Abstract
Cardiac excitation-contraction coupling (ECC) is the orchestrated process of initial myocyte electrical excitation, which leads to calcium entry, intracellular trafficking, and subsequent sarcomere shortening and myofibrillar contraction. Neurohumoral β-adrenergic signaling is a well-established mediator of ECC; other signaling mechanisms, such as paracrine signaling, have also demonstrated significant impact on ECC but are less well understood. For example, resident heart endothelial cells are well-known physiologic paracrine modulators of cardiac myocyte ECC mainly via nitric oxide and endothelin-1. Moreover, recent studies have demonstrated other resident non-cardiomyocyte heart cells (e.g., physiologic fibroblasts and pathologic myofibroblasts), and even experimental cardiotherapeutic cells (e.g., mesenchymal stem cells), are also capable of altering cardiomyocyte ECC through paracrine mechanisms. In this review, we first focus on the paracrine-mediated effects of resident and therapeutic non-cardiomyocytes on cardiomyocyte hypertrophy, electrophysiology, and calcium handling, each of which can modulate ECC, and then discuss the current knowledge about key paracrine factors and their underlying mechanisms of action. Next, we provide a case example demonstrating the promise of tissue engineering approaches to study paracrine effects on tissue-level contractility. More specifically, we present new functional and molecular data on the effects of human adult cardiac fibroblast conditioned media on human engineered cardiac tissue contractility and ion channel gene expression that generally agrees with previous murine studies, but also suggests possible species-specific differences. By contrast, paracrine secretions by human dermal fibroblasts had no discernible effect on human engineered cardiac tissue contractile function and gene expression. Finally, we discuss systems biology approaches to help identify key stem cell paracrine mediators of ECC and their associated mechanistic pathways. Such integration of tissue engineering and systems biology methods shows promise to reveal novel insights into paracrine mediators of ECC and their underlying mechanisms of action, ultimately leading to improved cell-based therapies for patients with heart disease.
Keywords: Paracrine signaling, mesenchymal stem cells, EC-coupling, tissue engineering, systems biology
Subject Terms: Electrophysiology, Calcium Cycling/Excitation-Contraction Coupling, Cell Therapy, Computational Biology, Contractile Function
INTRODUCTION
Cardiomyocyte excitation-contraction coupling (ECC) is the crucial process that links calcium (Ca2+) trafficking to active force generation and subsequent relaxation of the cardiomyocyte. Following supra-threshold myocyte excitation, Ca2+ rapidly influxes into the myocyte cytosol through depolarization-activated inward Ca2+ channels. This Ca2+ influx induces Ca2+ release from the sarcoplasmic reticulum; together, these mechanisms raise free cytosolic Ca2+ concentration, increasing the probability of Ca2+-troponin C binding and thus activating the contractile machinery.1 In normal physiology, a fine-tuned percentage of cytosolic Ca2+ is then pumped back into the sarcoplasmic reticulum reservoir for future calcium induced calcium release.1 A simplified overview of this highly orchestrated ECC process is shown in Figure 1.
Figure 1. β-Adrenergic Signaling and Cardiomyocyte ECC.
β–adrenergic receptor (β–AR) activation by binding of epinephrine/norepinephrine leads to the following signaling cascade: 1) activation of the guanosine triphosphate (GTP)-binding protein α; 2) stimulation of adenylyl cyclase (AC); and 3) a rise in cyclic adenosine monophosphate (cAMP). Increase in intracellular cAMP leads to protein kinase A (PKA) activation, which subsequently phosphorylates and thus increases ion flux through the L-type Ca2+ channel (LTCC) and ryanodine receptor (RyR). Additionally, PKA relieves phospholamban (PLB) inhibition on sarcoendoplasmic reticulum Ca2+-ATPase (SERCA), allowing for an increase in Ca2+ storage within the sarcoplasmic reticulum (SR). In normal physiology, SERCA removes approximately 70% of the activator cytosolic calcium (Ca2+) during diastole (inset). Lastly, PKA phosphorylates troponin I, leading to positive lusitropic effects via myofilament Ca2+ desensitization. Blue and red arrows denote calcium flow during systole and diastole, respectively.
Sympathetic β-adrenergic signaling is a well-established rapid inotropic2 and lusitropic3 modulator of ECC.1 Briefly, β-adrenergic receptor stimulation of Gs type G-protein coupled receptors activates adenylyl cyclase to form cyclic adenosine monophosphate. Cyclic adenosine monophosphate subsequently activates protein kinases that phosphorylate various proteins essential to electrophysiology, calcium handling, and thus ECC, including the L-type calcium channel,4,5 phospholamban,6 ryanodine receptor,7 troponin I,8 and myosin binding protein C9 (Figure 1). A comprehensive review of sympathetic activation effects on ECC is available elsewhere.1
Cardiomyocyte ECC and associated β-adrenergic signaling can be further modulated by factors secreted by both distant and nearby cells. For example, chemokine CXCL12, also known as stromal cell-derived factor, can activate its receptor CXCR4 to attenuate β-adrenergic-mediated increases in adult cardiomyocyte calcium handling and fractional shortening.10 In addition, it is well established that cardiac endothelial cells secrete certain paracrine factors (e.g., nitric oxide) that can act as physiological signaling mediators of myocyte ECC and compete with β-adrenergic signaling pathways.11,12
Recent work has demonstrated that several other neighboring physiological and pathological non-cardiomyocytes in the heart, as well as experimental cardiotherapeutic cells, are capable of modulating cardiomyocyte ECC by paracrine signaling mechanisms. Importantly, these effects are relatively slower than previously described β-adrenergic signaling, likely reflecting underlying cardiomyocyte remodeling. Note that while cardiomyocytes are also known to release cardioactive paracrine factors,13–15 their mechanisms of action are largely independent of direct ECC-modulation and are thus beyond the scope of this review; for more details, see elsewhere.16
In this review, we will first discuss both established and unresolved key ECC-related paracrine factors secreted by endothelial cells, fibroblasts, myofibroblasts, cardiac progenitor cells (CPCs), and mesenchymal stem cells (MSCs), together representing neighboring physiologic, pathologic, and therapeutic paracrine mediators of cardiomyocyte ECC (for a summary of neighboring non-cardiomyocyte paracrine effects on cardiomyocyte ECC, see Table 1). Next, we provide a case example to demonstrate the promise of tissue engineering approaches to study non-cardiomyocyte paracrine effects on contractile function and associated gene expression. Finally, we discuss systems biology approaches to help identify key cardiomyocyte ECC paracrine-mediators of cardiotherapeutic cells, the least studied cell types in this context.
Table 1.
Brief Summary of Non-Cardiomyocyte Paracrine Effects on Cardiomyocyte ECC
| Source | Likely Paracrine Mediator(s) | Paracrine-Mediated Effect on ECC | Potential Mechanisms of Action |
|---|---|---|---|
| Endothelial Cell | Nitric Oxide | ↑ basal myocardial contractility | LTCC17 and RyR2 nitrosylation.11,18 |
| Attenuate β–adrenergic inotropic effects | cGMP-dependent degradation of cAMP via PDEII;19,20 PKG-mediated decrease of LTCC activity.19,21 | ||
| Positive lusitropy | cGMP-dependent PKG phosphorylation of troponin I.22–24 | ||
| Endothelin-1 | ↑ Ca2+ entry | PKC-mediated activation of NCX forward/reverse mode;25–30 PKC-mediated activation of LTCC.31–35 | |
| Alter myofilament Ca2+ sensitivity | PKC/D-dependent phosphorylation of troponin I and myosin-binding protein C.36–40 | ||
| ↑ calcium-induced calcium release | IP3-induced calcium release from the SR via IP3R, which subsequently sensitizes RyR2.41–43 | ||
| Fibroblast | TGF-β | Hypertrophy;44,45 ↑ resting membrane potential;46 ↓ conduction/upstroke velocity;46 ↑ calcium transient amplitude;47 ↑ contractility (see Figure 4 and elsewhere48) | Non-canonical, non-Smad TGF-β MEK/ERK signaling pathway;48 Na+ and K+ ion channel remodeling (see Figure 5 and elsewhere46) |
| Exosomal miRNA-21-3p | Hypertrophy49 | Targeting the mediators sorbin and SH3 domain-containing protein 2, as well as PDZ and LIM domain 549 | |
| Myofibroblast | TGF-β | Hypertrophy;47 ↓ calcium transient amplitude47 | TGF-β signaling47 |
| CPC | Unresolved | ↑ contractility50 | ↑ SERCA/NCX ratio50 |
| MSC | Unresolved | ↑ action potential duration;51 ↑ calcium transient amplitude;52,53 ↑ contractility;52,54 ↓ calcium transient alternans;55,56 predominantly anti-arrhythmic effects53,54,57,58 | PI3K/Akt signaling;52,54 increase in SERCA and LTCC activity/gene expression52,54 |
Non-standard abbreviations: L-type calcium channel (LTCC); ryanodine receptor (RyR2); cyclic guanosine monophosphate (cGMP); cyclic adenosine monophosphate (cAMP); phosphodiesterase II (PDEII); protein kinase (PK); sodium-calcium exchanger (NCX); inositol trisphosphate (IP3); sarcoplasmic reticulum (SR); inositol trisphosphate receptor (IP3R); transforming growth factor beta (TGF-β); sarco/endoplasmic reticulum Ca2+-ATPase (SERCA).
PARACRINE EFFECTS OF CARDIAC ENDOTHELIAL CELLS ON CARDIOMYOCYTE ECC
Cardiac endothelial cells, including but not limited to endocardial59 and intramyocardial capillary endothelial cells,60 regulate cardiac function and are well-established paracrine mediators of myocyte ECC; they release a vast array of cardioactive factors, including nitric oxide, endothelin-1, prostaglandins, natriuretic peptides, angiotensin II, kinins, reactive oxygen species, adenylpurines, neuregulin-1, parathyroid hormone-related peptide, and others.61–63 Two major ECC factors, nitric oxide and endothelin-1,63 are discussed below. A schematic summarizing these mediators and their effects on cardiomyocyte ECC is shown in Figure 2.
Figure 2. Endothelial-Cardiomyocyte Interplay Through Paracrine Factors.
Endothelial cell nitric oxide (NO) increases basal contractility via nitrosylation of L-type Ca2+ channel (LTCC) and ryanodine receptor (RyR2). NO attenuates β–adrenergic effects on cardiomyocyte ECC via cyclic guanosine monophosphate (cGMP)-dependent degradation of cyclic adenosine monophosphate (cAMP) and protein kinase G (PKG)-mediated decrease of LTCC activity. PKG also phosphorylates troponin I, leading to myofilament calcium desensitization and thus increased lusitropy. Endothelin-1, which mainly acts through the endothelin A (ETA) receptor in ventricular cardiomyocytes, may increase calcium entry via protein kinase C (PKC)-mediated: 1) increase of LTCC activity; 2) indirect activation of sodium-calcium exchanger (NCX) reverse mode by increasing Na+-H+ exchanger activity; and 3) direct activation of NCX reverse (shown) and/or forward (not shown) mode. Endothelin-1 alters myofilament Ca2+ sensitivity via protein kinase C/D (PKC/D) phosphorylation of troponin I and myosin-binding protein C. Finally, endothelin-1 may increase calcium-induced calcium release via inositol trisphosphate (IP3) activation of inositol trisphosphate receptor (IP3R), which sensitizes RyR2 on the sarcoplasmic reticulum (SR). Green and red arrows denote activation and inhibition, respectively. Other non-standard abbreviations: phospholamban (PLB); sarcoendoplasmic reticulum Ca2+-ATPase (SERCA).
Endothelial Nitric Oxide
Nitric oxide is reported to have biphasic inotropic effects on basal contractility, where only low doses increase basal contractility.12,64–68 For example, various ex vivo studies on isolated perfused rat hearts65,69 and cardiac myocytes70 demonstrated low levels of nitric oxide modestly increase basal myocardial contractility.11 Mechanistically, these effects are likely cyclic guanosine monophosphate-independent, and instead predominantly via nitrosylation of the voltage-dependent L-type calcium channel17 and ryanodine receptor 2.11,18 Activation of these key calcium handling proteins can lead to increased Ca2+ entry and Ca2+-induced calcium release, respectively.
On the other hand, in the presence of β-adrenergic stimulation, nitric oxide attenuates cardiomyocyte ECC inotropic enhancement by: 1) cyclic guanosine monophosphate-dependent degradation of cyclic adenosine monophosphate via phosphodiesterase II, thus effectively reducing Ca2+ entry through the downstream voltage-dependent L-type calcium channel effector;19,20 and 2) protein kinase-G-mediated decrease of L-type calcium channel activity.19,21
Protein kinase-G has also been shown to phosphorylate troponin I via cyclic guanosine monophosphate-dependent mechanisms;22–24 this leads to myofilament calcium desensitization, which is associated with positive lusitropic effects.71
Endothelial Endothelin-1
Endothelin-1 is a peptide that is a potent vasoconstrictor.72 In addition to its vascular effects, endothelin-1 has demonstrated positive inotropic effects across various species61,73–75 via activation of endothelin receptors expressed on cardiomyocytes. Studies have shown the positive inotropic effect of endothelin-1 is predominantly attributed to enhanced Ca2+ entry, increased myofilament sensitivity to cytosolic Ca2+, and enhanced calcium-induced calcium release.76
Mechanistically, endothelin-1-mediated enhancement of Ca2+ entry results from: 1) activation of Na+/Ca2+ exchanger forward and/or reverse mode via protein kinase C downstream of the endothelin A receptor;25–28 and 2) indirect activation of the Na+/Ca2+ exchanger reverse mode by protein kinase C-dependent increase of Na+-H+ exchanger activity, effectively increasing intracellular Na+ and thus driving the Na+/Ca2+ exchanger to increase intracellular calcium.29,30 Other mechanisms, such as endothelin-1 activation of L-type calcium channel current remain controversial,31–34 but are also likely mediated via protein kinase C downstream of the endothelin receptor.35
The increased extrusion of H+ via the Na+-H+ exchanger was originally hypothesized to affect myofilament calcium sensitivity via cytosolic alkalinazation.76 However, this was later debunked, as the bicarbonate anion transporter largely compensates for changes in cytosolic H+ concentration.76 Instead, endothelin-1 likely alters myofilament sensitivity by protein kinase C/D-dependent phosphorylation of troponin I and myosin-binding protein C.36–40
Finally, endothelin-1 can stimulate inositol trisphosphate-induced calcium release from the sarcoplasmic reticulum of ventricular myocytes via inositol trisphosphate receptors, which can subsequently sensitize ryanodine receptors.41–43 However, these mechanisms are not considered large contributors to endothelin-1 inotropic effects on ventricular myocytes, which are known to have low abundance of inositol trisphosphate receptors.76,77
PARACRINE EFFECTS OF CARDIAC FIBROBLASTS ON MYOCYTE ECC
Conventionally, healthy cardiac tissue is believed to have a cellular composition that is up to two-thirds non-myocytes, the majority of which are cardiac fibroblasts.78–80 Interestingly, Pinto et al. recently demonstrated the number of fibroblasts might not be so high—potentially even less than myocytes—leaving this issue unresolved.81 Nevertheless, while under normal physiological conditions fibroblasts are key to maintaining homeostasis of myocardial structure and function, they are also activated in pathological wound healing leading to tissue remodeling and fibrosis. The maladaptive remodeling of fibrotic scar formations then forms obstacles at the tissue level that interfere with normal electrical propagation,82,83 and is considered pro-arrhythmic. Such pathophysiological responses have motivated genetic modification of unexcitable primary human fibroblasts to create engineered cells that can conduct action potentials and rescue conduction slowing in a cell culture model of fibrosis.84 In general, cardiac fibroblasts are capable of expressing gap junctions and coupling to cardiomyocytes (see elsewhere85,86 for a comprehensive review), and communicating via paracrine signaling.
As described below, physiologic fibroblasts and pathologic myofibroblasts have been found to alter myocyte size, electrophysiology, and calcium handling, thus influencing ECC at both cellular and tissue levels via paracrine signaling mechanisms.
Fibroblast-Derived Soluble Factors Can Mediate Cardiomyocyte Hypertrophy
In addition to their role in fibrosis, cardiac fibroblasts may influence the cardiac hypertrophic response. Indeed, a hypertrophic “nonmyocyte-derived growth factor” released by fibroblasts was hypothesized by Long et al. over 20 years ago.87 Since then, various soluble factors—such as angiotensin II, transforming growth factor beta (TGF-β), endothelin-1, insulin-like growth factor 1 (IGF-1), and interleukin 6—have been suggested as fibroblast paracrine mediators of hypertrophy.
Angiotensin II was one of the initial fibroblast paracrine factors hypothesized to induce hypertrophy in myocytes.88 Later work then demonstrated fibroblasts are required to be present for angiotensin II-dependent hypertrophic effects,45,89,90 motivating investigators to study downstream paracrine mediators released by angiotensin II stimulated-fibroblasts to induce myocyte hypertrophy.
For example, independent of its pro-fibrotic effects,91 TGF-β has been demonstrated to induce cardiac hypertrophy both in vitro and in vivo. Rosenkranz et al. demonstrated that over-expression of TGF-β in transgenic mice led to increased hypertrophy.92 Importantly, others showed stretched- or angiotensin II stimulated-fibroblast conditioned media neutralized of TGF-β circumvents the cultured myocyte hypertrophic responses,44,45 suggesting TGF-β is required for fibroblast paracrine-mediated effects on myocyte hypertrophy. Using a similar approach, fibroblast-released endothelin-1 has also been suggested to induce hypertrophy. Conditioned media from fibroblasts stimulated with cyclic stretching or angiotensin II did not induce hypertrophy in myocytes treated with endothelin receptor antagonists.44,89,93
Other paracrine factors have also been suggested to stimulate cardiac myocyte hypertrophy; neutralizing cardiotropin-1—a member of the interleukin 6 family—or IGF-1 also attenuated hypertrophic effects of fibroblast-conditioned media.93–95 Takeda et al. further demonstrated IGF-1 acts downstream of Küppel-like factor 5 to induce such hypertrophic effects on myocytes.96 Other fibroblast hypertrophic soluble mediators (e.g., fibroblast growth factor 297) have also been hypothesized, but are less studied. Taken together, the results suggest that beyond their role in fibrosis, cardiac fibroblasts secrete several soluble factors that may impact the hypertrophic response in the heart.
Fibroblast-Derived Insoluble Extracellular Vesicles Mediate Cardiomyocyte Hypertrophy
Recently, studies have shown that cell-cell communication can also take place via secreted exosomes, which are bilayer membrane-bound nanovesicles packaging cargoes of select lipids, proteins, and RNAs.98 In the context of cardiovascular disease, various resident heart cells and therapeutic stem cells have been shown to communicate with myocytes and myocardial tissue via exosomes, predominately involved in cardioprotective,99,100 pro-angiogenic,101,102 and anti-fibrotic100,101 effects.98 However, recently it was shown that exosomes are also potential mediators of hypertrophy. Bang et al. observed that cardiac fibroblasts release exosomal miRNA passenger strand miRNA-21-3p to induce cardiomyocyte hypertrophy in vitro. Mechanistically, they identified sorbin and SH3 domain-containing protein 2, as well as PDZ and LIM domain 5 as the main targets and mediators of miRNA-21-3p induced hypertrophy.49 Their findings translated in vivo, whereby microinjection of antagomiRNA-21-3p was found to attenuate angiotensin II-induced hypertrophy in mice.49 Thus, it is possible that beyond secreted proteins, cardiac fibroblast-derived exosomes may also be involved in the cardiac response to stress.
Fibroblast Paracrine Factor Effects on Electrophysiology
An established electrophysiological phenotype of cardiac hypertrophy is prolonged action potential duration (APD) via reduction of voltage-gated potassium channel currents.103,104 Thus, it is plausible that hypertrophy-inducing fibroblast conditioned media could also alter myocyte electrophysiology. Indeed, Guo et al. first demonstrated with neonatal rat cardiac myocytes in vitro that 72-hour treatment with fibroblast conditioned media not only induced hypertrophy, but also prolonged APD at 25% repolarization, and decreased transient outward current density.95 Nearly a decade later, electrophysiological remodeling effects of fibroblast-conditioned media were further elucidated. Pedrotty et al. demonstrated various consistent single-cell and tissue-level pathological, dose-dependent, electrophysiological effects of cardiac fibroblasts on neonatal rat cardiomyocytes, including: 1) decreased monolayer conduction velocity; 2) prolongation of APD; 3) increased resting membrane potential; and 4) decreased upstroke velocity.46 These effects were likely caused by ion channel remodeling, as inward rectifying potassium, sodium, and transient outward potassium channel genes were down-regulated while there were no significant changes in fibroblast proliferation, cardiomyocyte apoptosis, or connexin-43 function.46 Interestingly, these effects were not only dose-dependent but also microenvironment-dependent, as they were not reproduced when the treatment used conditioned media from cardiac myocyte-fibroblast co-cultures.46
Such observations of cardiac fibroblast conditioned media on the electrophysiological properties of cardiomyocytes are not limited to neonatal rat cardiac myocytes; indeed, they have been reported consistently across several myocyte sources. With adult rat cardiomyocytes, fibroblast conditioned media was shown to decrease peak transient outward current, lengthen APD, and decrease sodium channel current.105 Adult mouse cardiac fibroblast conditioned media was found to prolong APD to 50% repolarization, increase peak notch action potential voltage (a possible surrogate of transient outward current activity), and decrease beat rate of mouse embryonic stem cell-derived cardiomyocytes.106
The fibroblast-released factors mediating such electrical remodeling remain unresolved. Guo et al. demonstrated fibroblast-released IGF-1 may play a role in hypertrophy, but not ion channel remodeling. They observed that anti-IGF-1 antibody treated fibroblast conditioned media partially inhibited myocyte hypertrophy,95 while transient outward ion channel activity was unaffected relative to the fibroblast conditioned media group.95 While TGF-β has been shown to induce hypertrophy (as discussed above), it is also a valid candidate as a mediator of the electrophysiological effects of cardiac fibroblast conditioned media, as Kaur et al. showed that TGF-β was responsible for ion channel remodeling in adult rat cardiomyocytes.105 Such effects could potentially contribute to the electrical remodeling that typically accompanies myocardial injury. Additionally, Kaur et al.’s findings were consistent with other studies examining exogenous TGF-β, which has been found to: 1) reduce sodium, inward rectifier potassium, and outward sustained potassium currents in neonatal rat atrial cardiomyocytes;107 2) increase and decrease sodium and transient outward potassium currents, respectively, in adult rat cardiomyocytes;105 and 3) prolong APD in adult rat cardiomyocytes.105 One important area for future work involves testing the extension of these findings in large animal and human cardiomyocytes, as we begin to investigate below in Figures 3–5.
Figure 3. Human Engineered Cardiac Tissue Contractility Assay.
(A) hECTs are created, cultured, and tested in a custom bioreactor with integrated force-sensing end-posts; as the tissue beats, deflections of the end-posts are tracked. Output contractile metrics include, but are not limited to, developed force (DF), maximum rates of contraction and relaxation (+/− dF/dt, respectively), and beat rate. (B) Confocal microscopy of hECTs labeled with cardiac troponin I (green) and DAPI (blue) displays cardiomyocytes with striated sarcomeres and regions of aligned myofibrils. Inset shows magnified view of registered sarcomeres. (C) hECT labeled with sarcoendoplasmic reticulum Ca2+-ATPase 2 (red) and DAPI (blue) shows sarcoplasmic reticulum structures distributed throughout the tissue. Scale bar = 40 μm.
Figure 5. Molecular Characterization of hECTs.
hECTs from each group (n=3–4, mean±SEM) were snap-frozen for qRT-PCR on day 10, where expression of (A) cardiac-specific/calcium handling, (B) apoptotic, and (C) potassium/sodium channel genes were quantified. *p<0.05, **p<0.01; p-values from one-way ANOVA with Scheffe’s post-hoc test. Non-standard abbreviations: cardiac troponin T (cTnT); L-type calcium channel (LTCC); sarco/endoplasmic reticulum calcium-ATPase (SERCA2a); connexin-43 (Cx43); atrial natriuretic peptide (ANF); myosin heavy chain (MHC); caspase-3 (Casp3); caspase-9 (Casp9); B-cell lymphoma 2 (BCL2); BCL2-associated X protein (BAX); transient outward potassium current (Kv4.2); inward rectifier potassium current (Kir2.1); delayed rectifier potassium current (Kv11.1); sodium current (Nav1.5).
Fibroblast-Derived Paracrine Mediators of ECC and Contractility
Recently, fibroblast paracrine factors have also been shown to also alter calcium handling—a direct determinant of ECC—as well as cardiac tissue contractility. Motivated by the hypertrophy and electrophysiology studies described above, Cartledge et al. sought to further elucidate the role of physiological fibroblasts and pathological myofibroblasts on adult rat myocyte viability, size, and calcium transients.47 Using Transwells® to prevent direct cell-cell contact, they showed that physiological cardiac fibroblasts and pathological cardiac myofibroblasts both secrete paracrine factors that increase myocyte size, accelerate calcium transient decay, and decrease myocyte viability. However, effects on myocyte calcium transients were fibroblast phenotype-dependent: physiological fibroblast paracrine factors increased the myocyte calcium transient amplitude, while pathological myofibroblast paracrine factors decreased the calcium transient amplitude.47 Interestingly, Cartledge et al. observed TGF-β levels increased in Transwells® containing myocytes with either fibroblasts or myofibroblasts relative to myocyte-only cultures.47
To test the role of TGF-beta in fibroblast paracrine-mediated effects on myocyte calcium handling, viability, and hypertrophy, they used SB431542 to block TGF-β type 1 receptors. Blockage of the TGF-β type 1 receptor prevented the previously described effects on cardiomyocyte viability, size, and calcium transient amplitude.47
Liau et al. recently extended this work to investigate developmental stage-dependent effects of cardiac fibroblast conditioned media on contraction. Specifically, mouse embryonic stem cell derived-cardiomyocyte miniature tissue “micro-patches” were supplemented with conditioned media from adult or fetal cardiac fibroblasts.48 Adult cardiac fibroblast conditioned media significantly increased contraction of the mouse micro-patches; these effects were amplified with fetal cardiac fibroblast-conditioned media.48 Using pathway inhibitor experiments and Western blot analysis, they concluded these effects to be mediated via MEK-ERK signaling,48 which is a non-Smad pathway in TGF-β signaling.108 Altogether, these findings suggest TGF-β is a likely mediator of cardiac fibroblast paracrine-mediated effects on contraction via ion channel remodeling and hypertrophy.
STEM CELL SECRETOMES CAN IMPACT CARDIOMYOCYTE ECC
Resident adult cardiac stem cells—which include CPCs, endothelial progenitor cells, and MSCs109—also have the potential to increase cardiomyocyte ECC through paracrine mechanisms. For example, Tang et al. demonstrated cell-mimicking synthetic microparticles carrying the human cardiac stem cell secretome promoted neonatal rat cardiomyocyte contractility in vitro.110
Delivery of CPCs101,111–113 and bone marrow-derived MSCs58,114–116 are emerging approaches to treat heart disease. Like several other stem cell sources (e.g., embryonic and induced pluripotent stem cells, CD34+ stem cells, etc.), CPC and MSC paracrine factors have been shown to enhance angiogenesis,101,112,117–119 decrease fibrosis,101,112,119,120 and promote other cardiac repair mechanisms (comprehensive reviews on this topic are available elsewhere121,122), which can indirectly modulate cardiomyocyte contractility. Moreover, CPC and MSC paracrine factors have each shown the potential to more directly alter myocyte electrophysiology and calcium handling, and thus ECC.
Kinetics of Stem Cell Paracrine Effects on ECC
As depicted in Table 1, CPC and MSC paracrine effects on cardiomyocyte ECC are primarily associated with increasing expression of calcium handling genes. In comparison to nearly instantaneous β-adrenergic signaling mechanisms,123 paracrine mechanisms are expected to be slower and longer-lasting; indeed, Desantiago et al. demonstrated MSC paracrine effects on ECC take at least 5 minutes in isolated ventricular myocytes,52 and in our recent work, such in vitro effects of MSCs were evident at least five days post-treatment.54 Clearly, a better understanding of the kinetics of stem cell paracrine effects is warranted in order to optimize future stem cell cardiotherapies for both ischemic and non-ischemic cardiomyopathies. To do so, further insight is required into CPC and MSC paracrine effects on ECC; current knowledge on each of these respective fields is discussed below.
CPC Paracrine Effects on Cardiomyocyte ECC
Few studies have investigated CPC paracrine effects on cardiomyocyte ECC. Maxeiner et al. observed that human and rat CPC-conditioned media increased fractional shortening in isolated rat adult cardiomyocytes in a concentration-dependent manner relative to pure media control treatment, largely independent of CPC donor age.50 In addition, conditioned media derived from several human adult and juvenile CPC lines led to an increase in the ratio of sarcoendoplasmic reticulum Ca2+-ATPase (SERCA)/sodium-calcium exchanger gene expression.50 Interestingly, increases in contractile function were most prominent when the conditioned media was highly diluted,50 with further investigation required to understand these phenomena.
Several preliminary attempts have been made to identify which CPC-secreted factors may impact myocyte ECC. For example, in the same study, Maxeiner et al. performed cytokine arrays on released soluble factors by rat and human CPCs;50 the interleukin family, vascular endothelial growth factor, and tissue inhibitors of metalloproteinases-1 were consistently up-regulated in both juvenile and adult rat and human CPCs.50 Considering other components of the CPC secretome, a series of studies by the Davis group observed that the insoluble exosomal fraction of CPC-conditioned media led to no significant effect on rat ventricular myocyte calcium transient amplitude and sarcomere shortening at 12 hours post-treatment.101,112 It remains undetermined whether there may be a time- or species-dependence to these observations. Clearly, further investigation is necessary to better understand mechanisms and cargo involved in CPC paracrine-mediated effects on cardiomyocyte ECC. As discussed below, paracrine effects of MSCs on ECC have been more extensively studied.
MSC Paracrine Effects on Electrophysiology
The electrophysiological effects of MSC paracrine factors appear to vary across species and experimental conditions. Mureli et al. first showed mouse MSC conditioned media has a positive dromotropic effect on healthy atrial-like murine HL-1 monolayers by increasing connexin-43 mRNA expression and protein level.124 Hwang et al. found increased total connexin-43 protein levels significantly increased in hypoxic rat myocytes exposed to hypoxic human MSC (hMSC) conditioned media (exposure to normoxic hMSC conditioned media trended toward increased levels, with no statistical significance).53 However, these findings were not consistent with healthy ventricular myocytes using hMSC conditioned media,51 suggesting the effects of hMSC conditioned media may depend on cell phenotype and environment. Askar et al. showed with Transwell® experiments that hMSC conditioned media prolonged healthy neonatal rat cardiomyocyte APD in a dose-dependent manner, but did not affect conduction velocity in myocyte monolayers.51 Comparable to the 5–10% hMSC supplementation (percent per number of cardiomyocytes and fibroblasts in engineered cardiac tissue) used in our recent experiments,54 Askar et al. observed that dosages of 7% hMSC conditioned media (percent per number of myocytes in 2-D) led to significant effects on cardiomyocyte electrophysiology.51 Dose-dependent prolongation of APD led to increased APD dispersion and vulnerability of re-entry in the setting of healthy neonatal rat cardiomyocyte monolayers.51 On the other hand, in hydrogen peroxide treated human induced pluripotent stem cell derived-cardiac myocyte monolayers, hMSC conditioned media significantly decreased action potential alternans relative to control.55
In a recent study,54 in silico findings from mathematical models provide novel insights that help resolve disparate reports of potential pro-arrhythmic risks of hMSCs in vitro125 that appear contradictory to in vivo reports of hMSCs having no effect,57 or favorable cardioprotective effects,58 on arrhythmogenesis in pre-clinical animal studies and clinical trials. In simulations representing healthy myocyte monolayers (i.e., similar to experimental conditions from Askar et al.51), both hMSC paracrine signaling and heterocellular coupling have the potential to increase arrhythmogenicity (Online Figure IA; see Mayourian et al. for detailed methods54). However, in vulnerable window simulations of fibrotic cardiac tissue (i.e., similar to pre-clinical/clinical conditions for hMSC intervention), hMSC paracrine signaling-only conditions were anti-arrhythmic compared to control (Online Figure IB),54 consistent with a preclinical study in a rat myocardial infarction model, where hMSC paracrine factors were anti-arrhythmic, suppressed fibrosis, and restored conduction.53 Most importantly, in silico vulnerable window analyses predicted that hMSC supplementation (involving both hMSC paracrine signaling and heterocellular coupling mechanisms) did not adversely impact fibrotic cardiac tissue arrhythmogenesis, and may even be anti-arrhythmogenic (Online Figure IB).54 These simulations may help explain why hMSCs are mainly reported to be safe57 or even anti-arrhythmic58 in the clinical trial setting where paracrine effects can be present despite low cell engraftment efficiency.
While our simulations may suggest maximizing hMSC engraftment increases arrhythmogenic risk, it is important to note that several in vitro studies demonstrate that hMSCs have the potential to repair conduction block in neonatal rat cardiomyocyte cultures by filling conduction gaps via connexin-mediated heterocellular coupling.126,127 To our knowledge, this experimental preparation has yet to be simulated; the potentially important implications in post-myocardial infarction tissue warrant further investigation.
MSC Paracrine Effects on Calcium Handling
The effects of MSC paracrine factors on calcium handling are observed across several studies that look at healthy and injured myocytes. Desantiago et al. first showed that hMSC conditioned Tyrode-treated healthy mouse cardiomyocytes had increased calcium transient amplitude and accelerated calcium transient decay compared to control.52 Beneficial effects of hMSC paracrine factors on calcium handling were found in injured myocytes,53 where both normoxic and hypoxic hMSC conditioned media suppressed effects of hypoxia on neonatal rat ventricular myocyte cytosolic free calcium levels.53 Sattayaprasert et al. further showed that hMSC conditioned media reduces calcium transient alternans and shortens calcium transient duration in hydrogen peroxide treated human induced pluripotent stem cell derived-cardiac myocyte monolayers.55 Similarly, mouse MSC conditioned media attenuated the occurrence of arrhythmic calcium transients of ischemia/reperfusion injured isolated ventricular myocytes.56 Thus, across species, hMSC conditioned media appears to have favorable effects on calcium handling in cultured cardiomyocytes.
MSC Paracrine-Mediated Effects of Cardiac Contractile Function
In our most recent work, we demonstrated that the effects of hMSC paracrine factors on calcium handling translated to increased contractility in both computer model simulations and our custom human engineered cardiac tissue (hECT) system.54 We simulated ECC of human induced pluripotent stem cell-derived cardiomyocytes—representative of hECT cellular constituents—subjected to experimentally calibrated hMSC heterocellular coupling128 and paracrine signaling effects.54 We then compared the in silico results to hECT contractility measurements under matched hMSC treatments. The in silico and hECT data together corroborate the paracrine-mediated effects of hMSCs on cardiomyocyte contractility.54 Interestingly, when hMSCs were co-cultured within hECTs, the hMSC paracrine-mediated increase on contractility was attenuated;54 our simulations predicted underlying myocyte-hMSC heterocellular coupling may act as a “sink” and reduce calcium transient amplitude and thus decrease contraction.54 A similar phenomenon was recently reported by the Parker group, as neonatal cardiomyocytes coupled with stem cell-derived cardiomyocytes led to calcium transients and fractional shortening intermediate between the two cell types.129 Clearly, further investigation is needed to better understand direct cell-cell contact effects on cardiomyocyte ECC.
The previously described findings of MSC paracrine effects on contractility were consistent with a pair of studies by DeSantiago et al., where MSC conditioned Tyrodes’ solution increased cell shortening in healthy and injured mouse myocytes.52,56 Furthermore, they complement the findings by Luo et al., where hMSC secreted factors packaged into poly(lactic-co-glycolic acid) microparticles coated with the hMSC membrane promoted neonatal rat cardiomyocyte contraction in vitro.130 These positive inotropic effects of the hMSC secretome motivate efforts to: 1) investigate the underlying mechanisms; and 2) identify the responsible cardioactive factors.
Underlying Mechanisms of MSC Paracrine Effects on ECC
Substantial progress has been made toward elucidating the underlying mechanisms responsible for MSC paracrine effects on electrophysiology and calcium handling. Hwang et al. observed that hypoxic rat myocytes treated with normoxic hMSC conditioned media trended towards increased mRNA levels of L-type calcium channel and SERCA2a relative to control, with statistical significance achieved when hMSCs were cultured under hypoxic conditions.53 We recently demonstrated that treating hECTs with hMSC conditioned media also led to increased mRNA levels of L-type calcium channel and SERCA2a relative to untreated control;54 additionally, Desantiago et al. found that the activity of L-type calcium channel and SERCA2a activity increases in healthy mouse myocytes exposed to hMSC paracrine factors relative to control.52
Desantiago et al. were one of the first groups to find that the PI3K/Akt signaling pathway was activated in mouse ventricular myocytes by MSC conditioned media.52 Through proteomic analysis, we also recently also showed that PI3K/Akt signaling is predicted to be activated in hECT/hMSC conditioned media.54 This signaling pathway was also implicated in MSC paracrine-mediated cardioprotection from ischemia/reperfusion injury in vitro, where hMSC conditioned Tyrode’s solution attenuated the hyperpolarization of the mitochondrial membrane potential via PI3K/Akt signaling56, prolonging cell survival and reducing arrhythmic early after depolarizations. The role of the PI3K/Akt signaling pathway in hMSC conditioned media-mediated ECC enhancement was further defined in human induced pluripotent stem cell-derived cardiomyocyte calcium handling by Sattayaprasert et al.,55 where inhibitors of PI3K and endothelial nitric oxide synthase (a downstream mediator of PI3K) attenuated the benefits of hMSC paracrine media on calcium transient alternans and duration, whereas S-nitrosoglutathione, an nitric oxide donor, had opposite effects. Taken together, these findings confirm that the PI3K/AKT pathway is a critical mediator of the effects of hMSC paracrine factors on cardiomyocyte ECC.
Proposed MSC Factors Responsible for Enhancing ECC
Numerous studies have investigated the composition of the hMSC secretome;120,131–136 however, while many secreted factors have been associated with pro-angiogenic120,137–139 and anti-fibrotic120,139–141 effects, little is known regarding the hMSC paracrine factors responsible for the effects on electrophysiology, calcium handling, and contractility described above.
Cardioactive hMSC exosomes have been shown to modulate myocyte PI3K/Akt signaling,100,142 a crucial pathway involved in hMSC paracrine effects on cardiomyocyte ECC (Table 1). Indeed, we found that treating hECTs with the exosome-enriched fraction of the hMSC secretome recapitulated the effects observed with complete hMSC conditioned media on hECT contractile force and expression of calcium handling genes (e.g., SERCA2a and L-type calcium channel), whereas treatment with the exosome-depleted fraction was similar to unsupplemented controls.54 These findings motivate further investigation into the role of hMSC exosomes, their membrane (while certain exosomal membranes appear necessary for pro-angiogenic and anti-fibrotic cardioactivity,101 in general little is known about their role on cardiomyocyte ECC), and their cargo responsible for increased cardiomyocyte contractility.
hMSC exosomal cargo has been implicated in multiple studies related to cardiac repair;98,100,143–145 however, little is known about the specific effects of hMSC exosomes and their cargo on contractility. Based on previous literature, exosomal miRNA-21-5p may be one possible candidate responsible for increasing ECC in cardiomyocytes. miRNA-21-5p is the only miRNA consistently in the top 5 most abundantly found in hMSC exosomes across three independent studies.131,135,146 Interestingly, cardiac progenitor cell exosomal miRNA-21-5p has been shown to prevent cardiomyocyte apoptosis by targeting programmed cell death 4, known to be downstream of the PI3K/Akt signaling pathway.147 This is consistent with the findings by Wei et al.148 and Cheng et al.,149 where miRNA-21-5p also regulated cardiomyocyte apoptosis by targeting programmed cell death 4. miRNA-21-5p has also been shown to bind strongly to phospholamban150 and to activate endothelial nitric oxide synthase,151 both of which increase SERCA2a activity.52,150 Clearly, further work is necessary to elucidate the cardiotropic role of miRNA-21-5p and other key constituents of the hMSC secretome.
A major challenge in identifying key CPC and hMSC paracrine factors is that the composition of their secretomes strongly depends on the microenvironment. For example, the Davis group showed the cardioactivity of CPC exosomes increases under hypoxia.101,152 Similarly, Hwang et al. showed that hypoxic conditioning of hMSCs leads to more potent paracrine effects on calcium handling enhancement, fibrosis reduction, and cardioprotection.53 Kinnaird et al. demonstrated that 72 hours of hypoxic preconditioning of hMSCs led to a significant increase in the secretion of pro-angiogenic factors relative to normoxic control conditions.153 Furthermore, genetic manipulation of hMSCs154 or molecular preconditioning of hMSCs155 can also increase the potency of their paracrine effects. A systematic investigation utilizing a reliable contractility assay is warranted to optimize these environmental variables and to help identify key cardioactive hMSC paracrine factors.
TISSUE ENGINEERING APPROACHES TO STUDY NON-CARDIOMYOCYTE PARACRINE EFFECTS ON CARDIAC CONTRACTION
Human Engineered Cardiac Tissues as an in vitro Contractility Assay
A major advancement in the cardiac research field has been the growing sophistication of in vitro models of functional human myocardium by combining tissue engineering technology with human stem cell biology,156–158 as recently reviewed.159–161 In our laboratory, hECTs are created, cultured, and tested using a custom bioreactor162 with integrated force-sensing end-posts. As the tissue beats (either spontaneously or by electrical pacing), deflections of the end-posts are tracked in real time by high-speed video to non-invasively monitor contractile performance throughout a sequence of twitch cycles (Figure 3A). From the twitch tracings, extracted contractile metrics include, but are not limited to, developed force (DF), maximum rates of contraction and relaxation, and beat rate (Figure 3A, bottom).
These hECTs have been designed to ensure cell viability throughout the full tissue thickness,163,164 with cardiomyocytes that exhibit regions of aligned myofibrils and registered sarcomeres (Figure 3B). With SERCA2 found throughout the tissue (Figure 3C), hECTs recapitulate key aspects of native cardiac muscle physiology including ECC, the Frank-Starling mechanism, and pharmacologic responses.163 Such hECTs can be maintained for 4 weeks or longer, and are particularly suited for longitudinal in vitro investigation of healthy and diseased human cardiac contractile function and effects of therapeutic intervention. To this end, in the cardiac tissue engineering field, we are one of the first groups to model familial dilated cardiomyopathy165 and to test stem cell therapies.54,162,164 In the context of this review, our hECT system is advantageous for uniquely providing a configuration of co-cultured tissues in a shared bath in order to investigate paracrine signaling effects on cardiac contractility. Its ability to non-invasively measure contractile function facilitates the collection of multi-day longitudinal data on the effects of paracrine factors on contractile function. Altogether, hECTs offer a system that helps bridge a gap in previous experimental models of the heart, and provides new opportunities for advancing our understanding of stem cell paracrine signaling and exosome biology.
Indeed, our recent work on hMSC paracrine-mediated effects on hECT contractile function and gene expression recapitulated many findings from murine studies using alternative experimental setups,54 but also reveal some interesting species-specific and/or platform-specific differences. To further demonstrate the use of hECTs for testing paracrine-mediated effects on contractility, we provide new data examining how the effects of the fibroblast secretome on cardiomyocyte contractility and ECC-relevant gene expression translates to human cell sources (for detailed methods, see the Online Supplement). Following baseline contractile function testing on day 5 using our custom 3-D hECT contractility assay system (Figure 3A), we treated hECTs with either: 1) serum-free defined media control; 2) fresh human foreskin fibroblast conditioned media (HFF CdM); or 3) fresh human adult cardiac fibroblast conditioned media (hACF CdM). hECTs were cultured an additional 5 days in their respective treatments, after which contractile function was measured and compared to baseline measurements. Spontaneous beat rate and DF were also measured on a daily basis, providing a simple method of monitoring longitudinal contractile function without having to replace culture media (as is normally done following electrical pacing to eliminate electrolysis byproducts).
As shown in Figure 4A, the hACF CdM treatment led to a statistically significant increase in DF relative to pre-treatment at 0.5 Hz pacing, whereas the HFF CdM and untreated control groups were not significantly different from pre-treatment. Furthermore, hACF CdM led to a statistically significant increase in hECT DF relative to HFF CdM at 0.5 Hz pacing five days post-treatment (Figure 4A).
Figure 4. Effects of hACF Conditioned Media on hECT Contractile Function.

(A) Contractility assay shows hECT developed force during 0.5-Hz pacing (mean±SEM, n=4–6) at pre-treatment (day 5) and 5-days post-treatment with serum-free defined media (SFDM, control), hACF CdM, and HFF CdM. Daily measurements of (B) developed force and (C) beat rate during spontaneous contractions for each group (mean±SEM, n=3–4). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001, comparison of time point relative to pre-treatment in same group; #p<0.05, comparison of given group relative to HFF CdM at same time point; †p<0.05, ††p<0.01, comparison of same group at two different time points. p-values from repeated measures obtained using ANOVA followed by Bonferroni’s multiple comparisons test.
To understand the time-dependence of treatments, we looked at spontaneous beat rate and DF. As shown in Figure 4B, hACF CdM treatment led to statistically significant increases in DF days 2–5 post-treatment relative to day 0 pre-treatment, even though beat rate was not significantly different over time in this group (Figure 4C). In the HFF CdM and control groups, beat rate and DF tended to be relatively stable over time (the control group showed a significant decrease in beat rate on day 5 post-treatment, which coincided with a small but significant decrease in DF at this time point). These results complement murine studies on cardiac fibroblast paracrine-mediated contractility effects, as previously described.47,48
These hECT functional measures were corroborated by molecular characterization. Following five days of treatment and testing as above, hECTs from each group were snap-frozen for prospective qRT-PCR of cardiac-specific, calcium handling, apoptosis, and potassium/sodium ion channel (i.e., repolarization/depolarization, respectively) genes. In general, calcium handling and cardiac-specific genes were not significantly different between groups (Figure 5A). Furthermore, apoptosis genes were not significantly different between groups (Figure 5B). Interestingly, this hACF CdM-mediated gene profile is distinctly different from that of hMSC-treated hECTs (noted above and elsewhere54) despite similarities in the increase of hECT contractile function by the two cell types. Like others have suggested,46 this implies the effects of hACF CdM must be mediated through alternative mechanisms.
In particular, expression of the Kir2.1 inward rectifier and Kv11.1 delayed rectifier potassium channel genes significantly decrease in the hACF CdM intervention group compared to both HFF CdM and control groups (Figure 5C). These findings are also consistent with others.46 Surprisingly, there was a statistically significant increase in Kv4.2 transient outward potassium channel gene expression in the hACF CdM intervention group compared to both HFF CdM and control groups (opposite of previous reports in rodent cells46), while there is no significant difference in the Nav1.5 sodium channel gene between groups (which was found to decrease in rodent studies46). This may partly reflect species-dependent variations in ion channel distributions; for example, Kv4.2 is highly expressed in rat myocytes, but not in human myocytes.166 Accordingly, out of all genes analyzed, Kv4.2 had the highest threshold cycle (i.e., lowest expression) in control hECTs. Furthermore, transient outward activity plays a much less influential role in the human stem cell-derived myocyte action potential waveform relative to murine action potential (see multi-species sensitivity analysis in our previous work54 for justification). This suggests the increase in Kv4.2 is likely not physiologically significant, even though it is statistically significant. Altogether, these studies further support the use of hECT functional and molecular characterization for testing non-cardiomyocyte paracrine effects on cardiac ECC.
Based on these findings and the previous studies described, we hypothesize that reduction of outward potassium currents by hACF CdM leads to prolongation of the APD and, therefore, increased calcium handling with a concomitant increase in contractile force. The proposed mechanism is similar to the described inotropic effects of class III antiarrhythmic drugs.167–169 APD is an established correlate of contractile force;170–172 therefore, by effectively prolonging the plateau period, calcium availability is increased, augmenting calcium induced calcium release, elevating the calcium transient, and increasing ECC.
Limitations of Human Engineered Cardiac Tissues
Despite the advantages of hECT technology, one recognized limitation is the relatively immature cardiac phenotype that mimics newborn human heart muscle,163 reflecting an ongoing scientific challenge with cardiomyocytes derived from pluripotent stem cells.173 While perhaps not ideal for cardiac repair applications,174 immaturity may be less problematic for in vitro screening of factors that can modulate contraction given that aspects of the fetal gene program are reactivated as a stress-protective mechanism in failing adult myocardium.175 Also, hMSC therapy has recently been proposed to treat pediatric dilated cardiomyopathy.176 Thus, immature hECTs may adequately represent some aspects of the heart failure disease state and, as such, offer a unique contractility assay for studying cell-based treatment mechanisms and developing future cardiac therapies.
Nevertheless, like others in the field, we continue to develop strategies to promote phenotypic maturation of human pluripotent stem cell derived cardiomyocytes and hECTs including improved electrophysiology and contractility,177 structurally advanced sarcomeres and evidence of T-tubule formation,178 and the ability to model various forms of hypertrophic and dilated cardiomyopathies.165,179 Generating physiologic levels of force may be a prerequisite for surgical implantation applications of engineered cardiac tissues;158 however, for in vitro studies, the current sub-physiologic but cardio-mimetic performance of our hECTs appears suitable for therapeutic screening applications.
SYSTEMS BIOLOGY APPROACHES TO STUDY STEM CELL PARACRINE EFFECTS ON ECC
As previously reviewed,120,134 bioinformatics methods have proven valuable for investigating paracrine mechanisms of stem cell therapy, such as angiogenesis, fibrosis, and immune modulation. However, in the specific context of paracrine effects of stem cells on cardiac ECC, the application of computational and systems biology approaches is a relatively nascent field. As discussed below, recent systems biology approaches have shown promise for: 1) complementing experimental data to better understand non-cardiomyocyte paracrine effects on contractile function; and 2) identifying key paracrine mediators responsible for increases in contractility.
In our previous work,54 complementary experimental and computational methods were used to study hMSC paracrine-mediated effects on contractility and arrhythmogenicity. The mathematical models could simulate how paracrine signaling affects cardiac cell and tissue physiology (e.g., action potentials and calcium transients) based on calibrating the models with measured effects on cellular physiology and tissue-level fibrosis. One benefit of this approach is that the relative importance of specific effects can be independently controlled and readily delineated in the simulations, which is often challenging in an experimental setting. A second benefit is that because the model is calibrated using data covering a range of hMSC concentrations, the effects of intermediate concentrations can also be simulated. Our model was successful in reproducing paracrine-mediated effects on hECT contraction that were not included in the model calibration process, demonstrating the predictive power of the model. Together, these two approaches, which provide complementary information, can yield greater insights than either strategy used in isolation.
Nevertheless, such phenomenologic simulations cannot be used to infer which components of the secretome are crucial in altering myocyte physiology. Recent advancements in systems biology have enabled linking complex networks of cues (e.g., hMSC microenvironment), signals (e.g., hMSC factors), and responses (e.g., phenotypes of interest). Janes et al. first demonstrated that partial least squares regression (PLSR) analysis is useful for forming relationships between signaling molecules and cell decisions.180 The PLSR method has been applied to a range of bioanalytics problems;181–184 recently, the Davis group used PLSR in a pair of studies to form relationships between CPC exosomal miRNA effectors and pro-angiogenic, anti-fibrotic, and anti-hypertrophic responses.101,152 This method was successful in predicting various key exosomal miRNAs, though it has yet to be applied in the context of cardiomyocyte ECC.
INTEGRATING TISSUE ENGINEERING AND SYSTEMS BIOLOGY APPROACHES TO IDENTIFY NOVEL CARDIOACTIVE FACTORS
Ranganath et al. first proposed a systematic approach to optimize and harness the hMSC secretion profile by conditioning hMSCs with an array of small molecules, and subsequently characterizing the customized secretomes via high-throughput human cytokine antibody array.120 In the context of identifying key components of the hMSC secretome responsible for increasing contractility, this method could address the issue of hMSC secretome dependence on microenvironment/preconditioning, but it would not reveal: 1) the key factors responsible for a given contractile phenotype; nor 2) the role of insoluble factors, including exosomes and their cargo, which have been demonstrated to influence contractile function.54
We propose utilizing the PLSR method to help identify key hMSC factors responsible for increasing contractility. By collecting sets of quantitative measurements of “input” signaling molecules and “output” metrics of interest across an array of culturing conditions, PLSR can be utilized to predict relationships between the cues, signals, and responses (details of this method are available elsewhere101,152). This method is an extension of that proposed by Ranganath et al.;120 in addition to signals and cues, it would also require responses of interest. Given our stated focus on ECC, we propose utilizing hECTs as an in vitro contractility assay to generate physiologically relevant response data for the PLSR analysis. By testing the contractile function of hECTs supplemented with hMSC-conditioned media from an array of preconditioning environments, PLSR can be applied to systematically reveal relationships between factors identified via protein and miRNA profiling techniques, and functional contractile outputs of hECTs. Such methodology could equally be applied to identify key CPC-secreted ECC-modulating factors. Examples of conditioning environments for these cell types that may alter their secretome composition and potency include: normoxia versus hypoxia;101,112 two-dimensional versus three-dimensional substrates;54 soft versus hard stiffness;185,186 and homocellular versus heterocellular culture.54,187
CONCLUSIONS
Recent studies have demonstrated that in addition to classical endothelial cell paracrine modulation of ECC, other resident non-cardiomyocyte heart cells (e.g., physiologic fibroblasts and pathologic myofibroblasts), or even experimental cardiotherapeutic cells (e.g., MSCs and CPCs), are also capable of altering cardiac ECC through indirect paracrine mechanisms. Based on our recent work and the new experimental data herein, we demonstrate that hECTs are a promising contractility assay to further study non-cardiomyocyte paracrine effects on tissue-level contractility. Systems biology approaches can provide complementary information to identify the key stem cell paracrine mediators of ECC and their mechanisms of action. Altogether, integrating tissue engineering and systems biology approaches shows exciting potential for providing novel insight into paracrine mediators of ECC and their underlying mechanisms of action, ultimately leading to improved cell-based therapies for patients with heart disease.
Supplementary Material
Acknowledgments
This work was supported in part through the resources and staff expertise provided by the Scientific Computing at the Icahn School of Medicine at Mount Sinai. Microscopy and image analysis were performed at the Microscopy CoRE at the Icahn School of Medicine at Mount Sinai. The authors thank the Pluripotent Stem Cell Facility at the Icahn School of Medicine at Mount Sinai for providing the H7 human embryonic stem cell line and reagents.
SOURCES OF FUNDING
This work was supported by NIH/NHLBI 1F30HL134283-01A1 (JM), American Heart Association 15POST25090116 (DKC), NIH/NHLBI F30HL118923 (TJC), NIH HL124187 and American Heart Association 17GRNT33460554 (SS), NIH HL119046, HL129814, HL128072, HL131404, and HL135093 (RJH), and NIH/NHLBI R01HL132226 (KDC).
Nonstandard Abbreviations and Acronyms
- APD
Action potential duration
- Ca2+
Calcium
- CPC
Cardiac progenitor cell
- DF
Developed force
- ECC
Excitation-contraction coupling
- hACF CdM
Human adult cardiac fibroblast conditioned media
- hECT
Human engineered cardiac tissue
- HFF CdM
Human foreskin fibroblast conditioned media
- IGF-1
Insulin-like growth factor 1
- MSC
Mesenchymal stem cell
- PLSR
Partial least squares regression
- SERCA
Sarcoendoplasmic reticulum Ca2+-ATPase
- TGF-β
Transforming growth factor beta
Footnotes
DISCLOSURES
Dr. Costa discloses his role as scientific co-founder and Chief Scientific Officer of NovoHeart Ltd. NovoHeart did not play any role in the design or conduct of this study. The other authors report no conflicts.
References
- 1.Bers DM. Cardiac excitation-contraction coupling. Nature. 2002;415:198–205. doi: 10.1038/415198a. [DOI] [PubMed] [Google Scholar]
- 2.Lohse MJ, Engelhardt S, Eschenhagen T. What is the role of beta-adrenergic signaling in heart failure? Circ Res. 2003;93:896–906. doi: 10.1161/01.RES.0000102042.83024.CA. [DOI] [PubMed] [Google Scholar]
- 3.Kaumann A, Bartel S, Molenaar P, Sanders L, Burrell K, Vetter D, Hempel P, Karczewski P, Krause EG. Activation of beta2-adrenergic receptors hastens relaxation and mediates phosphorylation of phospholamban, troponin i, and c-protein in ventricular myocardium from patients with terminal heart failure. Circulation. 1999;99:65–72. doi: 10.1161/01.cir.99.1.65. [DOI] [PubMed] [Google Scholar]
- 4.Zhao XL, Gutierrez LM, Chang CF, Hosey MM. The alpha 1-subunit of skeletal muscle l-type ca channels is the key target for regulation by a-kinase and protein phosphatase-1c. Biochem Biophys Res Commun. 1994;198:166–173. doi: 10.1006/bbrc.1994.1024. [DOI] [PubMed] [Google Scholar]
- 5.Gerhardstein BL, Puri TS, Chien AJ, Hosey MM. Identification of the sites phosphorylated by cyclic amp-dependent protein kinase on the beta 2 subunit of l-type voltage-dependent calcium channels. Biochemistry. 1999;38:10361–10370. doi: 10.1021/bi990896o. [DOI] [PubMed] [Google Scholar]
- 6.Simmerman HK, Jones LR. Phospholamban: Protein structure, mechanism of action, and role in cardiac function. Physiol Rev. 1998;78:921–947. doi: 10.1152/physrev.1998.78.4.921. [DOI] [PubMed] [Google Scholar]
- 7.Marx SO, Reiken S, Hisamatsu Y, Jayaraman T, Burkhoff D, Rosemblit N, Marks AR. Pka phosphorylation dissociates fkbp12.6 from the calcium release channel (ryanodine receptor): Defective regulation in failing hearts. Cell. 2000;101:365–376. doi: 10.1016/s0092-8674(00)80847-8. [DOI] [PubMed] [Google Scholar]
- 8.Sulakhe PV, Vo XT. Regulation of phospholamban and troponin-i phosphorylation in the intact rat cardiomyocytes by adrenergic and cholinergic stimuli: Roles of cyclic nucleotides, calcium, protein kinases and phosphatases and depolarization. Mol Cell Biochem. 1995;149–150:103–126. doi: 10.1007/BF01076569. [DOI] [PubMed] [Google Scholar]
- 9.Kunst G, Kress KR, Gruen M, Uttenweiler D, Gautel M, Fink RH. Myosin binding protein c, a phosphorylation-dependent force regulator in muscle that controls the attachment of myosin heads by its interaction with myosin s2. Circ Res. 2000;86:51–58. doi: 10.1161/01.res.86.1.51. [DOI] [PubMed] [Google Scholar]
- 10.Pyo RT, Sui J, Dhume A, Palomeque J, Blaxall BC, Diaz G, Tunstead J, Logothetis DE, Hajjar RJ, Schecter AD. Cxcr4 modulates contractility in adult cardiac myocytes. J Mol Cell Cardiol. 2006;41:834–844. doi: 10.1016/j.yjmcc.2006.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hare JM. Nitric oxide and excitation-contraction coupling. J Mol Cell Cardiol. 2003;35:719–729. doi: 10.1016/s0022-2828(03)00143-3. [DOI] [PubMed] [Google Scholar]
- 12.Massion PB, Feron O, Dessy C, Balligand JL. Nitric oxide and cardiac function: Ten years after, and continuing. Circ Res. 2003;93:388–398. doi: 10.1161/01.RES.0000088351.58510.21. [DOI] [PubMed] [Google Scholar]
- 13.Malik ZA, Kott KS, Poe AJ, Kuo T, Chen L, Ferrara KW, Knowlton AA. Cardiac myocyte exosomes: Stability, hsp60, and proteomics. Am J Physiol Heart Circ Physiol. 2013;304:H954–965. doi: 10.1152/ajpheart.00835.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Datta R, Bansal T, Rana S, Datta K, Datta Chaudhuri R, Chawla-Sarkar M, Sarkar S. Myocyte-derived hsp90 modulates collagen upregulation via biphasic activation of stat-3 in fibroblasts during cardiac hypertrophy. Mol Cell Biol. 2017:37. doi: 10.1128/MCB.00611-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Garcia NA, Ontoria-Oviedo I, Gonzalez-King H, Diez-Juan A, Sepulveda P. Glucose starvation in cardiomyocytes enhances exosome secretion and promotes angiogenesis in endothelial cells. PLoS One. 2015;10:e0138849. doi: 10.1371/journal.pone.0138849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sahoo S, Losordo DW. Exosomes and cardiac repair after myocardial infarction. Circ Res. 2014;114:333–344. doi: 10.1161/CIRCRESAHA.114.300639. [DOI] [PubMed] [Google Scholar]
- 17.Campbell DL, Stamler JS, Strauss HC. Redox modulation of l-type calcium channels in ferret ventricular myocytes. Dual mechanism regulation by nitric oxide and s-nitrosothiols. J Gen Physiol. 1996;108:277–293. doi: 10.1085/jgp.108.4.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Xu L, Eu JP, Meissner G, Stamler JS. Activation of the cardiac calcium release channel (ryanodine receptor) by poly-s-nitrosylation. Science. 1998;279:234–237. doi: 10.1126/science.279.5348.234. [DOI] [PubMed] [Google Scholar]
- 19.Han X, Kobzik L, Balligand JL, Kelly RA, Smith TW. Nitric oxide synthase (nos3)-mediated cholinergic modulation of ca2+ current in adult rabbit atrioventricular nodal cells. Circ Res. 1996;78:998–1008. doi: 10.1161/01.res.78.6.998. [DOI] [PubMed] [Google Scholar]
- 20.Hartzell HC, Fischmeister R. Opposite effects of cyclic gmp and cyclic amp on ca2+ current in single heart cells. Nature. 1986;323:273–275. doi: 10.1038/323273a0. [DOI] [PubMed] [Google Scholar]
- 21.Gallo MP, Ghigo D, Bosia A, Alloatti G, Costamagna C, Penna C, Levi RC. Modulation of guinea-pig cardiac l-type calcium current by nitric oxide synthase inhibitors. J Physiol. 1998;506(Pt 3):639–651. doi: 10.1111/j.1469-7793.1998.639bv.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Seddon M, Shah AM, Casadei B. Cardiomyocytes as effectors of nitric oxide signalling. Cardiovasc Res. 2007;75:315–326. doi: 10.1016/j.cardiores.2007.04.031. [DOI] [PubMed] [Google Scholar]
- 23.Shah AM, Spurgeon HA, Sollott SJ, Talo A, Lakatta EG. 8-bromo-cgmp reduces the myofilament response to ca2+ in intact cardiac myocytes. Circ Res. 1994;74:970–978. doi: 10.1161/01.res.74.5.970. [DOI] [PubMed] [Google Scholar]
- 24.Layland J, Li JM, Shah AM. Role of cyclic gmp-dependent protein kinase in the contractile response to exogenous nitric oxide in rat cardiac myocytes. J Physiol. 2002;540:457–467. doi: 10.1113/jphysiol.2001.014126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ballard C, Schaffer S. Stimulation of the na+/ca2+ exchanger by phenylephrine, angiotensin ii and endothelin 1. J Mol Cell Cardiol. 1996;28:11–17. doi: 10.1006/jmcc.1996.0002. [DOI] [PubMed] [Google Scholar]
- 26.Yang HT, Sakurai K, Sugawara H, Watanabe T, Norota I, Endoh M. Role of na+/ca2+ exchange in endothelin-1-induced increases in ca2+ transient and contractility in rabbit ventricular myocytes: Pharmacological analysis with kb-r7943. Br J Pharmacol. 1999;126:1785–1795. doi: 10.1038/sj.bjp.0702454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Aiello EA, Villa-Abrille MC, Dulce RA, Cingolani HE, Perez NG. Endothelin-1 stimulates the na+/ca2+ exchanger reverse mode through intracellular na+ (na+i)-dependent and na+i-independent pathways. Hypertension. 2005;45:288–293. doi: 10.1161/01.HYP.0000152700.58940.b2. [DOI] [PubMed] [Google Scholar]
- 28.Dulce RA, Hurtado C, Ennis IL, Garciarena CD, Alvarez MC, Caldiz C, Pierce GN, Portiansky EL, Chiappe de Cingolani GE, Camilion de Hurtado MC. Endothelin-1 induced hypertrophic effect in neonatal rat cardiomyocytes: Involvement of na+/h+ and na+/ca2+ exchangers. J Mol Cell Cardiol. 2006;41:807–815. doi: 10.1016/j.yjmcc.2006.05.016. [DOI] [PubMed] [Google Scholar]
- 29.Alvarez BV, Perez NG, Ennis IL, Camilion de Hurtado MC, Cingolani HE. Mechanisms underlying the increase in force and ca(2+) transient that follow stretch of cardiac muscle: A possible explanation of the anrep effect. Circ Res. 1999;85:716–722. doi: 10.1161/01.res.85.8.716. [DOI] [PubMed] [Google Scholar]
- 30.Cingolani HE, Ennis IL, Aiello EA, Perez NG. Role of autocrine/paracrine mechanisms in response to myocardial strain. Pflugers Arch. 2011;462:29–38. doi: 10.1007/s00424-011-0930-9. [DOI] [PubMed] [Google Scholar]
- 31.Lauer MR, Gunn MD, Clusin WT. Endothelin activates voltage-dependent ca2+ current by a g protein-dependent mechanism in rabbit cardiac myocytes. J Physiol. 1992;448:729–747. doi: 10.1113/jphysiol.1992.sp019067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.He JQ, Pi Y, Walker JW, Kamp TJ. Endothelin-1 and photoreleased diacylglycerol increase l-type ca2+ current by activation of protein kinase c in rat ventricular myocytes. J Physiol. 2000;524(Pt 3):807–820. doi: 10.1111/j.1469-7793.2000.00807.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ono K, Tsujimoto G, Sakamoto A, Eto K, Masaki T, Ozaki Y, Satake M. Endothelin-a receptor mediates cardiac inhibition by regulating calcium and potassium currents. Nature. 1994;370:301–304. doi: 10.1038/370301a0. [DOI] [PubMed] [Google Scholar]
- 34.Cheng TH, Chang CY, Wei J, Lin CI. Effects of endothelin 1 on calcium and sodium currents in isolated human cardiac myocytes. Can J Physiol Pharmacol. 1995;73:1774–1783. doi: 10.1139/y95-242. [DOI] [PubMed] [Google Scholar]
- 35.Kamp TJ, Hell JW. Regulation of cardiac l-type calcium channels by protein kinase a and protein kinase c. Circ Res. 2000;87:1095–1102. doi: 10.1161/01.res.87.12.1095. [DOI] [PubMed] [Google Scholar]
- 36.Pi Y, Zhang D, Kemnitz KR, Wang H, Walker JW. Protein kinase c and a sites on troponin i regulate myofilament ca2+ sensitivity and atpase activity in the mouse myocardium. J Physiol. 2003;552:845–857. doi: 10.1113/jphysiol.2003.045260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang H, Grant JE, Doede CM, Sadayappan S, Robbins J, Walker JW. Pkc-betaii sensitizes cardiac myofilaments to ca2+ by phosphorylating troponin i on threonine-144. J Mol Cell Cardiol. 2006;41:823–833. doi: 10.1016/j.yjmcc.2006.08.016. [DOI] [PubMed] [Google Scholar]
- 38.Cuello F, Bardswell SC, Haworth RS, Yin X, Lutz S, Wieland T, Mayr M, Kentish JC, Avkiran M. Protein kinase d selectively targets cardiac troponin i and regulates myofilament ca2+ sensitivity in ventricular myocytes. Circ Res. 2007;100:864–873. doi: 10.1161/01.RES.0000260809.15393.fa. [DOI] [PubMed] [Google Scholar]
- 39.Cuello F, Bardswell SC, Haworth RS, Ehler E, Sadayappan S, Kentish JC, Avkiran M. Novel role for p90 ribosomal s6 kinase in the regulation of cardiac myofilament phosphorylation. J Biol Chem. 2011;286:5300–5310. doi: 10.1074/jbc.M110.202713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chen PP, Patel JR, Rybakova IN, Walker JW, Moss RL. Protein kinase a-induced myofilament desensitization to ca(2+) as a result of phosphorylation of cardiac myosin-binding protein c. J Gen Physiol. 2010;136:615–627. doi: 10.1085/jgp.201010448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Proven A, Roderick HL, Conway SJ, Berridge MJ, Horton JK, Capper SJ, Bootman MD. Inositol 1,4,5-trisphosphate supports the arrhythmogenic action of endothelin-1 on ventricular cardiac myocytes. J Cell Sci. 2006;119:3363–3375. doi: 10.1242/jcs.03073. [DOI] [PubMed] [Google Scholar]
- 42.Domeier TL, Zima AV, Maxwell JT, Huke S, Mignery GA, Blatter LA. Ip3 receptor-dependent ca2+ release modulates excitation-contraction coupling in rabbit ventricular myocytes. Am J Physiol Heart Circ Physiol. 2008;294:H596–604. doi: 10.1152/ajpheart.01155.2007. [DOI] [PubMed] [Google Scholar]
- 43.Harzheim D, Movassagh M, Foo RS, Ritter O, Tashfeen A, Conway SJ, Bootman MD, Roderick HL. Increased insp3rs in the junctional sarcoplasmic reticulum augment ca2+ transients and arrhythmias associated with cardiac hypertrophy. Proc Natl Acad Sci U S A. 2009;106:11406–11411. doi: 10.1073/pnas.0905485106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.van Wamel AJ, Ruwhof C, van der Valk-Kokshoom LE, Schrier PI, van der Laarse A. The role of angiotensin ii, endothelin-1 and transforming growth factor-beta as autocrine/paracrine mediators of stretch-induced cardiomyocyte hypertrophy. Mol Cell Biochem. 2001;218:113–124. doi: 10.1023/a:1007279700705. [DOI] [PubMed] [Google Scholar]
- 45.Gray MO, Long CS, Kalinyak JE, Li HT, Karliner JS. Angiotensin ii stimulates cardiac myocyte hypertrophy via paracrine release of tgf-beta 1 and endothelin-1 from fibroblasts. Cardiovasc Res. 1998;40:352–363. doi: 10.1016/s0008-6363(98)00121-7. [DOI] [PubMed] [Google Scholar]
- 46.Pedrotty DM, Klinger RY, Kirkton RD, Bursac N. Cardiac fibroblast paracrine factors alter impulse conduction and ion channel expression of neonatal rat cardiomyocytes. Cardiovasc Res. 2009;83:688–697. doi: 10.1093/cvr/cvp164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cartledge JE, Kane C, Dias P, Tesfom M, Clarke L, McKee B, Al Ayoubi S, Chester A, Yacoub MH, Camelliti P, Terracciano CM. Functional crosstalk between cardiac fibroblasts and adult cardiomyocytes by soluble mediators. Cardiovasc Res. 2015;105:260–270. doi: 10.1093/cvr/cvu264. [DOI] [PubMed] [Google Scholar]
- 48.Liau B, Jackman CP, Li Y, Bursac N. Developmental stage-dependent effects of cardiac fibroblasts on function of stem cell-derived engineered cardiac tissues. Sci Rep. 2017;7:42290. doi: 10.1038/srep42290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bang C, Batkai S, Dangwal S, et al. Cardiac fibroblast-derived microrna passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest. 2014;124:2136–2146. doi: 10.1172/JCI70577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Maxeiner H, Krehbiehl N, Muller A, Woitasky N, Akinturk H, Muller M, Weigand MA, Abdallah Y, Kasseckert S, Schreckenberg R, Schluter KD, Wenzel S. New insights into paracrine mechanisms of human cardiac progenitor cells. Eur J Heart Fail. 2010;12:730–737. doi: 10.1093/eurjhf/hfq063. [DOI] [PubMed] [Google Scholar]
- 51.Askar SF, Ramkisoensing AA, Atsma DE, Schalij MJ, de Vries AA, Pijnappels DA. Engraftment patterns of human adult mesenchymal stem cells expose electrotonic and paracrine proarrhythmic mechanisms in myocardial cell cultures. Circ Arrhythm Electrophysiol. 2013;6:380–391. doi: 10.1161/CIRCEP.111.000215. [DOI] [PubMed] [Google Scholar]
- 52.DeSantiago J, Bare DJ, Semenov I, Minshall RD, Geenen DL, Wolska BM, Banach K. Excitation-contraction coupling in ventricular myocytes is enhanced by paracrine signaling from mesenchymal stem cells. J Mol Cell Cardiol. 2012;52:1249–1256. doi: 10.1016/j.yjmcc.2012.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hwang HJ, Chang W, Song BW, et al. Antiarrhythmic potential of mesenchymal stem cell is modulated by hypoxic environment. J Am Coll Cardiol. 2012;60:1698–1706. doi: 10.1016/j.jacc.2012.04.056. [DOI] [PubMed] [Google Scholar]
- 54.Mayourian J, Cashman TJ, Ceholski DK, Johnson BV, Sachs D, Kaji DA, Sahoo S, Hare JM, Hajjar RJ, Sobie EA, Costa KD. Experimental and computational insight into human mesenchymal stem cell paracrine signaling and heterocellular coupling effects on cardiac contractility and arrhythmogenicity. Circ Res. 2017 doi: 10.1161/CIRCRESAHA.117.310796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sattayaprasert P, Nassal DM, Wan X, Deschenes I, Laurita KR. Mesenchymal stem cells suppress cardiac alternans by activation of pi3k mediated nitroso-redox pathway. J Mol Cell Cardiol. 2016;98:138–145. doi: 10.1016/j.yjmcc.2016.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.DeSantiago J, Bare DJ, Banach K. Ischemia/reperfusion injury protection by mesenchymal stem cell derived antioxidant capacity. Stem Cells Dev. 2013;22:2497–2507. doi: 10.1089/scd.2013.0136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.de Jong R, Houtgraaf JH, Samiei S, Boersma E, Duckers HJ. Intracoronary stem cell infusion after acute myocardial infarction: A meta-analysis and update on clinical trials. Circ Cardiovasc Interv. 2014;7:156–167. doi: 10.1161/CIRCINTERVENTIONS.113.001009. [DOI] [PubMed] [Google Scholar]
- 58.Hare JM, Fishman JE, Gerstenblith G, et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The poseidon randomized trial. JAMA. 2012;308:2369–2379. doi: 10.1001/jama.2012.25321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.De Hert SG, Gillebert TC, Andries LJ, Brutsaert DL. Role of the endocardial endothelium in the regulation of myocardial function. Physiologic and pathophysiologic implications. Anesthesiology. 1993;79:1354–1366. doi: 10.1097/00000542-199312000-00027. [DOI] [PubMed] [Google Scholar]
- 60.Brutsaert DL, De Keulenaer GW, Fransen P, Mohan P, Kaluza GL, Andries LJ, Rouleau JL, Sys SU. The cardiac endothelium: Functional morphology, development, and physiology. Prog Cardiovasc Dis. 1996;39:239–262. doi: 10.1016/s0033-0620(96)80004-1. [DOI] [PubMed] [Google Scholar]
- 61.Noireaud J, Andriantsitohaina R. Recent insights in the paracrine modulation of cardiomyocyte contractility by cardiac endothelial cells. Biomed Res Int. 2014;2014:923805. doi: 10.1155/2014/923805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Shah AM, MacCarthy PA. Paracrine and autocrine effects of nitric oxide on myocardial function. Pharmacol Ther. 2000;86:49–86. doi: 10.1016/s0163-7258(99)00072-8. [DOI] [PubMed] [Google Scholar]
- 63.Lewis MJ, Shah AM. Endothelial modulation of cardiac function. Australia ; United Kingdom: Harwood Academic; 1997. [Google Scholar]
- 64.Massion PB, Balligand JL. Modulation of cardiac contraction, relaxation and rate by the endothelial nitric oxide synthase (enos): Lessons from genetically modified mice. J Physiol. 2003;546:63–75. doi: 10.1113/jphysiol.2002.025973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kojda G, Kottenberg K, Nix P, Schluter KD, Piper HM, Noack E. Low increase in cgmp induced by organic nitrates and nitrovasodilators improves contractile response of rat ventricular myocytes. Circ Res. 1996;78:91–101. doi: 10.1161/01.res.78.1.91. [DOI] [PubMed] [Google Scholar]
- 66.Mohan P, Brutsaert DL, Paulus WJ, Sys SU. Myocardial contractile response to nitric oxide and cgmp. Circulation. 1996;93:1223–1229. doi: 10.1161/01.cir.93.6.1223. [DOI] [PubMed] [Google Scholar]
- 67.Vila-Petroff MG, Younes A, Egan J, Lakatta EG, Sollott SJ. Activation of distinct camp-dependent and cgmp-dependent pathways by nitric oxide in cardiac myocytes. Circ Res. 1999;84:1020–1031. doi: 10.1161/01.res.84.9.1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wegener JW, Godecke A, Schrader J, Nawrath H. Effects of nitric oxide donors on cardiac contractility in wild-type and myoglobin-deficient mice. Br J Pharmacol. 2002;136:415–420. doi: 10.1038/sj.bjp.0704740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Paolocci N, Ekelund UE, Isoda T, Ozaki M, Vandegaer K, Georgakopoulos D, Harrison RW, Kass DA, Hare JM. Cgmp-independent inotropic effects of nitric oxide and peroxynitrite donors: Potential role for nitrosylation. Am J Physiol Heart Circ Physiol. 2000;279:H1982–1988. doi: 10.1152/ajpheart.2000.279.4.H1982. [DOI] [PubMed] [Google Scholar]
- 70.Chesnais JM, Fischmeister R, Mery PF. Positive and negative inotropic effects of no donors in atrial and ventricular fibres of the frog heart. J Physiol. 1999;518(Pt 2):449–461. doi: 10.1111/j.1469-7793.1999.0449p.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yasuda S, Coutu P, Sadayappan S, Robbins J, Metzger JM. Cardiac transgenic and gene transfer strategies converge to support an important role for troponin i in regulating relaxation in cardiac myocytes. Circ Res. 2007;101:377–386. doi: 10.1161/CIRCRESAHA.106.145557. [DOI] [PubMed] [Google Scholar]
- 72.Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, Yazaki Y, Goto K, Masaki T. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature. 1988;332:411–415. doi: 10.1038/332411a0. [DOI] [PubMed] [Google Scholar]
- 73.Endoh M. Signal transduction and ca2+ signaling in intact myocardium. J Pharmacol Sci. 2006;100:525–537. doi: 10.1254/jphs.cpj06009x. [DOI] [PubMed] [Google Scholar]
- 74.Brunner F, Bras-Silva C, Cerdeira AS, Leite-Moreira AF. Cardiovascular endothelins: Essential regulators of cardiovascular homeostasis. Pharmacol Ther. 2006;111:508–531. doi: 10.1016/j.pharmthera.2005.11.001. [DOI] [PubMed] [Google Scholar]
- 75.Nagasaka T, Izumi M, Hori M, Ozaki H, Karaki H. Positive inotropic effect of endothelin-1 in the neonatal mouse right ventricle. Eur J Pharmacol. 2003;472:197–204. doi: 10.1016/s0014-2999(03)01936-8. [DOI] [PubMed] [Google Scholar]
- 76.Drawnel FM, Archer CR, Roderick HL. The role of the paracrine/autocrine mediator endothelin-1 in regulation of cardiac contractility and growth. Br J Pharmacol. 2013;168:296–317. doi: 10.1111/j.1476-5381.2012.02195.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Lipp P, Laine M, Tovey SC, Burrell KM, Berridge MJ, Li W, Bootman MD. Functional insp3 receptors that may modulate excitation-contraction coupling in the heart. Curr Biol. 2000;10:939–942. doi: 10.1016/s0960-9822(00)00624-2. [DOI] [PubMed] [Google Scholar]
- 78.Camelliti P, Borg TK, Kohl P. Structural and functional characterisation of cardiac fibroblasts. Cardiovasc Res. 2005;65:40–51. doi: 10.1016/j.cardiores.2004.08.020. [DOI] [PubMed] [Google Scholar]
- 79.Nag AC. Study of non-muscle cells of the adult mammalian heart: A fine structural analysis and distribution. Cytobios. 1980;28:41–61. [PubMed] [Google Scholar]
- 80.Banerjee I, Yekkala K, Borg TK, Baudino TA. Dynamic interactions between myocytes, fibroblasts, and extracellular matrix. Ann N Y Acad Sci. 2006;1080:76–84. doi: 10.1196/annals.1380.007. [DOI] [PubMed] [Google Scholar]
- 81.Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D’Antoni ML, Debuque R, Chandran A, Wang L, Arora K, Rosenthal NA, Tallquist MD. Revisiting cardiac cellular composition. Circ Res. 2016;118:400–409. doi: 10.1161/CIRCRESAHA.115.307778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.de Bakker JM, van Capelle FJ, Janse MJ, Tasseron S, Vermeulen JT, de Jonge N, Lahpor JR. Slow conduction in the infarcted human heart. ‘Zigzag’ course of activation. Circulation. 1993;88:915–926. doi: 10.1161/01.cir.88.3.915. [DOI] [PubMed] [Google Scholar]
- 83.Spach MS, Boineau JP. Microfibrosis produces electrical load variations due to loss of side-to-side cell connections: A major mechanism of structural heart disease arrhythmias. Pacing Clin Electrophysiol. 1997;20:397–413. doi: 10.1111/j.1540-8159.1997.tb06199.x. [DOI] [PubMed] [Google Scholar]
- 84.Nguyen HX, Kirkton RD, Bursac N. Engineering prokaryotic channels for control of mammalian tissue excitability. Nat Commun. 2016;7:13132. doi: 10.1038/ncomms13132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ongstad E, Kohl P. Fibroblast-myocyte coupling in the heart: Potential relevance for therapeutic interventions. J Mol Cell Cardiol. 2016;91:238–246. doi: 10.1016/j.yjmcc.2016.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Mahoney VM, Mezzano V, Morley GE. A review of the literature on cardiac electrical activity between fibroblasts and myocytes. Prog Biophys Mol Biol. 2016;120:128–133. doi: 10.1016/j.pbiomolbio.2015.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Long CS, Henrich CJ, Simpson PC. A growth factor for cardiac myocytes is produced by cardiac nonmyocytes. Cell Regul. 1991;2:1081–1095. doi: 10.1091/mbc.2.12.1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Aceto JF, Baker KM. [sar1]angiotensin ii receptor-mediated stimulation of protein synthesis in chick heart cells. Am J Physiol. 1990;258:H806–813. doi: 10.1152/ajpheart.1990.258.3.H806. [DOI] [PubMed] [Google Scholar]
- 89.Harada M, Itoh H, Nakagawa O, Ogawa Y, Miyamoto Y, Kuwahara K, Ogawa E, Igaki T, Yamashita J, Masuda I, Yoshimasa T, Tanaka I, Saito Y, Nakao K. Significance of ventricular myocytes and nonmyocytes interaction during cardiocyte hypertrophy: Evidence for endothelin-1 as a paracrine hypertrophic factor from cardiac nonmyocytes. Circulation. 1997;96:3737–3744. doi: 10.1161/01.cir.96.10.3737. [DOI] [PubMed] [Google Scholar]
- 90.Ponicke K, Heinroth-Hoffmann I, Becker K, Brodde OE. Trophic effect of angiotensin ii in neonatal rat cardiomyocytes: Role of endothelin-1 and non-myocyte cells. Br J Pharmacol. 1997;121:118–124. doi: 10.1038/sj.bjp.0701084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Leask A. Tgfbeta, cardiac fibroblasts, and the fibrotic response. Cardiovasc Res. 2007;74:207–212. doi: 10.1016/j.cardiores.2006.07.012. [DOI] [PubMed] [Google Scholar]
- 92.Rosenkranz S, Flesch M, Amann K, Haeuseler C, Kilter H, Seeland U, Schluter KD, Bohm M. Alterations of beta-adrenergic signaling and cardiac hypertrophy in transgenic mice overexpressing tgf-beta(1) Am J Physiol Heart Circ Physiol. 2002;283:H1253–1262. doi: 10.1152/ajpheart.00578.2001. [DOI] [PubMed] [Google Scholar]
- 93.Kuwahara K, Saito Y, Harada M, Ishikawa M, Ogawa E, Miyamoto Y, Hamanaka I, Kamitani S, Kajiyama N, Takahashi N, Nakagawa O, Masuda I, Nakao K. Involvement of cardiotrophin-1 in cardiac myocyte-nonmyocyte interactions during hypertrophy of rat cardiac myocytes in vitro. Circulation. 1999;100:1116–1124. doi: 10.1161/01.cir.100.10.1116. [DOI] [PubMed] [Google Scholar]
- 94.Horio T, Maki T, Kishimoto I, Tokudome T, Okumura H, Yoshihara F, Suga S, Takeo S, Kawano Y, Kangawa K. Production and autocrine/paracrine effects of endogenous insulin-like growth factor-1 in rat cardiac fibroblasts. Regul Pept. 2005;124:65–72. doi: 10.1016/j.regpep.2004.06.029. [DOI] [PubMed] [Google Scholar]
- 95.Guo W, Kamiya K, Yasui K, Kodama I, Toyama J. Paracrine hypertrophic factors from cardiac non-myocyte cells downregulate the transient outward current density and kv4.2 k+ channel expression in cultured rat cardiomyocytes. Cardiovasc Res. 1999;41:157–165. doi: 10.1016/s0008-6363(98)00157-6. [DOI] [PubMed] [Google Scholar]
- 96.Takeda N, Manabe I, Uchino Y, Eguchi K, Matsumoto S, Nishimura S, Shindo T, Sano M, Otsu K, Snider P, Conway SJ, Nagai R. Cardiac fibroblasts are essential for the adaptive response of the murine heart to pressure overload. J Clin Invest. 2010;120:254–265. doi: 10.1172/JCI40295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Pellieux C, Foletti A, Peduto G, Aubert JF, Nussberger J, Beermann F, Brunner HR, Pedrazzini T. Dilated cardiomyopathy and impaired cardiac hypertrophic response to angiotensin ii in mice lacking fgf-2. J Clin Invest. 2001;108:1843–1851. doi: 10.1172/JCI13627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Emanueli C, Shearn AI, Angelini GD, Sahoo S. Exosomes and exosomal mirnas in cardiovascular protection and repair. Vascul Pharmacol. 2015;71:24–30. doi: 10.1016/j.vph.2015.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Barile L, Lionetti V, Cervio E, Matteucci M, Gherghiceanu M, Popescu LM, Torre T, Siclari F, Moccetti T, Vassalli G. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res. 2014;103:530–541. doi: 10.1093/cvr/cvu167. [DOI] [PubMed] [Google Scholar]
- 100.Arslan F, Lai RC, Smeets MB, Akeroyd L, Choo A, Aguor EN, Timmers L, van Rijen HV, Doevendans PA, Pasterkamp G, Lim SK, de Kleijn DP. Mesenchymal stem cell-derived exosomes increase atp levels, decrease oxidative stress and activate pi3k/akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013;10:301–312. doi: 10.1016/j.scr.2013.01.002. [DOI] [PubMed] [Google Scholar]
- 101.Gray WD, French KM, Ghosh-Choudhary S, Maxwell JT, Brown ME, Platt MO, Searles CD, Davis ME. Identification of therapeutic covariant microrna clusters in hypoxia-treated cardiac progenitor cell exosomes using systems biology. Circ Res. 2015;116:255–263. doi: 10.1161/CIRCRESAHA.116.304360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sahoo S, Klychko E, Thorne T, Misener S, Schultz KM, Millay M, Ito A, Liu T, Kamide C, Agrawal H, Perlman H, Qin G, Kishore R, Losordo DW. Exosomes from human cd34(+) stem cells mediate their proangiogenic paracrine activity. Circ Res. 2011;109:724–728. doi: 10.1161/CIRCRESAHA.111.253286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Hart G. Cellular electrophysiology in cardiac hypertrophy and failure. Cardiovasc Res. 1994;28:933–946. doi: 10.1093/cvr/28.7.933. [DOI] [PubMed] [Google Scholar]
- 104.Boyden PA, Jeck CD. Ion channel function in disease. Cardiovasc Res. 1995;29:312–318. [PubMed] [Google Scholar]
- 105.Kaur K, Zarzoso M, Ponce-Balbuena D, Guerrero-Serna G, Hou L, Musa H, Jalife J. Tgf-beta1, released by myofibroblasts, differentially regulates transcription and function of sodium and potassium channels in adult rat ventricular myocytes. PLoS One. 2013;8:e55391. doi: 10.1371/journal.pone.0055391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Trieschmann J, Bettin D, Haustein M, Koster A, Molcanyi M, Halbach M, Hanna M, Fouad M, Brockmeier K, Hescheler J, Pfannkuche K, Hannes T. The interaction between adult cardiac fibroblasts and embryonic stem cell-derived cardiomyocytes leads to proarrhythmic changes in in vitro cocultures. Stem Cells Int. 2016;2016:2936126. doi: 10.1155/2016/2936126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ramos-Mondragon R, Vega AV, Avila G. Long-term modulation of na+ and k+ channels by tgf-beta1 in neonatal rat cardiac myocytes. Pflugers Arch. 2011;461:235–247. doi: 10.1007/s00424-010-0912-3. [DOI] [PubMed] [Google Scholar]
- 108.Zhang YE. Non-smad pathways in tgf-beta signaling. Cell Res. 2009;19:128–139. doi: 10.1038/cr.2008.328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Monsanto MM, White KS, Kim T, Wang BJ, Fisher K, Ilves K, Khalafalla FG, Casillas A, Broughton K, Mohsin S, Dembitsky WP, Sussman MA. Concurrent isolation of 3 distinct cardiac stem cell populations from a single human heart biopsy. Circ Res. 2017;121:113–124. doi: 10.1161/CIRCRESAHA.116.310494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Tang J, Shen D, Caranasos TG, Wang Z, Vandergriff AC, Allen TA, Hensley MT, Dinh PU, Cores J, Li TS, Zhang J, Kan Q, Cheng K. Therapeutic microparticles functionalized with biomimetic cardiac stem cell membranes and secretome. Nat Commun. 2017;8:13724. doi: 10.1038/ncomms13724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tang YL, Zhu W, Cheng M, Chen L, Zhang J, Sun T, Kishore R, Phillips MI, Losordo DW, Qin G. Hypoxic preconditioning enhances the benefit of cardiac progenitor cell therapy for treatment of myocardial infarction by inducing cxcr4 expression. Circ Res. 2009;104:1209–1216. doi: 10.1161/CIRCRESAHA.109.197723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Agarwal U, George A, Bhutani S, Ghosh-Choudhary S, Maxwell JT, Brown ME, Mehta Y, Platt MO, Liang Y, Sahoo S, Davis ME. Experimental, systems, and computational approaches to understanding the microrna-mediated reparative potential of cardiac progenitor cell-derived exosomes from pediatric patients. Circ Res. 2017;120:701–712. doi: 10.1161/CIRCRESAHA.116.309935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Bolli R, Chugh AR, D’Amario D, et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (scipio): Initial results of a randomised phase 1 trial. Lancet. 2011;378:1847–1857. doi: 10.1016/S0140-6736(11)61590-0. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 114.Heldman AW, DiFede DL, Fishman JE, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: The tac-hft randomized trial. JAMA. 2014;311:62–73. doi: 10.1001/jama.2013.282909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Karantalis V, DiFede DL, Gerstenblith G, et al. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The prospective randomized study of mesenchymal stem cell therapy in patients undergoing cardiac surgery (prometheus) trial. Circ Res. 2014;114:1302–1310. doi: 10.1161/CIRCRESAHA.114.303180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Karantalis V, Hare JM. Use of mesenchymal stem cells for therapy of cardiac disease. Circ Res. 2015;116:1413–1430. doi: 10.1161/CIRCRESAHA.116.303614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Liang X, Zhang L, Wang S, Han Q, Zhao RC. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring mir-125a. J Cell Sci. 2016;129:2182–2189. doi: 10.1242/jcs.170373. [DOI] [PubMed] [Google Scholar]
- 118.Merino-Gonzalez C, Zuniga FA, Escudero C, Ormazabal V, Reyes C, Nova-Lamperti E, Salomon C, Aguayo C. Mesenchymal stem cell-derived extracellular vesicles promote angiogenesis: Potencial clinical application. Front Physiol. 2016;7:24. doi: 10.3389/fphys.2016.00024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Liu JF, Wang BW, Hung HF, Chang H, Shyu KG. Human mesenchymal stem cells improve myocardial performance in a splenectomized rat model of chronic myocardial infarction. J Formos Med Assoc. 2008;107:165–174. doi: 10.1016/S0929-6646(08)60130-8. [DOI] [PubMed] [Google Scholar]
- 120.Ranganath SH, Levy O, Inamdar MS, Karp JM. Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell. 2012;10:244–258. doi: 10.1016/j.stem.2012.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Mirotsou M, Jayawardena TM, Schmeckpeper J, Gnecchi M, Dzau VJ. Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J Mol Cell Cardiol. 2011;50:280–289. doi: 10.1016/j.yjmcc.2010.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103:1204–1219. doi: 10.1161/CIRCRESAHA.108.176826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Eschenhagen T. Beta-adrenergic signaling in heart failure-adapt or die. Nat Med. 2008;14:485–487. doi: 10.1038/nm0508-485. [DOI] [PubMed] [Google Scholar]
- 124.Mureli S, Gans CP, Bare DJ, Geenen DL, Kumar NM, Banach K. Mesenchymal stem cells improve cardiac conduction by upregulation of connexin 43 through paracrine signaling. Am J Physiol Heart Circ Physiol. 2013;304:H600–609. doi: 10.1152/ajpheart.00533.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Chang MG, Tung L, Sekar RB, Chang CY, Cysyk J, Dong P, Marban E, Abraham MR. Proarrhythmic potential of mesenchymal stem cell transplantation revealed in an in vitro coculture model. Circulation. 2006;113:1832–1841. doi: 10.1161/CIRCULATIONAHA.105.593038. [DOI] [PubMed] [Google Scholar]
- 126.Beeres SL, Atsma DE, van der Laarse A, Pijnappels DA, van Tuyn J, Fibbe WE, de Vries AA, Ypey DL, van der Wall EE, Schalij MJ. Human adult bone marrow mesenchymal stem cells repair experimental conduction block in rat cardiomyocyte cultures. J Am Coll Cardiol. 2005;46:1943–1952. doi: 10.1016/j.jacc.2005.07.055. [DOI] [PubMed] [Google Scholar]
- 127.Pijnappels DA, Schalij MJ, van Tuyn J, Ypey DL, de Vries AA, van der Wall EE, van der Laarse A, Atsma DE. Progressive increase in conduction velocity across human mesenchymal stem cells is mediated by enhanced electrical coupling. Cardiovasc Res. 2006;72:282–291. doi: 10.1016/j.cardiores.2006.07.016. [DOI] [PubMed] [Google Scholar]
- 128.Mayourian J, Savizky RM, Sobie EA, Costa KD. Modeling electrophysiological coupling and fusion between human mesenchymal stem cells and cardiomyocytes. PLoS Comput Biol. 2016;12:e1005014. doi: 10.1371/journal.pcbi.1005014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Aratyn-Schaus Y, Pasqualini FS, Yuan H, McCain ML, Ye GJ, Sheehy SP, Campbell PH, Parker KK. Coupling primary and stem cell-derived cardiomyocytes in an in vitro model of cardiac cell therapy. J Cell Biol. 2016;212:389–397. doi: 10.1083/jcb.201508026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Luo L, Tang J, Nishi K, Yan C, Dinh PU, Cores J, Kudo T, Zhang J, Li TS, Cheng K. Fabrication of synthetic mesenchymal stem cells for the treatment of acute myocardial infarction in mice. Circ Res. 2017;120:1768–1775. doi: 10.1161/CIRCRESAHA.116.310374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Baglio SR, Rooijers K, Koppers-Lalic D, Verweij FJ, Perez Lanzon M, Zini N, Naaijkens B, Perut F, Niessen HW, Baldini N, Pegtel DM. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive mirna and trna species. Stem Cell Res Ther. 2015;6:127. doi: 10.1186/s13287-015-0116-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Teixeira FG, Panchalingam KM, Assuncao-Silva R, Serra SC, Mendes-Pinheiro B, Patricio P, Jung S, Anjo SI, Manadas B, Pinto L, Sousa N, Behie LA, Salgado AJ. Modulation of the mesenchymal stem cell secretome using computer-controlled bioreactors: Impact on neuronal cell proliferation, survival and differentiation. Sci Rep. 2016;6:27791. doi: 10.1038/srep27791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Anderson JD, Johansson HJ, Graham CS, et al. Comprehensive proteomic analysis of mesenchymal stem cell exosomes reveals modulation of angiogenesis via nuclear factor-kappab signaling. Stem Cells. 2016;34:601–613. doi: 10.1002/stem.2298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Kupcova Skalnikova H. Proteomic techniques for characterisation of mesenchymal stem cell secretome. Biochimie. 2013;95:2196–2211. doi: 10.1016/j.biochi.2013.07.015. [DOI] [PubMed] [Google Scholar]
- 135.Phinney DG, Di Giuseppe M, Njah J, et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle micrornas. Nat Commun. 2015;6:8472. doi: 10.1038/ncomms9472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Park CW, Kim KS, Bae S, Son HK, Myung PK, Hong HJ, Kim H. Cytokine secretion profiling of human mesenchymal stem cells by antibody array. Int J Stem Cells. 2009;2:59–68. doi: 10.15283/ijsc.2009.2.1.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Okumura H, Nagaya N, Itoh T, Okano I, Hino J, Mori K, Tsukamoto Y, Ishibashi-Ueda H, Miwa S, Tambara K, Toyokuni S, Yutani C, Kangawa K. Adrenomedullin infusion attenuates myocardial ischemia/reperfusion injury through the phosphatidylinositol 3-kinase/akt-dependent pathway. Circulation. 2004;109:242–248. doi: 10.1161/01.CIR.0000109214.30211.7C. [DOI] [PubMed] [Google Scholar]
- 138.Tao Z, Chen B, Tan X, Zhao Y, Wang L, Zhu T, Cao K, Yang Z, Kan YW, Su H. Coexpression of vegf and angiopoietin-1 promotes angiogenesis and cardiomyocyte proliferation reduces apoptosis in porcine myocardial infarction (mi) heart. Proc Natl Acad Sci U S A. 2011;108:2064–2069. doi: 10.1073/pnas.1018925108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Cashman TJ, Gouon-Evans V, Costa KD. Mesenchymal stem cells for cardiac therapy: Practical challenges and potential mechanisms. Stem Cell Rev. 2013;9:254–265. doi: 10.1007/s12015-012-9375-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Urbanek K, Rota M, Cascapera S, et al. Cardiac stem cells possess growth factor-receptor systems that after activation regenerate the infarcted myocardium, improving ventricular function and long-term survival. Circ Res. 2005;97:663–673. doi: 10.1161/01.RES.0000183733.53101.11. [DOI] [PubMed] [Google Scholar]
- 141.Bujak M, Dobaczewski M, Gonzalez-Quesada C, Xia Y, Leucker T, Zymek P, Veeranna V, Tager AM, Luster AD, Frangogiannis NG. Induction of the cxc chemokine interferon-gamma-inducible protein 10 regulates the reparative response following myocardial infarction. Circ Res. 2009;105:973–983. doi: 10.1161/CIRCRESAHA.109.199471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Kang K, Ma R, Cai W, Huang W, Paul C, Liang J, Wang Y, Zhao T, Kim HW, Xu M, Millard RW, Wen Z, Wang Y. Exosomes secreted from cxcr4 overexpressing mesenchymal stem cells promote cardioprotection via akt signaling pathway following myocardial infarction. Stem Cells Int. 2015;2015:659890. doi: 10.1155/2015/659890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Lai RC, Chen TS, Lim SK. Mesenchymal stem cell exosome: A novel stem cell-based therapy for cardiovascular disease. Regen Med. 2011;6:481–492. doi: 10.2217/rme.11.35. [DOI] [PubMed] [Google Scholar]
- 144.Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, Salto-Tellez M, Timmers L, Lee CN, El Oakley RM, Pasterkamp G, de Kleijn DP, Lim SK. Exosome secreted by msc reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4:214–222. doi: 10.1016/j.scr.2009.12.003. [DOI] [PubMed] [Google Scholar]
- 145.Baglio SR, Pegtel DM, Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front Physiol. 2012;3:359. doi: 10.3389/fphys.2012.00359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Nakamura Y, Miyaki S, Ishitobi H, Matsuyama S, Nakasa T, Kamei N, Akimoto T, Higashi Y, Ochi M. Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration. FEBS Lett. 2015;589:1257–1265. doi: 10.1016/j.febslet.2015.03.031. [DOI] [PubMed] [Google Scholar]
- 147.Xiao J, Pan Y, Li XH, Yang XY, Feng YL, Tan HH, Jiang L, Feng J, Yu XY. Cardiac progenitor cell-derived exosomes prevent cardiomyocytes apoptosis through exosomal mir-21 by targeting pdcd4. Cell Death Dis. 2016;7:e2277. doi: 10.1038/cddis.2016.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Wei C, Li L, Kim IK, Sun P, Gupta S. Nf-kappab mediated mir-21 regulation in cardiomyocytes apoptosis under oxidative stress. Free Radic Res. 2014;48:282–291. doi: 10.3109/10715762.2013.865839. [DOI] [PubMed] [Google Scholar]
- 149.Cheng Y, Liu X, Zhang S, Lin Y, Yang J, Zhang C. Microrna-21 protects against the h(2)o(2)-induced injury on cardiac myocytes via its target gene pdcd4. J Mol Cell Cardiol. 2009;47:5–14. doi: 10.1016/j.yjmcc.2009.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Soller KJ, Yang J, Veglia G, Bowser MT. Reversal of phospholamban inhibition of the sarco(endo)plasmic reticulum ca2+-atpase (serca) using short, protein-interacting rnas and oligonucleotide analogs. J Biol Chem. 2016;291:21510–21518. doi: 10.1074/jbc.M116.738807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Qiao S, Olson JM, Paterson M, Yan Y, Zaja I, Liu Y, Riess ML, Kersten JR, Liang M, Warltier DC, Bosnjak ZJ, Ge ZD. Microrna-21 mediates isoflurane-induced cardioprotection against ischemia-reperfusion injury via akt/nitric oxide synthase/mitochondrial permeability transition pore pathway. Anesthesiology. 2015;123:786–798. doi: 10.1097/ALN.0000000000000807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Agarwal U, George A, Bhutani S, Ghosh-Choudhary S, Maxwell JT, Brown ME, Mehta Y, Platt MO, Liang Y, Sahoo S, Davis ME. Experimental, systems and computational approaches to understanding the microrna-mediated reparative potential of cardiac progenitor cell-derived exosomes from pediatric patients. Circ Res. 2016 doi: 10.1161/CIRCRESAHA.116.309935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, Epstein SE. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res. 2004;94:678–685. doi: 10.1161/01.RES.0000118601.37875.AC. [DOI] [PubMed] [Google Scholar]
- 154.Gnecchi M, He H, Noiseux N, Liang OD, Zhang L, Morello F, Mu H, Melo LG, Pratt RE, Ingwall JS, Dzau VJ. Evidence supporting paracrine hypothesis for akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. Faseb J. 2006;20:661–669. doi: 10.1096/fj.05-5211com. [DOI] [PubMed] [Google Scholar]
- 155.Herrmann JL, Wang Y, Abarbanell AM, Weil BR, Tan J, Meldrum DR. Preconditioning mesenchymal stem cells with transforming growth factor-alpha improves mesenchymal stem cell-mediated cardioprotection. Shock. 2010;33:24–30. doi: 10.1097/SHK.0b013e3181b7d137. [DOI] [PubMed] [Google Scholar]
- 156.Schaaf S, Shibamiya A, Mewe M, Eder A, Stohr A, Hirt MN, Rau T, Zimmermann WH, Conradi L, Eschenhagen T, Hansen A. Human engineered heart tissue as a versatile tool in basic research and preclinical toxicology. PLoS One. 2011;6:e26397. doi: 10.1371/journal.pone.0026397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Tulloch NL, Muskheli V, Razumova MV, Korte FS, Regnier M, Hauch KD, Pabon L, Reinecke H, Murry CE. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ Res. 2011;109:47–59. doi: 10.1161/CIRCRESAHA.110.237206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Zhang D, Shadrin IY, Lam J, Xian HQ, Snodgrass HR, Bursac N. Tissue-engineered cardiac patch for advanced functional maturation of human esc-derived cardiomyocytes. Biomaterials. 2013;34:5813–5820. doi: 10.1016/j.biomaterials.2013.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Hirt MN, Hansen A, Eschenhagen T. Cardiac tissue engineering: State of the art. Circ Res. 2014;114:354–367. doi: 10.1161/CIRCRESAHA.114.300522. [DOI] [PubMed] [Google Scholar]
- 160.Liau B, Zhang D, Bursac N. Functional cardiac tissue engineering. Regen Med. 2012;7:187–206. doi: 10.2217/rme.11.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Ogle BM, Bursac N, Domian I, Huang NF, Menasche P, Murry CE, Pruitt B, Radisic M, Wu JC, Wu SM, Zhang J, Zimmermann WH, Vunjak-Novakovic G. Distilling complexity to advance cardiac tissue engineering. Sci Transl Med. 2016;8:342ps313. doi: 10.1126/scitranslmed.aad2304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Cashman TJ, Josowitz R, Gelb BD, Li RA, Dubois NC, Costa KD. Construction of defined human engineered cardiac tissues to study mechanisms of cardiac cell therapy. J Vis Exp. 2016:e53447. doi: 10.3791/53447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Turnbull IC, Karakikes I, Serrao GW, Backeris P, Lee JJ, Xie C, Senyei G, Gordon RE, Li RA, Akar FG, Hajjar RJ, Hulot JS, Costa KD. Advancing functional engineered cardiac tissues toward a preclinical model of human myocardium. Faseb J. 2014;28:644–654. doi: 10.1096/fj.13-228007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Serrao GW, Turnbull IC, Ancukiewicz D, Kim DE, Kao E, Cashman TJ, Hadri L, Hajjar RJ, Costa KD. Myocyte-depleted engineered cardiac tissues support therapeutic potential of mesenchymal stem cells. Tissue Eng Part A. 2012;18:1322–1333. doi: 10.1089/ten.tea.2011.0278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Stillitano F, Turnbull IC, Karakikes I, Nonnenmacher M, Backeris P, Hulot JS, Kranias EG, Hajjar RJ, Costa KD. Genomic correction of familial cardiomyopathy in human engineered cardiac tissues. Eur Heart J. 2016;37:3282–3284. doi: 10.1093/eurheartj/ehw307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Niwa N, Nerbonne JM. Molecular determinants of cardiac transient outward potassium current (i(to)) expression and regulation. J Mol Cell Cardiol. 2010;48:12–25. doi: 10.1016/j.yjmcc.2009.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Peralta AO, John RM, Gaasch WH, Taggart PI, Martin DT, Venditti FJ. The class iii antiarrhythmic effect of sotalol exerts a reverse use-dependent positive inotropic effect in the intact canine heart. J Am Coll Cardiol. 2000;36:1404–1410. doi: 10.1016/s0735-1097(00)00833-0. [DOI] [PubMed] [Google Scholar]
- 168.Tande PM, Refsum H. Class iii antiarrhythmic action linked with positive inotropy: Acute electrophysiological and inotropic effects of amiodarone in vitro. Pharmacol Toxicol. 1990;66:18–22. doi: 10.1111/j.1600-0773.1990.tb00694.x. [DOI] [PubMed] [Google Scholar]
- 169.Hoey A, Amos GJ, Ravens U. Comparison of the action potential prolonging and positive inotropic activity of dpi 201–106 and bdf 9148 in human ventricular myocardium. J Mol Cell Cardiol. 1994;26:985–994. doi: 10.1006/jmcc.1994.1119. [DOI] [PubMed] [Google Scholar]
- 170.Bouchard RA, Clark RB, Giles WR. Effects of action potential duration on excitation-contraction coupling in rat ventricular myocytes. Action potential voltage-clamp measurements. Circ Res. 1995;76:790–801. doi: 10.1161/01.res.76.5.790. [DOI] [PubMed] [Google Scholar]
- 171.Allen DG. On the relationship between action potential duration and tension in cat papillary muscle. Cardiovasc Res. 1977;11:210–218. doi: 10.1093/cvr/11.3.210. [DOI] [PubMed] [Google Scholar]
- 172.Wohlfart B. Relationships between peak force, action potential duration and stimulus interval in rabbit myocardium. Acta Physiol Scand. 1979;106:395–409. doi: 10.1111/j.1748-1716.1979.tb06419.x. [DOI] [PubMed] [Google Scholar]
- 173.Zhu R, Blazeski A, Poon E, Costa KD, Tung L, Boheler KR. Physical developmental cues for the maturation of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther. 2014;5:117. doi: 10.1186/scrt507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Hartman ME, Dai DF, Laflamme MA. Human pluripotent stem cells: Prospects and challenges as a source of cardiomyocytes for in vitro modeling and cell-based cardiac repair. Adv Drug Deliv Rev. 2016;96:3–17. doi: 10.1016/j.addr.2015.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Rajabi M, Kassiotis C, Razeghi P, Taegtmeyer H. Return to the fetal gene program protects the stressed heart: A strong hypothesis. Heart Fail Rev. 2007;12:331–343. doi: 10.1007/s10741-007-9034-1. [DOI] [PubMed] [Google Scholar]
- 176.Selem SM, Kaushal S, Hare JM. Stem cell therapy for pediatric dilated cardiomyopathy. Curr Cardiol Rep. 2013;15:369. doi: 10.1007/s11886-013-0369-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Chen G, Li S, Karakikes I, Ren L, Chow MZ, Chopra A, Keung W, Yan B, Chan CW, Costa KD, Kong CW, Hajjar RJ, Chen CS, Li RA. Phospholamban as a crucial determinant of the inotropic response of human pluripotent stem cell-derived ventricular cardiomyocytes and engineered 3-dimensional tissue constructs. Circ Arrhythm Electrophysiol. 2015;8:193–202. doi: 10.1161/CIRCEP.114.002049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Kerscher P, Turnbull IC, Hodge AJ, Kim J, Seliktar D, Easley CJ, Costa KD, Lipke EA. Direct hydrogel encapsulation of pluripotent stem cells enables ontomimetic differentiation and growth of engineered human heart tissues. Biomaterials. 2016;83:383–395. doi: 10.1016/j.biomaterials.2015.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Cashman TJ, Josowitz R, Johnson BV, Gelb BD, Costa KD. Human engineered cardiac tissues created using induced pluripotent stem cells reveal functional characteristics of braf-mediated hypertrophic cardiomyopathy. PLoS One. 2016;11:e0146697. doi: 10.1371/journal.pone.0146697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Janes KA, Kelly JR, Gaudet S, Albeck JG, Sorger PK, Lauffenburger DA. Cue-signal-response analysis of tnf-induced apoptosis by partial least squares regression of dynamic multivariate data. J Comput Biol. 2004;11:544–561. doi: 10.1089/cmb.2004.11.544. [DOI] [PubMed] [Google Scholar]
- 181.Janes KA, Albeck JG, Gaudet S, Sorger PK, Lauffenburger DA, Yaffe MB. A systems model of signaling identifies a molecular basis set for cytokine-induced apoptosis. Science. 2005;310:1646–1653. doi: 10.1126/science.1116598. [DOI] [PubMed] [Google Scholar]
- 182.Miller-Jensen K, Janes KA, Brugge JS, Lauffenburger DA. Common effector processing mediates cell-specific responses to stimuli. Nature. 2007;448:604–608. doi: 10.1038/nature06001. [DOI] [PubMed] [Google Scholar]
- 183.Platt MO, Wilder CL, Wells A, Griffith LG, Lauffenburger DA. Multipathway kinase signatures of multipotent stromal cells are predictive for osteogenic differentiation: Tissue-specific stem cells. Stem Cells. 2009;27:2804–2814. doi: 10.1002/stem.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Park KY, Li WA, Platt MO. Patient specific proteolytic activity of monocyte-derived macrophages and osteoclasts predicted with temporal kinase activation states during differentiation. Integr Biol (Camb) 2012;4:1459–1469. doi: 10.1039/c2ib20197f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Mosqueira D, Pagliari S, Uto K, et al. Hippo pathway effectors control cardiac progenitor cell fate by acting as dynamic sensors of substrate mechanics and nanostructure. ACS Nano. 2014;8:2033–2047. doi: 10.1021/nn4058984. [DOI] [PubMed] [Google Scholar]
- 186.Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126:677–689. doi: 10.1016/j.cell.2006.06.044. [DOI] [PubMed] [Google Scholar]
- 187.Williams AR, Hatzistergos KE, Addicott B, McCall F, Carvalho D, Suncion V, Morales AR, Da Silva J, Sussman MA, Heldman AW, Hare JM. Enhanced effect of combining human cardiac stem cells and bone marrow mesenchymal stem cells to reduce infarct size and to restore cardiac function after myocardial infarction. Circulation. 2013;127:213–223. doi: 10.1161/CIRCULATIONAHA.112.131110. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.




