. erative cells for refractory chronic myocardial ischemia with left
Hamdi, H., Planat-Benard, V., Bel, A., Puymirat, E., Geha, R., Pidial, L., ventricular dysfunction. Catheter. Cardiovasc. Interv. 89(2): 169–177.
et al. 2011. Epicardial adipose stem cell sheets results in greater post- doi:10.1002/ccd.26601.
infarction survival than intramyocardial injections. Cardiovasc. Res. Hentze, H., Soong, P.L., Wang, S.T., Phillips, B.W., Putti, T.C., and Dunn,
91(3): 483–491. doi:10.1093/cvr/cvr099. N.R. 2009. Teratoma formation by human embryonic stem cells: eval-
Han, X., Alu, A., Liu, H., Shi, Y., Wei, X., Cai, L., and Wei, Y. 2022. uation of essential parameters for future safety studies. Stem Cell Res.
Biomaterial-assisted biotherapy: a brief review of biomaterials used 2(3): 198–210. doi:10.1016/j.scr.2009.02.002.
in drug delivery, vaccine development, gene therapy, and stem cell Hollweck, T., Hartmann, I., Eblenkamp, M., Wintermantel, E., Re-
therapy. Bioact. Mater. 17: 29–48. doi:10.1016/j.bioactmat.2022.01. ichart, B., Uberfuhr, P., and Eissner, G. 2011. Cardiac differentia-
011. tion of human Wharton’s jelly stem cells— —experimental compari-
Hao, D., He, C., Ma, B., Lankford, L., Reynaga, L., Farmer, D.L., et al. 2019. son of protocols. Open Tissue Eng. Regen. Med. J. 4(1). doi:10.2174/
Hypoxic preconditioning enhances survival and proangiogenic ca- 1875043501104010095.
pacity of human first trimester chorionic villus-derived mesenchymal Hu, X., Xu, Y., Zhong, Z., Wu, Y., Zhao, J., Wang, Y., et al. 2016. A
stem cells for fetal tissue engineering. Stem Cells Int. 2019: 9695239. large-scale investigation of hypoxia-preconditioned allogeneic mes-
doi:10.1155/2019/9695239. enchymal stem cells for myocardial repair in nonhuman primates:
Hare, J.M. 2015a. A phase I/II, randomized, double-blinded, placebo- paracrine activity without remuscularization. Circ. Res. 118(6): 970–
controlled study of the safety and efficacy of transendocardial in- 983. doi:10.1161/CIRCRESAHA.115.307516.
jection of autologous human cells (bone marrow or mesenchymal) Huang, C.-C., Tsai, H.-W., Lee, W.-Y., Lin, W.-W., Chen, D.-Y., Hung, Y.-W.,
in patients with chronic ischemic left ventricular dysfunction and et al. 2013. A translational approach in using cell sheet fragments of
heart failure secondary to myocardial infarction. (Clinical Trial Reg- autologous bone marrow-derived mesenchymal stem cells for cellu-
istration NCT00768066). Available from https://clinicaltrials.gov/ct2/s lar cardiomyoplasty in a porcine model. Biomaterials, 34(19): 4582–
how/NCT00768066. 4591. doi:10.1016/j.biomaterials.2013.03.003.
Hare, J.M. 2015b. A phase I/II, randomized pilot study of the comparative Huang, X.-P., Sun, Z., Miyagi, Y., McDonald Kinkaid, H., Zhang, L., Weisel,
safety and efficacy of transendocardial injection of autologous mes- R.D., and Li, R.-K. 2010. Differentiation of allogeneic mesenchymal
enchymal stem cells versus allogeneic mesenchymal stem cells in pa- stem cells induces immunogenicity and limits their long-term ben-
tients with chronic ischemic left ventricular dysfunction secondary efits for myocardial repair. Circulation, 122(23): 2419–2429. doi:10.
to myocardial infarction (Clinical Trial Registration NCT01087996). 1161/CIRCULATIONAHA.110.955971.
Available from https://clinicaltrials.gov/ct2/show/NCT01087996. Ip, J.E., Wu, Y., Huang, J., Zhang, L., Pratt, R.E., and Dzau, V.J. 2007. Mes-
Hare, J.M. 2018. A phase I/II, randomized pilot study of the comparative enchymal stem cells use integrin beta1 not CXC chemokine receptor
safety and efficacy of transendocardial injection of autologous mes- 4 for myocardial migration and engraftment. Mol. Biol. Cell, 18(8):
enchymal stem cells versus allogeneic mesenchymal stem cells in pa- 2873–2882. doi:10.1091/mbc.e07-02-0166.
tients with non-ischemic dilated cardiomyopathy. (Clinical Trial Reg- Ishikawa, K. 2015. Intracoronary injection of large stem cells. Circ. Car-
istration NCT01392625). Available from https://clinicaltrials.gov/ct2/s diovasc. Interv. 8(5): e002648. doi:10.1161/CIRCINTERVENTIONS.115.
how/NCT01392625. 002648.
Hare, J.M., Traverse, J.H., Henry, T.D., Dib, N., Strumpf, R.K., Schulman, Jafarinia, M., Alsahebfosoul, F., Salehi, H., Eskandari, N., and
S.P., et al. 2009. A randomized, double-blind, placebo-controlled, Ganjalikhani-Hakemi, M. 2020. Mesenchymal stem cell-derived
dose-escalation study of intravenous adult human mesenchymal extracellular vesicles: a novel cell-free therapy. Immunol. Investig.
stem cells (prochymal) after acute myocardial infarction. J. Am. Coll. 49(7): 758–780. doi:10.1080/08820139.2020.1712416.
Cardiol. 54(24): 2277–2286. doi:10.1016/j.jacc.2009.06.055. Jiang, A., Chen, Y., Shi, L., and Li, F. 2018. Differentiation of brown
Hasan, A., Waters, R., Roula, B., Dana, R., Yara, S., Alexandre, T., and adipose-derived stem cells into cardiomyocyte-like cells is regu-
Paul, A. 2016. Engineered biomaterials to enhance stem cell-based lated by a combination of low 5-azacytidine concentration and
cardiac tissue engineering and therapy. Macromol. Biosci. 16(7): 958– bone morphogenetic protein 4. Int. J. Clin. Exp. Pathol. 11(11):
977. doi:10.1002/mabi.201500396. 5514–5524.

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 175


Canadian Science Publishing

Jiang, W., and Xu, J. 2020. Immune modulation by mesenchymal stem plications. Stem Cells Transl. Med. 10(s2): S1–S3. doi:10.1002/sctm.
cells. Cell Prolif. 53(1): e12712. doi:10.1111/cpr.12712. 21-0269.
Jin, J., Jeong, S.I., Shin, Y.M., Lim, K.S., Shin, H.soo, Lee, Y.M., et al. 2009. Lee, B., Jung, S., Hashimura, Y., Lee, M., Borys, B.S., Dang, T., et al. 2022.
Transplantation of mesenchymal stem cells within a poly(lactide-co-ε- Cell culture process scale-up challenges for commercial-scale manu-
caprolactone) scaffold improves cardiac function in a rat myocardial facturing of allogeneic pluripotent stem cell products. Bioengineer-
infarction model. Eur. J. Heart Fail. 11(2): 147–153. doi:10.1093/eurjhf/ ing (Basel, Switzerland), 9(3): 92. doi:10.3390/bioengineering9030092.
hfn017. Li, C., Klco, J.M., Helton, N.M., George, D.R., Mudd, J.L., Miller, C.A., et al.
JKastrup. 2021. Stem cell therapy in ischEmic non-treatable cardiac 2015. Genetic heterogeneity of induced pluripotent stem cells: results
disease— —SCIENCE a European Multi-centre Trial (Clinical Trial Reg- from 24 clones derived from a single C57BL/6 mouse. PLoS ONE, 10(3):
istration NCT02673164). Available from https://clinicaltrials.gov/ct2/s e0120585. doi:10.1371/journal.pone.0120585.
how/NCT02673164. Li, J., Zhu, K., Wang, Y., Zheng, J., Guo, C., Lai, H., and Wang, C. 2015. Com-
JKastrup. 2022. Allogeneic adipose tissue-derived stromal/stem cell ther- bination of IGF–1 gene manipulation and 5–AZA treatment promotes
apy in patients with ischemic heart disease and heart failure: differentiation of mesenchymal stem cells into cardiomyocyte–like
a phase II Danish Multicentre Study (Clinical Trial Registration cells. Mol. Med. Rep. 11(2): 815–820. doi:10.3892/mmr.2014.2812.
NCT03092284). Available from https://clinicaltrials.gov/ct2/show/NC Li, J., Hu, S., Zhu, D., Huang, K., Mei, X., López de Juan Abad, B., and
T03092284. Cheng, K. 2021. All roads lead to rome (the heart): cell retention and
Jung, J.W., Kwon, M., Choi, J.C., Shin, J.W., Park, I.W., Choi, B.W., and outcomes from various delivery routes of cell therapy products to
Kim, J.Y. 2013. Familial occurrence of pulmonary embolism after in- the heart. J. Am. Heart Assoc. 10(8): e020402. doi:10.1161/JAHA.120.
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

travenous, adipose tissue-derived stem cell therapy. Yonsei Med. J. 020402.


54(5): 1293–1296. doi:10.3349/ymj.2013.54.5.1293. Liang, X., Ding, Y., Zhang, Y., Chai, Y.-H., He, J., Chiu, S.-M., et al. 2015.
Kabat, M., Bobkov, I., Kumar, S., and Grumet, M. 2020. Trends in mes- Activation of NRG1-ERBB4 signaling potentiates mesenchymal stem
enchymal stem cell clinical trials 2004–2018: is efficacy optimal in a cell-mediated myocardial repairs following myocardial infarction.
narrow dose range? Stem Cells Transl. Med. 9(1): 17–27. doi:10.1002/ Cell Death Dis. 6(5): Article 5. doi:10.1038/cddis.2015.91.
sctm.19-0202. Lin, B., Li, Y., Han, L., Kaplan, A.D., Ao, Y., Kalra, S., et al. 2015. Mod-
Kadota, S., Tanaka, Y., and Shiba, Y. 2020. Heart regeneration using eling and study of the mechanism of dilated cardiomyopathy us-
pluripotent stem cells. J. Cardiol. 76(5): 459–463. doi:10.1016/j.jjcc. ing induced pluripotent stem cells derived from individuals with
2020.03.013. Duchenne muscular dystrophy. Dis. Models Mech. 8(5): 457–466.
Kang, K., Ma, R., Cai, W., Huang, W., Paul, C., Liang, J., et al. 2015. Exo- doi:10.1242/dmm.019505.
somes secreted from CXCR4 overexpressing mesenchymal stem cells Liu, C.-B., Huang, H., Sun, P., Ma, S.-Z., Liu, A.-H., Xue, J., et al. 2016. Hu-
promote cardioprotection via Akt signaling pathway following my- man umbilical cord-derived mesenchymal stromal cells improve left
ocardial infarction. Stem Cells Int. 2015: 659890. doi:10.1155/2015/ ventricular function, perfusion, and remodeling in a porcine model
659890. of chronic myocardial ischemia. Stem Cells Transl. Med. 5(8): 1004–
Kelishadi, R., and Poursafa, P. 2014. A review on the genetic, environ- 1013. doi:10.5966/sctm.2015-0298.
mental, and lifestyle aspects of the early-life origins of cardiovascu- Liu, S., Liu, F., Zhou, Y., Jin, B., Sun, Q., and Guo, S. 2020. Immunosup-
lar disease. Curr. Probl. Pediatr. Adolesc. Health Care, 44(3): 54–72. pressive property of MSCs mediated by cell surface receptors. Front.
doi:10.1016/j.cppeds.2013.12.005. Immunol. 11.
Keshtkar, S., Azarpira, N., and Ghahremani, M.H. 2018. Mesenchy- Liu, Y.-W., Chen, B., Yang, X., Fugate, J.A., Kalucki, F.A., Futakuchi-
mal stem cell-derived extracellular vesicles: novel frontiers in re- Tsuchida, A., et al. 2018. Human embryonic stem cell-derived
generative medicine. Stem Cell Res. Ther. 9(1): 63. doi:10.1186/ cardiomyocytes restore function in infarcted hearts of non-
s13287-018-0791-7. human primates. Nat. Biotechnol. 36(7): Article 7. doi:10.1038/nbt.
Kim, J.-H., Joo, H.J., Kim, M., Choi, S.-C., Lee, J.I., Hong, S.J., and Lim, D.- 4162.
S. 2017. Transplantation of adipose-derived stem cell sheet attenu- Lo, B., and Parham, L. 2009. Ethical issues in stem cell research. Endocr.
ates adverse cardiac remodeling in acute myocardial infarction. Tis- Rev. 30(3): 204–213. doi:10.1210/er.2008-0031.
sue Eng. A, 23(1–2): 1–11. doi:10.1089/ten.tea.2016.0023. Majid, Q.A., Fricker, A.T.R., Gregory, D.A., Davidenko, N., Hernandez
Kinzebach, S., and Bieback, K. 2013. Expansion of mesenchymal Cruz, O., Jabbour, R.J., et al. 2020. Natural biomaterials for cardiac tis-
stem/stromal cells under xenogenic-free culture conditions. In Mes- sue engineering: a highly biocompatible solution. Front. Cardiovasc.
enchymal stem cells— —basics and clinical application I. Edited by B. Med. 7: 554597. doi:10.3389/fcvm.2020.554597.
Weyand, M. Dominici, R. Hass, R. Jacobs and C. Kasper. Springer. Martin, I., Galipeau, J., Kessler, C., Le Blanc, K., and Dazzi, F. 2019.
pp. 33–57. doi:10.1007/10_2012_134. Challenges for mesenchymal stromal cell therapies. Sci. Transl. Med.
Kisselbach, L., Merges, M., Bossie, A., and Boyd, A. 2009. CD90 expression 11(480): eaat2189. doi:10.1126/scitranslmed.aat2189.
on human primary cells and elimination of contaminating fibrob- Masterson, C.H., Tabuchi, A., Hogan, G., Fitzpatrick, G., Kerrigan, S.W.,
lasts from cell cultures. Cytotechnology, 59(1): 31–44. doi:10.1007/ Jerkic, M., et al. 2021. Intra-vital imaging of mesenchymal stromal
s10616-009-9190-3. cell kinetics in the pulmonary vasculature during infection. Sci. Rep.
Kocaoemer, A., Kern, S., Klüter, H., and Bieback, K. 2007. Human AB 11(1): Article 1. doi:10.1038/s41598-021-83894-7.
serum and thrombin-activated platelet-rich plasma are suitable al- Menasché, P. 2020. Cell therapy with human ESC-derived cardiac cells:
ternatives to fetal calf serum for the expansion of mesenchymal stem clinical perspectives. Front. Bioeng. Biotechnol. 8. doi:10.3389/fbioe.
cells from adipose tissue. Stem Cells (Dayton, Ohio), 25(5): 1270–1278. 2020.601560.
doi:10.1634/stemcells.2006-0627. Menasché, P., Vanneaux, V., Hagège, A., Bel, A., Cholley, B., Cacciapuoti, I.,
Korshunova, I., Rhein, S., García-González, D., Stölting, I., Pfisterer, U., et al. 2015. Human embryonic stem cell-derived cardiac progenitors
Barta, A., et al. 2020. Genetic modification increases the survival and for severe heart failure treatment: first clinical case report. Eur. Heart
the neuroregenerative properties of transplanted neural stem cells. J. 36(30): 2011–2017. doi:10.1093/eurheartj/ehv189.
JCI Insight, 5(4): e126268. doi:10.1172/jci.insight.126268. Menasché, P., Vanneaux, V., Hagège, A., Bel, A., Cholley, B., Parouchev, A.,
Kurtz, A. 2008. Mesenchymal stem cell delivery routes and fate. Int. J. et al. 2018. Transplantation of human embryonic stem cell-derived
Stem Cells, 1(1): 1–7. doi:10.15283/ijsc.2008.1.1.1. cardiovascular progenitors for severe ischemic left ventricular dys-
Kwon, D., Kim, J.-S., Cha, B.-H., Park, K.-S., Han, I., Park, K.-S., et al. 2016. function. J. Am. Coll. Cardiol. 71(4): 429–438. doi:10.1016/j.jacc.2017.
The effect of fetal bovine serum (FBS) on efficacy of cellular repro- 11.047.
gramming for induced pluripotent stem cell (iPSC) generation. Cell Meng, W., He, C., Hao, Y., Wang, L., Li, L., and Zhu, G. 2020. Prospects and
Transplant. 25(6): 1025–1042. doi:10.3727/096368915X689703. challenges of extracellular vesicle-based drug delivery system: consid-
Labusca, L., Herea, D.D., and Mashayekhi, K. 2018. Stem cells as deliv- ering cell source. Drug Deliv. 27(1): 585–598. doi:10.1080/10717544.
ery vehicles for regenerative medicine-challenges and perspectives. 2020.1748758.
World J. Stem Cells, 10(5): 43–56. doi:10.4252/wjsc.v10.i5.43. Minghua, W., Zhijian, G., Chahua, H., Qiang, L., Minxuan, X., Luqiao,
Lappin, T., and Cheng, T. 2021. An urgent need for standardization W., et al. 2018. Plasma exosomes induced by remote ischaemic
of stem cells and stem cell-derived products toward clinical ap- preconditioning attenuate myocardial ischaemia/reperfusion injury

176 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

by transferring miR-24. Cell Death Dis. 9(3): Article 3. doi:10.1038/ Pattison, J.M., Melo, S.P., Piekos, S.N., Torkelson, J.L., Bashkirova, E.,
s41419-018-0274-x. Mumbach, M.R., et al. 2018. Retinoic acid and BMP4 cooperate
Molina-Ruiz, F.J., Introna, C., Bombau, G., Galofre, M., and Canals, J.M. with TP63 to alter chromatin dynamics during surface epithelial
2022. Standardization of cell culture conditions and routine genomic commitment. Nat. Genet. 50(12): 1658–1665. doi:10.1038/
screening under a quality management system leads to reduced s41588-018-0263-0.
genomic instability in hPSCs. Cells, 11(13): Article 13. doi:10.3390/ Perin, E.C., Sanz-Ruiz, R., Sánchez, P.L., Lasso, J., Pérez-Cano, R., Alonso-
cells11131984. Farto, J.C., et al. 2014. Adipose-derived regenerative cells in patients
Moscoso, I., Barallobre, J., de Ilarduya, Ó.M., Añón, P., Fraga, M., Calviño, with ischemic cardiomyopathy: the PRECISE trial. Am. Heart J. 168(1):
R., et al. 2009. Analysis of different routes of administration of het- 88–95.e2. doi:10.1016/j.ahj.2014.03.022.
erologous 5-azacytidine–treated mesenchymal stem cells in a porcine Planat-Bénard, V., Menard, C., André, M., Puceat, M., Perez, A., Garcia-
model of myocardial infarction. Transplant. Proc. 41(6): 2273–2275. Verdugo, J.-M., et al. 2004. Spontaneous cardiomyocyte differenti-
doi:10.1016/j.transproceed.2009.06.011. ation from adipose tissue stroma cells. Circ. Res. 94(2): 223–229.
Mu, Y., Cao, G., Zeng, Q., and Li, Y. 2011. Transplantation of induced bone doi:10.1161/01.RES.0000109792.43271.47.
marrow mesenchymal stem cells improves the cardiac function of Poomani, M.S., Mariappan, I., Perumal, R., Regurajan, R., Muthan, K., and
rabbits with dilated cardiomyopathy via upregulation of vascular en- Subramanian, V. 2022. Mesenchymal stem cell (MSCs) therapy for is-
dothelial growth factor and its receptors. Exp. Biol. Med. (Maywood, chemic heart disease: a promising frontier. Global Heart, 17(1): 19.
N.J.), 236(9): 1100–1107. doi:10.1258/ebm.2011.011066. doi:10.5334/gh.1098.
Muller-Borer, B.J., Collins, M.C., Gunst, P.R., Cascio, W.E., and Kypson, Qayyum, A.A., Mouridsen, M., Nilsson, B., Gustafsson, I., Schou, M.,
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

A.P. 2007. Quantum dot labeling of mesenchymal stem cells. J. Nielsen, O.W., et al. 2023. Danish phase II trial using adipose tissue
Nanobiotechnol. 5: 9. doi:10.1186/1477-3155-5-9. derived mesenchymal stromal cells for patients with ischaemic heart
Mushahary, D., Spittler, A., Kasper, C., Weber, V., and Charwat, V. 2018. failure. ESC Heart Fail. 10(2): 1170–1183. doi:10.1002/ehf2.14281.
Isolation, cultivation, and characterization of human mesenchymal Rafieerad, A., Yan, W., Sequiera, G.L., Sareen, N., Abu-El-Rub, E., Moudgil,
stem cells. Cytometry A, 93(1): 19–31. doi:10.1002/cyto.a.23242. M., and Dhingra, S. 2019. Application of Ti3C2 MXene quantum dots
Musunuru, K., Sheikh, F., Gupta, R.M., Houser, S.R., Maher, K.O., Milan, for immunomodulation and regenerative medicine. Adv. Healthcare
D.J., et al. 2018. Induced pluripotent stem cells for cardiovascular dis- Mater. 8(16): 1900569. doi:10.1002/adhm.201900569.
ease modeling and precision medicine: a scientific statement from Rafieerad, A., Sequiera, G.L., Yan, W., Kaur, P., Amiri, A., and Dhingra,
the American Heart Association. Circ. Genom. Precis. Med. 11(1): S. 2020. Sweet-MXene hydrogel with mixed-dimensional components
e000043. doi:10.1161/HCG.0000000000000043. for biomedical applications. J. Mech. Behav. Biomed. Mater. 101:
Narcisi, R., Cleary, M.A., Brama, P.A.J., Hoogduijn, M.J., Tüysüz, N., ten 103440. doi:10.1016/j.jmbbm.2019.103440.
Berge, D., and van Osch, G.J.V.M. 2015. Long-term expansion, en- Rafieerad, A., Yan, W., Alagarsamy, K.N., Srivastava, A., Sareen, N., Arora,
hanced chondrogenic potential, and suppression of endochondral os- R.C., and Dhingra, S. 2021. Fabrication of smart tantalum carbide
sification of adult human MSCs via WNT signaling modulation. Stem MXene quantum dots with intrinsic immunomodulatory properties
Cell Rep. 4(3): 459–472. doi:10.1016/j.stemcr.2015.01.017. for treatment of allograft vasculopathy. Adv. Funct. Mater. 31(46):
Nascimento, D.S., Mosqueira, D., Sousa, L.M., Teixeira, M., Filipe, M., Re- 2106786. doi:10.1002/adfm.202106786.
sende, T.P., et al. 2014. Human umbilical cord tissue-derived mes- Rheault-Henry, M., White, I., Grover, D., and Atoui, R. 2021. Stem cell
enchymal stromal cells attenuate remodeling after myocardial infarc- therapy for heart failure: medical breakthrough, or dead end? World
tion by proangiogenic, antiapoptotic, and endogenous cell-activation J. Stem Cells, 13(4): 236. doi:10.4252/wjsc.v13.i4.236.
mechanisms. Stem Cell Res. Ther. 5(1): 5. doi:10.1186/scrt394. Ribeiro, A., Laranjeira, P., Mendes, S., Velada, I., Leite, C., Andrade, P.,
Neri, S. 2019. Genetic stability of mesenchymal stromal cells for regen- et al. 2013. Mesenchymal stem cells from umbilical cord matrix, adi-
erative medicine applications: a fundamental biosafety aspect. Int. J. pose tissue and bone marrow exhibit different capability to suppress
Mol. Sci. 20(10): Article 10. doi:10.3390/ijms20102406. peripheral blood B, natural killer and T cells. Stem Cell Res. Ther. 4(5):
Nesselmann, C., Ma, N., Bieback, K., Wagner, W., Ho, A., Konttinen, Y.T., 125. doi:10.1186/scrt336.
et al. 2008. Mesenchymal stem cells and cardiac repair. J. Cell. Mol. Rikhtegar, R., Pezeshkian, M., Dolati, S., Safaie, N., Afrasiabi Rad, A.,
Med. 12(5b): 1795–1810. doi:10.1111/j.1582-4934.2008.00457.x. Mahdipour, M., et al. 2019. Stem cells as therapy for heart disease: IP-
Ni, H., Zhao, Y., Ji, Y., Shen, J., Xiang, M., and Xie, Y. 2019. Adipose-derived SCs, ESCs, CSCs, and skeletal myoblasts. Biomed. Pharmacother. 109:
stem cells contribute to cardiovascular remodeling. Aging (Albany 304–313. doi:10.1016/j.biopha.2018.10.065.
NY), 11(23): 11756–11769. doi:10.18632/aging.102491. Rodrigues, C.A.V., Diogo, M.M., da Silva, C.L., and Cabral, J.M.S. 2010. Hy-
Nowakowski, A., Walczak, P., Lukomska, B., and Janowski, M. 2016. Ge- poxia enhances proliferation of mouse embryonic stem cell-derived
netic engineering of mesenchymal stem cells to induce their migra- neural stem cells. Biotechnol. Bioeng. 106(2): 260–270. doi:10.1002/
tion and survival. Stem Cells Int. 2016: e4956063. doi:10.1155/2016/ bit.22648.
4956063. Rosellini, E., Zhang, Y.S., Migliori, B., Barbani, N., Lazzeri, L., Shin, S.R.,
Okano, H., Nakamura, M., Yoshida, K., Okada, Y., Tsuji, O., Nori, S., et al. 2018. Protein/polysaccharide-based scaffolds mimicking native
et al. 2013. Steps toward safe cell therapy using induced pluripotent extracellular matrix for cardiac tissue engineering applications. J.
stem cells. Circ. Res. 112(3): 523–533. doi:10.1161/CIRCRESAHA.111. Biomed. Mater. Res. A, 106(3): 769–781. doi:10.1002/jbm.a.36272.
256149. Rossi, E., Bernabeu, C., and Smadja, D.M. 2019. Endoglin as an adhesion
Padda, J., Sequiera, G.L., Sareen, N., and Dhingra, S. 2015. Stem cell ther- molecule in mature and progenitor endothelial cells: a function be-
apy for cardiac regeneration: hits and misses. Can. J. Physiol. Pharma- yond TGF-β. Front. Med. 6. doi:10.3389/fmed.2019.00010.
col. 93(10): 835–841. doi:10.1139/cjpp-2014-0468. Rossignol, P., Hernandez, A.F., Solomon, S.D., and Zannad, F. 2019. Heart
Palombella, S., Perucca Orfei, C., Castellini, G., Gianola, S., Lopa, S., Mas- failure drug treatment. Lancet, 393(10175): 1034–1044. doi:10.1016/
trogiacomo, M., et al. 2022. Systematic review and meta-analysis on S0140-6736(18)31808-7.
the use of human platelet lysate for mesenchymal stem cell cul- Saini, U., Gumina, R.J., Wolfe, B., Kuppusamy, M.L., Kuppusamy, P., and
tures: comparison with fetal bovine serum and considerations on Boudoulas, K.D. 2013. Preconditioning mesenchymal stem cells with
the production protocol. Stem Cell Res. Ther. 13(1): 142. doi:10.1186/ caspase inhibition and hyperoxia prior to hypoxia exposure increases
s13287-022-02815-1. cell proliferation. J. Cell. Biochem. 114(11): 2612–2623. doi:10.1002/
Parikh, S.S., Blackwell, D.J., Gomez-Hurtado, N., Frisk, M., Wang, L., Kim, jcb.24609.
K., et al. 2017. Thyroid and glucocorticoid hormones promote func- Santoso, M.R., Ikeda, G., Tada, Y., Jung, J., Vaskova, E., Sierra, R.G., et al.
tional T-tubule development in human-induced pluripotent stem 2020. Exosomes from induced pluripotent stem cell–derived car-
cell-derived cardiomyocytes. Circ. Res. 121(12): 1323–1330. doi:10. diomyocytes promote autophagy for myocardial repair. J. Am. Heart
1161/CIRCRESAHA.117.311920. Assoc. 9(6): e014345. doi:10.1161/JAHA.119.014345.
Park, S.-J., Kim, R.Y., Park, B.-W., Lee, S., Choi, S.W., Park, J.-H., et al. Sanz-Ros, J., Mas-Bargues, C., Romero-García, N., Huete-Acevedo, J., Dro-
2019. Dual stem cell therapy synergistically improves cardiac func- mant, M., and Borrás, C. 2023. Extracellular vesicles as therapeutic
tion and vascular regeneration following myocardial infarction. Nat. resources in the clinical environment. Int. J. Mol. Sci. 24(3): 2344.
Commun. 10: 3123. doi:10.1038/s41467-019-11091-2. doi:10.3390/ijms24032344.

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 177


Canadian Science Publishing

Sareen, N., Abu-El-Rub, E., Ammar, H.I., Yan, W., Sequiera, G.L., Sham- Benefit of cardiopoietic mesenchymal stem cell therapy on left ven-
sEldeen, A.M., et al. 2020. Hypoxia-induced downregulation of cy- tricular remodelling: results from the Congestive Heart Failure Car-
clooxygenase 2 leads to the loss of immunoprivilege of allogeneic diopoietic Regenerative Therapy (CHART-1) study. Eur. J. Heart Fail.
mesenchymal stem cells. FASEB J. 34(11): 15236–15251. doi:10.1096/ 19(11): 1520–1529. doi:10.1002/ejhf.898.
fj.202001478R. Toda, K. 2021. Clinical trial of human (allogeneic) iPS cell-derived
Sareen, N., Srivastava, A., Alagarsamy, K.N., Lionetti, V., and Dhingra, S. cardiomyocytes sheet for ischemic (Clinical Trial Registration
2023. Stem cells derived exosomes and biomaterials to modulate au- NCT04696328). Available from https://clinicaltrials.gov/ct2/show/NC
tophagy and mend broken hearts. Biochim. Biophys. Acta, 1869(7): T04696328.
166806. doi:10.1016/j.bbadis.2023.166806. Togarrati, P.P., Dinglasan, N., Desai, S., Ryan, W.R., and Muench, M.O.
Schuleri, K.H., Feigenbaum, G.S., Centola, M., Weiss, E.S., Zimmet, J.M., 2018. CD29 is highly expressed on epithelial, myoepithelial and mes-
Turney, J., et al. 2009. Autologous mesenchymal stem cells produce enchymal stromal cells of human salivary glands. Oral Dis. 24(4): 561–
reverse remodelling in chronic ischaemic cardiomyopathy. Eur. Heart 572. doi:10.1111/odi.12812.
J. 30(22): 2722–2732. doi:10.1093/eurheartj/ehp265. Tompkins, B.A., Balkan, W., Winkler, J., Gyöngyösi, M., Goliasch, G.,
Segers, V.F.M., Van Riet, I., Andries, L.J., Lemmens, K., Demolder, M.J., Fernández-Avilés, F., and Hare, J.M. 2018. Preclinical studies of stem
De Becker, A.J.M.L., et al. 2006. Mesenchymal stem cell adhesion cell therapy for heart disease. Circ. Res. 122(7): 1006–1020. doi:10.
to cardiac microvascular endothelium: activators and mechanisms. 1161/CIRCRESAHA.117.312486.
Am. J. Physiol. Heart Circ. Physiol. 290(4): H1370–H1377. doi:10.1152/ University Hospital, Toulouse. 2014. Administration of mesenchymal
ajpheart.00523.2005. stem cells in patients with chronic ischemic cardiomyopathy (Clin-
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

Sen, A., and Ta, M. 2020. Altered adhesion and migration of human mes- ical Trial Registration NCT01076920). Available from https://clinical
enchymal stromal cells under febrile temperature stress involves NF- trials.gov/ct2/show/NCT01076920.
κβ pathway. Sci. Rep. 10: 4473. doi:10.1038/s41598-020-61361-z. van den Berg, C.W., Okawa, S., Chuva de Sousa Lopes, S.M., van Iperen,
Sequiera, G.L., Srivastava, A., Sareen, N., Yan, W., Alagarsamy, K.N., L., Passier, R., Braam, S.R., et al. 2015. Transcriptome of human foetal
Verma, E., et al. 2022. Development of iPSC-based clinical trial se- heart compared with cardiomyocytes from pluripotent stem cells.
lection platform for patients with ultrarare diseases. Sci. Adv. 8(14): Development, 142(18): 3231–3238. doi:10.1242/dev.123810.
eabl4370. doi:10.1126/sciadv.abl4370. Volarevic, V., Markovic, B.S., Gazdic, M., Volarevic, A., Jovicic, N., Arsenije-
Shafei, A.E.-S., Ali, M.A., Ghanem, H.G., Shehata, A.I., Abdelgawad, A.A., vic, N., et al. 2018. Ethical and safety issues of stem cell-based therapy.
Handal, H.R., et al. 2017. Mesenchymal stem cell therapy: a promis- Int. J. Med. Sci. 15(1): 36–45. doi:10.7150/ijms.21666.
ing cell-based therapy for treatment of myocardial infarction. J. Gene Vrtovec, B. 2015. The effects of autologous intracoronary stem cell trans-
Med. 19(12):. doi:10.1002/jgm.2995. plantation in patients with end-stage dilated cardiomyopathy (Clini-
Shan, S., Liu, Z., Guo, T., Wang, M., Tian, S., Zhang, Y., et al. 2018. Growth cal Trial Registration NCT00629018). Available from https://clinicaltr
arrest-specific gene 6 transfer promotes mesenchymal stem cell sur- ials.gov/ct2/show/NCT00629018.
vival and cardiac repair under hypoxia and ischemia via enhanced au- Wang, T., Pine, A.R., Kotini, A.G., Yuan, H., Zamparo, L., Starczynowski,
tocrine signaling and paracrine action. Arch. Biochem. Biophys. 660: D.T., et al. 2021. Sequential CRISPR gene editing in human iPSCs
108–120. doi:10.1016/j.abb.2018.10.016. charts the clonal evolution of myeloid leukemia and identifies early
Shanghai East Hospital. 2021. Treatment of heart failure using human disease targets. Cell Stem Cell, 28(6): 1074–1089.e7. doi:10.1016/j.
umbilical cord mesenchymal stem cells (hUC-MSC) (Clinical Trial Reg- stem.2021.01.011.
istration NCT04939077). Available from https://clinicaltrials.gov/ct2/s Wei, X., Yang, X., Han, Z., Qu, F., Shao, L., and Shi, Y. 2013. Mesenchymal
how/NCT04939077. stem cells: a new trend for cell therapy. Acta Pharmacol. Sin. 34(6):
Shao, L., Zhang, Y., Lan, B., Wang, J., Zhang, Z., Zhang, L., et al. 2017. 747–754. doi:10.1038/aps.2013.50.
MiRNA-sequence indicates that mesenchymal stem cells and exo- Weiss, M.L., Anderson, C., Medicetty, S., Seshareddy, K.B., Weiss, R.J., Van-
somes have similar mechanism to enhance cardiac repair. BioMed derWerff, I., et al. 2008. Immune properties of human umbilical cord
Res. Int. 2017: e4150705. doi:10.1155/2017/4150705. Wharton’s jelly-derived cells. Stem Cells (Dayton, Ohio), 26(11): 2865–
Sheng, C.C., Zhou, L., and Hao, J. 2013. Current stem cell delivery meth- 2874. doi:10.1634/stemcells.2007-1028.
ods for myocardial repair. BioMed Res. Int. 2013: 547902. doi:10.1155/ Wong, S.S., and Bernstein, H.S. 2010. Cardiac regeneration using human
2013/547902. embryonic stem cells: producing cells for future therapy. Regen. Med.
Shiba, Y., Gomibuchi, T., Seto, T., Wada, Y., Ichimura, H., Tanaka, Y., et al. 5(5): 763–775. doi:10.2217/rme.10.52.
2016. Allogeneic transplantation of iPS cell-derived cardiomyocytes Wu, L.W., Wang, Y.-L., Christensen, J.M., Khalifian, S., Schneeberger,
regenerates primate hearts. Nature, 538(7625): 388–391. doi:10.1038/ S., Raimondi, G., et al. 2014. Donor age negatively affects the im-
nature19815. munoregulatory properties of both adipose and bone marrow de-
Stephen, J., Bravo, E.L., Colligan, D., Fraser, A.R., Petrik, J., and Camp- rived mesenchymal stem cells. Transplant Immunol. 30(4): 122–127.
bell, J.D.M. 2016. Mesenchymal stromal cells as multifunctional cellu- doi:10.1016/j.trim.2014.03.001.
lar therapeutics— —a potential role for extracellular vesicles. Transfus. Wu, Q., Wang, J., Tan, W.L.W., Jiang, Y., Wang, S., Li, Q., et al. 2020. Ex-
Apher. Sci. 55(1): 62–69. doi:10.1016/j.transci.2016.07.011. tracellular vesicles from human embryonic stem cell-derived cardio-
Sun, C., Wang, H., Ma, Q., Chen, C., Yue, J., Li, B., and Zhang, X. vascular progenitor cells promote cardiac infarct healing through re-
2021. Time-course single-cell RNA sequencing reveals transcriptional ducing cardiomyocyte death and promoting angiogenesis. Cell Death
dynamics and heterogeneity of limbal stem cells derived from Dis. 11(5): Article 5. doi:10.1038/s41419-020-2508-y.
human pluripotent stem cells. Cell Biosci. 11(1): 24. doi:10.1186/ Wu, Y., and Zhao, R.C.H. 2012. The role of chemokines in mesenchymal
s13578-021-00541-4. stem cell homing to myocardium. Stem Cell Rev. Rep. 8(1): 243–250.
Tabei, R., Kawaguchi, S., Kanazawa, H., Tohyama, S., Hirano, A., Handa, doi:10.1007/s12015-011-9293-z.
N., et al. 2019. Development of a transplant injection device for opti- Yamanaka, S. 2020. Pluripotent stem cell-based cell therapy— —promise
mal distribution and retention of human induced pluripotent stem and challenges. Cell Stem Cell, 27(4): 523–531. doi:10.1016/j.stem.
cell−derived cardiomyocytes. J. Heart Lung Transplant. 38(2): 203– 2020.09.014.
214. doi:10.1016/j.healun.2018.11.002. Yan, F., Yao, Y., Chen, L., Li, Y., Sheng, Z., and Ma, G. 2012. Hypoxic pre-
Tamargo, J., and López-Sendón, J. 2011. Novel therapeutic targets for conditioning improves survival of cardiac progenitor cells: role of
the treatment of heart failure. Nat. Rev. Drug Discov. 10(7): 536–555. stromal cell derived factor-1α–CXCR4 axis. PLoS ONE, 7(7): e37948.
doi:10.1038/nrd3431. doi:10.1371/journal.pone.0037948.
Tambrchi, P., Mahdavi, A.H., DaliriJoupari, M., and Soltani, L. 2022. Yan, W., Abu-El-Rub, E., Saravanan, S., Kirshenbaum, L.A., Arora, R.C., and
Polycaprolactone-co-polylactic acid nanofiber scaffold in combina- Dhingra, S. 2019. Inflammation in myocardial injury: mesenchymal
tion with 5-azacytidine and transforming growth factor-β to induce stem cells as potential immunomodulators. Am. J. Physiol. Heart Cir.
cardiomyocyte differentiation of adipose-derived mesenchymal stem Physiol. 317(2): H213–H225. doi:10.1152/ajpheart.00065.2019.
cells. Cell Biochem. Funct. 40(7): 668–682. doi:10.1002/cbf.3728. Yang, S., Cho, Y., and Jang, J. 2021. Single cell heterogeneity in hu-
Teerlink, J.R., Metra, M., Filippatos, G.S., Davison, B.A., Bartunek, J., man pluripotent stem cells. BMB Rep. 54(10): 505–515. doi:10.5483/
Terzic, A.CHART Investigators, and et al.,CHART Investigators, 2017. BMBRep.2021.54.10.094.

178 Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202


Canadian Science Publishing

Yang, X., Pabon, L., and Murry, C.E. 2014. Eng. Adolesc. Circ. Res. 114(3): inhibiting TNF-α and TGF-β1/ERK1/2 signaling pathways. Mol. Med.
511–523. doi:10.1161/CIRCRESAHA.114.300558. Rep. 17(1): 71–78. doi:10.3892/mmr.2017.7882.
Yannarelli, G., Dayan, V., Pacienza, N., Lee, C.-J., Medin, J., and Keating, A. Zhang, C.-S., Shao, K., Liu, C.-W., Li, C.-J., and Yu, B.-T. 2019. Hypoxic pre-
2013a. Human umbilical cord perivascular cells exhibit enhanced car- conditioning BMSCs-exosomes inhibit cardiomyocyte apoptosis af-
diomyocyte reprogramming and cardiac function after experimen- ter acute myocardial infarction by upregulating microRNA-24. Eur.
tal acute myocardial infarction. Cell Transplant. 22(9): 1651–1666. Rev. Med. Pharmacol. Sci. 23(15): 6691–6699. doi:10.26355/eurrev_
doi:10.3727/096368912X657675. 201908_18560.
Yannarelli, G., Pacienza, N., Cuniberti, L., Medin, J., Davies, J., and Keat- Zhang, D., Fan, G.-C., Zhou, X., Zhao, T., Pasha, Z., Xu, M., et al. 2008. Over-
ing, A. 2013b. Brief report: the potential role of epigenetics on mul- expression of CXCR4 on mesenchymal stem cells augments myoan-
tipotent cell differentiation capacity of mesenchymal stromal cells. giogenesis in the infarcted myocardium. J. Mol. Cell. Cardiol. 44(2):
Stem Cells, 31(1): 215–220. doi:10.1002/stem.1262. 281–292. doi:10.1016/j.yjmcc.2007.11.010.
Yao, C., Wu, W., Tang, H., Jia, X., Tang, J., Ruan, X., et al. 2020. Zhu, W.-Z., Santana, L.F., and Laflamme, M.A. 2009. Local control of
Self-assembly of stem cell membrane-camouflaged nanocom- excitation–contraction coupling in human embryonic stem cell-
plex for microRNA-mediated repair of myocardial infarction derived cardiomyocytes. PLoS ONE, 4(4): e5407. doi:10.1371/journal.
injury. Biomaterials, 257: 120256. doi:10.1016/j.biomaterials.2020. pone.0005407.
120256. Zhu, X., Shi, W., Tai, W., and Liu, F. 2012. The comparition of biological
Ye, L., Chang, Y.-H., Xiong, Q., Zhang, P., Zhang, L., Somasundaram, P., characteristics and multilineage differentiation of bone marrow and
et al. 2014. Cardiac repair in a porcine model of acute myocardial adipose derived mesenchymal stem cells. Cell Tissue Res. 350(2): 277–
Can. J. Physiol. Pharmacol. Downloaded from cdnsciencepub.com by 119.18.3.4 on 04/16/24

infarction with human induced pluripotent stem cell-derived cardio- 287. doi:10.1007/s00441-012-1453-1.
vascular cell populations. Cell Stem Cell, 15(6): 750–761. doi:10.1016/ Zhu, Z., Gan, X., Fan, H., and Yu, H. 2015. Mechanical stretch endows
j.stem.2014.11.009. mesenchymal stem cells stronger angiogenic and anti-apoptotic ca-
Yukawa, H., Watanabe, M., Kaji, N., Okamoto, Y., Tokeshi, M., Miyamoto, pacities via NFκB activation. Biochem. Biophys. Res. Commun. 468(4):
Y., et al. 2012. Monitoring transplanted adipose tissue-derived stem 601–605. doi:10.1016/j.bbrc.2015.10.157.
cells combined with heparin in the liver by fluorescence imaging Zuk, P. 2013. Adipose-derived stem cells in tissue regeneration: a review.
using quantum dots. Biomaterials, 33(7): 2177–2186. doi:10.1016/j. Int. Sch. Res. Notices, 2013: e713959. doi:10.1155/2013/713959.
biomaterials.2011.12.009. Zuk, P.A., Zhu, M., Mizuno, H., Huang, J., Futrell, J.W., Katz, A.J.,
Zakrzewski, W., Dobrzyński, M., Szymonowicz, M., and Rybak, Z. 2019. et al. 2001. Multilineage cells from human adipose tissue: implica-
Stem cells: past, present, and future. Stem Cell Res. Ther., 10(1): 68. tions for cell-based therapies. Tissue Eng. 7(2): 211–228. doi:10.1089/
doi:10.1186/s13287-019-1165-5. 107632701300062859.
Zhang, C., Zhou, G., Chen, Y., Liu, S., Chen, F., Xie, L., et al. 2018. Human Zuk, P.A., Zhu, M., Ashjian, P., De Ugarte, D.A., Huang, J.I., Mizuno, H.,
umbilical cord mesenchymal stem cells alleviate interstitial fibrosis et al. 2002. Human adipose tissue is a source of multipotent stem
and cardiac dysfunction in a dilated cardiomyopathy rat model by cells. Mol. Biol. Cell, 13(12): 4279–4295. doi:10.1091/mbc.e02-02-0105.

Can. J. Physiol. Pharmacol. 102: 161–179 (2024) | dx.doi.org/10.1139/cjpp-2023-0202 179

You might also like