Abstract
Studies in genetically modified mice have demonstrated that neuregulin-1 (NRG-1), along with the erythroblastic leukemia viral oncogene homolog (ErbB) 2, 3, and 4 receptor tyrosine kinases, is necessary for multiple aspects of cardiovascular development. These observations stimulated in vitro and in vivo animal studies, implicating NRG-1/ErbB signaling in the regulation of cardiac cell biology throughout life. Cardiovascular effects of ErbB2-targeted cancer therapies provide evidence in humans that ErbB signaling plays a role in the maintenance of cardiac function. These and other studies suggest a conceptual model in which a key function of NRG-1/ErbB signaling is to mediate adaptations of the heart to physiological and pathological stimuli through activation of intracellular kinase cascades that regulate tissue plasticity. Recent work implicates NRG-1/ErbB signaling in the regulation of multiple aspects of cardiovascular biology, including angiogenesis, blood pressure, and skeletal muscle responses to exercise. The therapeutic potential of recombinant NRG-1 as a potential treatment for heart failure has been demonstrated in animal models and is now being explored in clinical studies. NRG-1 is found in human serum and plasma, and it correlates with some clinical parameters, suggesting that it may have value as an indicator of prognosis. In this review, we bring together this growing literature on NRG-1 and its significance in cardiovascular development and disease.
Keywords: cardiac development, ErbB2, ErbB3, ErbB4, heart failure, heregulin, heart failure therapy
In 2012, there are 2 forms of recombinant neuregulin (NRG)-1β being examined as potential therapies for heart failure. It is unlikely that the cardiovascular effects of NRG-1 were anticipated by the early investigators who cloned gene products of the NRG-1 gene1 and later inactivated NRG-1 in mice.2 Neuroscientists were exploring the developmental requirement of NRG-1 and erythroblastic leukemia viral oncogene homolog (ErbB) receptors in the central nervous system when they discovered that not only NRG-1 but also ErbB2 and ErbB4 receptors were critical to normal cardiac development. During the past 17 years since those publications, interest in the role of NRG-1/ErbB signaling in the heart has grown steadily fueled in part by the observations of oncologists developing ErbB2-targeted therapies for cancer. A growing body of literature suggests diverse functions for NRG-1/ErbB signaling in the adult cardiovascular system. In this review, we bring together this growing literature and identify important gaps that require further investigation.
Overview of NRG and ErbB Receptor Biology
The NRG-1 gene is a member of the epidermal growth factor (EGF) gene family. There are 4 structurally related genes encoding NRGs (NRG-1, NRG-2, NRG-3, and NRG-4), with NRG-1 being the most abundant in the cardiovascular system.3,4 Alternative splicing results in multiple isoforms of NRG-1, all of which share the common EGF domain that is essential for NRG-1 ligand function. Alternative splicing at the C-terminal of the EGF domain of NRG-1 leads to α- and β-variants that determine receptor affinity. NRG-1 gene products can be further subdivided into 3 types (types I, II, and III). Type I NRG-1 proteins are type 1 transmembrane proteins with a C-2 immunoglobulin-like (Ig) domain. Type II NRG-1 proteins express an N-terminal secretory signal peptide, a matrix-interacting kringle, and immunoglobulin domains, and are truncated after the β-domain, resulting in an active ligand on secretion. Type III NRG-1 proteins are almost exclusively expressed in neuronal cells and have a cysteine-rich domain that binds membranes and creates a tethered ligand. In type I NRG-1 proteins, the active form of NRG-1 is released after proteolytic cleavage at the juxtamembrane region lying on the carboxyl-terminal side of the EGF-like domain. The cleavage of type I NRG-1 is essential for its function and it is catalyzed by 3 type I transmembrane proteases: tumor necrosis factor-α–converting enzyme (also known as a disintegrin and metalloprotease 17);5 β-site of amyloid precursor protein–cleaving enzyme (also known as memapsin 2);6 and meltrin β (also known as a disintegrin and metalloprotease 19).7
NRG-1 exerts its effect in a paracrine manner via the ErbB family of tyrosine kinase receptors (ErbB2, ErbB3, and ErbB4).8 ErbB receptors consist of an extracellular ligand-binding domain, a transmembrane domain, and a cytoplasmic tyrosine kinase domain. NRG-1 binds to ErbB3 and ErbB4 (but not ErbB2) and induces a conformational change in ErbB3 or ErbB4 that allows dimerization with ErbB2 or ligand-activated ErbB3 or ErbB4 receptors. Intracellular kinase domains subsequently phosphorylate the intracellular C-terminal domain of the dimerization partner, providing docking sites for various adaptor proteins, such as SHC-adaptor protein, growth factor receptor-bound protein 2, and the regulatory subunit of phosphatidylinositol-3-kinase (PI3K), and downstream signaling that mediates several processes in many tissues that are time- and context-dependent.
NRG/ErbB Signaling Regulates Multiple Aspects of Cardiac Development
Specific disruption of NRG-1 or the ErbB2, ErbB3, or ErbB4 receptors in genetically altered mice leads to failure of cardiac development (Figure 1). Mice with disrupted NRG-1, ErbB2, or ErbB4 expression die at days 9 to 10 in utero with many defects, including lack of ventricular trabeculation, a process necessary for development of a full-thickness ventricular wall.2,9,10 Mice expressing a kinase-dead ErbB2 showed similar embryonic defects.11 At this stage of development, NRG-1 is expressed in the endocardium, whereas ErbB2 and ErbB4 are expressed in ventricular myocytes. ErbB3 is expressed in the mesenchymal cells of the endocardial cushions of the fetal heart, which is involved in the formation of the valves.12 ErbB3-null mice demonstrate defects in the endocardial cushion, also leading to embryonic lethality. However, trabeculation was essentially normal. ErbB3 also is expressed in prenatal cardiac myocytes and vascular endothelial cells13 and has been reported in adult mouse cardiac myocytes,14 suggesting that there may be additional roles of ErbB3 in myocardial development beyond valve formation.
Figure 1. Role of neuregulin-1 signaling in cardiac development.
NRG-1/ErbB signaling is necessary for multiple aspect of cardiac development (see text for details). SA indicates sinoatrial; AV, atrioventricular; AVC, atrioventricular canal.
Mice with targeted deletion of the EGF domain of NRG-1, required for all isoforms of the gene, die in utero and have developmental defects in trabeculation identical to the ErbB2 and ErbB4 knockout mice, as well as endocardial cushion defects similar to those of the ErbB3 knockout mice.2 Interestingly, mice with specific disruption of exons encoding NRG-1 cytoplasmic/transmembrane domains15 or Ig-like domain16 die in utero with similar cardiac developmental defects. The NRG-1β/ErbB signaling pathway is involved in embryonic stem cell differentiation into cardiac myocyte lineages17,18 and in the developing embryo regulates induction of the cardiac gene regulatory network in nontrabecular as well as trabecular myocardium.19 These results can be collectively interpreted as showing that endocardium-derived NRG-1 acts on myocyte ErbB2/ErbB4 heterodimers to mediate ventricular myocyte proliferation as well as differentiation, regulating the progression from a single-cell layer of ventricular myocardium to the formation of trabeculae. Interestingly, NRG-1/ErbB signaling also is critical for the development of the cardiac conduction system. NRG-1β stimulates differentiation of embryonic cardiomyocytes into cells of the cardiac conduction system, with an increase in pacemaker current density, and transforms fetal cardiomyocytes into cardiac pacemaker-like cells.20 The concurrent events that determine whether NRG-1 stimulation drives a cell toward contractile vs conduction phenotype are yet unknown.
NRG-1/ErbB Signaling in Adult Heart Structure and Function
NRG-1/ErbB signaling has a role in the postnatal/adult heart that is distinct from the prenatal/embryogenesis stages of cardiac development (Figure 2). The first evidence for this was found in humans during the development of ErbB2-targeted (also known as human epidermal growth factor receptor-2/neu) therapies for cancer. Patients with meta-static breast cancer receiving the humanized monoclonal anti-ErbB2 antibody trastuzumab alone or in combination with anthracyclines were found to have a high incidence of heart failure and left ventricular systolic dysfunction.21 In vitro and in vivo work has since developed several possible mechanisms for this observation.
Figure 2. Biological and physiological role of neuregulin-1 signaling in adult heart.
Tissue activities known to be regulated by NRG-1/ErbB signaling in the adult cardiovascular system are shown (see text for details). EPC indicates endothelial progenitor cell.
Conditional cardiac myocyte–specific disruption of ErbB222,23 and ErbB424 has been examined in mice expressing myosin heavy chain promoter–regulated Cre-recombinase and floxed-ErbB receptors. These mice survive to birth but have development of spontaneous dilated cardiomyopathy, with increased susceptibility to stresses such as pressure overload and cardiotoxic anthracycline antibiotics. Ultrastructure analysis shows excess mitochondria and thin myofilaments.23 ErbB2 conditional mutant mice displayed a lengthened ventricular repolarization time, suggesting some impairment in the conduction system as well.22 Cardiac myocytes from conditional ErbB4 deletion mice exhibit both delayed conduction and impaired contractility.24
One interpretation of these findings is that the NRG-1/ErbB system is required for physiological adaptation of the heart to changes in cardiac demand. This concept is supported by studies examining NRG-1/ErbB signaling during cardiac adaptation to pregnancy.25 During pregnancy in mice, left ventricular NRG-1/ErbB signaling is activated. Suppressing ErbB signaling with an ErbB1/2 tyrosine kinase inhibitor during pregnancy was associated with increased ventricular dilation and decreased systolic function, with premature death of pregnant mice. These studies collectively demonstrate the importance of an intact NRG-1β/ErbB signaling axis for adaptation of cardiac structure and function.
In vitro studies have demonstrated likely cellular mechanisms of how NRG/ErbB signaling may regulate cardiac adaptation to stress. This work collectively supports the concept that this is a paracrine and juxtacrine system that regulates the interaction between microvascular endothelial cells and cardiac myocytes (Figure 3). Multiple isoforms of NRG-1 are expressed in microvascular endothelial cells.26 Because these are almost entirely type I pre-NRGs, proteolytic activation is required for NRG-1 release from endothelial cells as an active ligand. Mild oxidative stress activates NRG-1 release and activity in vitro via a member of the matrix metalloproteinase family,27 as predicted by work in neuronal cell systems.5 Other physiological stimuli implicated in the regulation of NRG-1 expression and activity include angiotensin and phenylephrine, as well as endothelin.28 Although multiple NRG-1α isoforms are expressed in cardiac microvascular endothelial cells, it is yet unclear what role these play in the heart.
Figure 3. Neuregulin-1 signaling in cardiac myocytes.
NRG-1 is expressed in the microvascular endothelium, whereas ErbB2 and ErbB4 are expressed in ventricular cardiac myocytes. NRG-1 activates several distinct and interacting signaling cascades, including extracellular signal-regulated kinase (Erk)1/2, mitogen-activated protein kinase (MAPK), phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt), and Src/focal adhesion kinase (FAK), leading to downstream changes in myocyte function (see text for details). EPC indicates endothelial progenitor cell; ROS, reactive oxygen species; eNOS, endothelial NO synthase; PKG, protein kinase G; PLN, phospholamban; SERCA, sarcoendoplasmic reticulum calcium ATPase.
ErbB2 and ErbB4 are expressed in cardiac myocytes of the adult heart.13 ErbB3 expression also has been demonstrated in the adult myocardium, although its function has not been fully examined.14 Recombinant NRG-1β activates ErbB2 and ErbB4 receptor phosphorylation in isolated cardiac myocytes, with activation of protein kinase B (Akt)/PI3K, MEK/extracellular signal-regulated kinase, Src/focal adhesion kinase, and NO synthase, all of which have been linked to cellular responses, including cell survival,13,27,29 mitochondrial function,30 proliferation,31 growth,32 glucose uptake,26 sarcoplasmic reticulum calcium uptake,33 and focal adhesion formation.34 NRG-1β also regulates adrenergic responses in cardiac myocytes, having a negative inotropic effect in isolated rabbit papillary muscles,35 and protects myocytes from β1-adrenergic receptor–induced cell death.36 NRG-1β also regulates muscarinic cholinergic receptor expression and activity.37 Whether these antiadrenergic effects account for the deleterious effects of trastuzumab on cardiac function are yet to be fully examined.
Examination of what happens to cardiac myocytes when ErbB receptor expression and activity are disrupted in cell culture suggests other potential mechanisms for trastuzum-ab-associated cardiac dysfunction. Treatment of adult myocytes with an antibody to extracellular domain of ErbB2 causes early activation of extracellular signal-regulated kinase 1/2 that is likely caused by activation of ErbB2-ErbB2 dimers and downstream ErbB2-specific signaling while increasing myocyte sensitivity to anthra cyclines, leading to myofilament disorganization in adult myocytes.38
NRG-1/ErbB activity interacts with myocardial metabolism, at least in vitro. NRG-1β induces cardiac myocyte glucose uptake in a PI3K-dependent manner to a similar magnitude as insulin.26 Interestingly, excess saturated fatty acid exposure, at least in vitro, induces a form of NRG resistance similar to the well-established and clinically important insulin resistance of type II diabetes mellitus. Treatment of cardiac myocytes with the saturated fatty acid palmitate seems to prevent ErbB coupling to the antiapoptotic PI3K/Akt pathway.39 These in vitro effects are suppressed by small amounts of the monounsaturated fatty acid oleate. It is interesting to consider whether disruption of NRG-1/ErbB signaling, like insulin signaling, is a general phenomenon that contributes to end-organ damage in the setting of type II diabetes mellitus.
Metabolic stress, as seen in ischemic heart disease and chronic heart failure, also disrupts NRG-1/ErbB signaling via effects on heat shock protein 90 chaperone activity. Heat shock protein 90 requires ATP for functional protection of client proteins, including ErbB2.40 Inhibition of glycolysis or mitochondrial respiration in isolated myocytes causes heat shock protein 90 to rapidly dissociate from ErbB2.41 This, in turn, leads to degradation of ErbB2 and reduced NRG activation of extracellular signal-regulated kinase and Akt signaling. This phenomenon may account for reduced sensitivity of cardiac myocytes to NRG-1β under conditions of combined treatment with β-adrenergic receptor activation and electric pacing36 or of the intact heart late after myocardial infarction.42 Myocardial responsiveness to endogenous or exogenous NRG-1β is likely to be a function of these and other effects on receptor expression and stability.
NRG Regulation of Vascular Biology and Angiogenesis
The developmental role of NRG-1/ErbB signaling described has focused a lot of attention on its role in cardiac myocytes. In addition, NRG-1 seems to play an important role in regulating the structure and function of the vasculature in many organs. Both NRG-1 and ErbB receptors are expressed in vascular endothelial cells, and stimulation of endothelial cells in vitro or in vivo induces an angiogenic response similar to and independent of vascular endothelial growth factor.43 NRG-1 also can induce angiogenesis via enhanced expression of vascular endothelial growth factor.44 The ErbB3/4 ligands β-cellulin and amphiregulin have been implicated in the recruitment of vascular smooth muscle cells necessary for arteriogenesis.45,46 These findings suggest that one mechanism for the antitumor effect of ErbB2-targeted cancer therapies may be inhibition of angiogenesis.47 The concept that NRG-1 regulates angiogenesis in the setting of tissue ischemia recently has been demonstrated in a model of hindlimb ischemia.48
Endothelial progenitor cells (EPCs) originating in the bone marrow play a role in tissue repair after ischemic injury.49 Stimuli such as ischemic injury and exercise, which have been shown to activate tissue NRG-1,27,50 are also potent inducers of EPC mobilization and recruitment to the heart and skeletal muscle.49,51 It is interesting that EPCs express ErbB2 and ErbB3, and NRG-1 regulates EPC biology, with effects on survival as well as induction of differentiation signaling.52 The extent to which NRG-1 modulation of EPC function is involved in myocardial recovery after ischemic injury, ischemia-induced angiogenesis, and the beneficial effects of exercise warrant further investigation.
NRG in the Central and Peripheral Nervous System: Implications for Cardiovascular Function
NRG-1 and ErbB receptors are distributed extensively in the central and peripheral nervous systems.53,54 The biological function of NRG-1/ErbB signaling in the nervous system is similar to that seen in the cardiovascular system: it is required for proper tissue organization and maturation in development, is activated in response to injury, and is a mediator for repair/recovery (Table 1). NRG-1 and its ErbB receptors are upregulated in the central nervous system after ischemia/injury, and administration of exogenous NRG-1β exerts a neuroprotective effect.55 Pretreatment with exogenous NRG-1β before cerebral ischemia and reperfusion resulted in significant improvement in the recovery of neurological function in rats, as well as infarct volume reduction.56,57 NRG-1 and its receptors are upregulated after traumatic brain injury in rat models.58,59 NRG-1β exerts a cytoprotective effect against oxidant injury in multiple cell types in the brain, including the microvascular endothelial cell.60
Table 1.
Therapeutic Application and Effect of Recombinant NRG-1 in Noncardiac Adult Tissue
| Author | Formulation of NRG-1 | Tissue | Pathology | Outcome |
|---|---|---|---|---|
| Marchionni et al94 | rhGGF2 | CNS | Encephalomyelitis (mouse model of multiple sclerosis) | Delayed signs, decreased severity, reduction in relapse rate, and increased remyelination |
| Xu et al95 | rhNRG-1 | Acute ischemic stroke (transient occlusion) | Reduced cortical infarct volume, blocked apoptosis, prevented macrophage/microglial infiltration and astrocyte activation | |
| Iaci et al57 | rhGGF2 | Chronic stroke (permanent occlusion) | Neuro-restorative effects; functional improvements and improved sensorimotor recovery | |
| Cai et al96 | rhNRG-1β | PNS | Sciatic nerve lesion | Improved nerve regeneration across gap |
| Krag et al97 | rhNRG-1β | Skeletal muscle | Muscular dystrophy mouse | Improved muscle function in muscular dystrophy model |
CNS indicates central nervous system; NRG indicates neuregulin-1; PNS, peripheral nervous system; rhGGF2, recombinant human glial growth factor 2; rhNRG, recombinant human neuregulin-1.
Some recent findings suggest that central nervous system NRG-1/ErbB signaling plays an important role in cardiovascular homeostasis. The rostral ventrolateral medulla of the brainstem is 1 of the most important regions involved in the central regulation of vasomotor tone. Injection of recombinant NRG-1β into the rostral ventrolateral medulla in rats lowered blood pressure, heart rate, and renal sympathetic nerve activity.61 Suppressing ErbB expression with small interfering RNA had the opposite effect, suggesting that baseline NRG-1/ErbB signaling modulates rostral ventrolateral medulla sympathetic tone. Whether these intriguing findings translate to new mechanisms for blood pressure control and hypertension warrants further investigation.
NRG in Skeletal Muscle: Mediator of Cardiovascular Benefits of Exercise?
NRG-1 and the ErbB receptors are important determinants of skeletal muscle biology, and this may have implications for cardiovascular function. NRG-1β induces myogenesis62 and myoblast differentiation.63 NRG-1 and ErbB receptors localize to the neuromuscular junction in skeletal muscle, where activation induces expression of postsynaptic acetylcholine receptor expression.64–66 Conditional knockout of ErbB2 in skeletal muscle leads to multiple defects in muscle, including decreased numbers of muscle spindles, impaired regeneration after injury, and defects in proprioception.67 Skeletal muscle NRG-1/ErbB signaling is activated in animals acutely by exercise,50 leading to the hypothesis that NRG-1β may mediate many of the effects of exercise (growth, glucose homeostasis) on skeletal muscle. In this context, it is interesting that NRG-1β stimulates skeletal muscle glucose uptake at comparable levels and is additive to the effects of insulin.68 This effect is mediatedviaPI3K/Akt/Akt-dependent translocation of glucose transporters (GLUT) to muscle cell membrane. Exercise training is known to improve glucose homeostasis, with clear benefits to cardiovascular function. Skeletal muscle ErbB3 expression was significantly increased in skeletal muscle in response to a progressive resistance training intervention in healthy adults.69 Fitness in healthy humans is associated with higher levels of circulating NRG-1β.70 Collectively, these findings lead us to speculate that skeletal muscle NRG-1β is an exercise hormone with actions at a distance, including the heart.
Alteration of ErbB Receptor Expression in Cardiac Pathology
NRG-1/ErbB signaling and expression have been studied in several cardiovascular disease states with interesting patho-physiological implications. In the setting of pressure overload induced by aortic banding in mice, expression of ErbB2 and ErbB4 mRNA levels in the left ventricle increase early during compensatory cardiac hypertrophy.71 After the transition to heart failure, however, myocardial ErbB2/ErbB4 mRNA and protein levels are decreased compared with control hearts. In diabetic animals, myocardial NRG-1 protein expression and ErbB2/ErbB4 phosphorylation and mRNA expression are also reduced.72 In ventricular tissue from patients with advanced heart failure, increased NRG-1 and decreased ErbB2/ErbB4 mRNA expression have been observed.73 Based on the deleterious effects of conditional deletion of ErbB2 and ErbB4 on myocardial structure and animal survival, it seems reasonable to speculate that the decreased ErbB receptor expression in late stages of heart failure may contribute to progressive myocardial dysfunction. Interestingly, in patients who had left ventricular unloading with ventricular assist devices, the changes reversed.73,74 Whether this contributes to the reversal of gene expression changes and improved myocardial function that is seen in some patients remains to be determined.
Endogenous NRG-1/ErbB Signaling Protects the Heart From Ischemic Injury
Ischemia/reperfusion injury is a potent activator of NRG-1/ErbB signaling in the intact heart.27 Selective disruption of endothelial NRG-1 expression impairs recovery of cardiac contractile function after an ischemic insult.75 Similar results are observed if the heart is perfused with a small-molecule inhibitor of ErbB2 tyrosine kinase activity.76 NRG-1 preconditioning has cardioprotective effects against ischemia/reperfusion injury via a PI3K/Akt-dependent mechanism.77 Whether the activation of NRG-1 by ischemia/reperfusion also participates in the well-known phenomenon of ischemic preconditioning is unknown.
NRG as a Potential Therapy for Myocardial Injury and Heart Failure
Multiple in vivo studies have established the therapeutic potential for recombinant human (rh) NRG-1β (Table 2). An EGF-domain fragment of rhNRG-1β was used in a series of small and large animal models of systolic heart failure to demonstrate an effect on heart function and survival.42 In rats with systolic dysfunction induced by surgical myocardial infarction, intravenous doses of the rhNRG-1β fragment administered daily for 5 days improved cardiac function. Similar beneficial effects on cardiac function were reported in models of anthracycline and virally induced cardiac injury in mice. In large animals with rapid-pacing–induced heart failure, the rhNRG-1β fragment similarly improved cardiac contractility and relaxation.42,78
Table 2.
Studies of Effects of Recombinant NRG-1 on Heart Failure (Animal Studies)
| Author | Formulation of NRG-1 | Heart Failure Model | Outcome |
|---|---|---|---|
| Liu et al42 | rhNRG-1 EGF domain only | Ischemic/post-MI (rats) | Attenuated dysfunction and improvements in cardiac performance. Increased capillary abundance in the fibrotic peri-infarct area (angiogenesis) |
| Viral myocarditis (mice) | Improved echocardiographic parameter and survival | ||
| Chronic rapid pacing model (dogs) | Improved LVEDP and LVESP, improved cardiac contractility and relaxation | ||
| Drug-induced (mice) | Prevented cardiac dysfunction and improved survival | ||
| Bersell et al31 | rhNRG-1 (+Ig domain) | Ischemic/post-MI (mice) | Induced cell cycle in cardiac myocyte and improved cardiac function and structure after MI |
| Gu et al98 | rhNRG-1 EGF domain only | Post-MI–induced heart failure (mice) | Upregulated cardiac myosin light chain kinase associated with improved cardiac function |
| Guo et al82 | rhNRG-1 EGF domain only | Post-MI–induced heart failure (rats) | Improved LV remodeling and cardiac function, reducing mitochondrial dysfunction, myocyte apoptosis, and oxidative stress |
| Bian et al99 | rhGGF2 | Doxorubicin-induced heart failure (mice) | Improved survival and cardiac function in doxorubicin-induced heart failure |
| Hill et al80 | rhGGF2 | Post-MI–induced heart failure (rats) | Improved LV contractile function, corrected altered gene expression. |
| Kasasbeh et al81 | rhGGF2 | Post-MI–induced heart failure (swine) | Improved LV contractive function, no effect on diastolic function or inotropic reserve |
EGF indicates epidermal growth factor; LV, left ventricular; LVEDP, left ventricular end-diastolic pressure; LVESP, left ventricular end-systolic pressure; MI, myocardial infarction; NRG indicates neuregulin-1; rhNRG, recombinant human neuregulin-1; rhGGF2, recombinant human glial growth factor 2.
A second form of rhNRG-1β undergoing examination is a kringle and Ig domain–containing version of NRG-1β known as glial growth factor 2 (GGF2). The Ig domain is expressed in all myocardial NRG-1 gene products and localizes ligand through interaction with matrix molecules. In in vitro assays of myocyte response to recombinant NRG-1β, the EGF domain–only fragment of NRG-1β has equal potency in activating intracellular signaling compared with the larger GGF2, but is shorter-acting.79 GGF2 treatment of rats or swine with myocardial infarction–induced myocardial dysfunction is associated with improved systolic function and reduced progressive remodeling.80,81
As discussed, there are many effects of NRG-1β on the biology of heart and vascular cells that could be contributing to the beneficial effects of NRG-1β on cardiac function. Protection of myocytes from stimuli that lead to cell death, together with effects on sarcomere gene expression and stability, may lead to improved cardiac function through repair of dysfunctional cardiac myocytes. NRG-1β seems to induce myocyte proliferation in rodents.31 At this point, it is unclear whether NRG-1β-induced cardiac myocyte proliferation contributes to the improvement in cardiac function observed. In swine, GGF2 treatment is associated with markedly improved mitochondrial structure by electron microscopy.81 This is consistent with reports of improved mitochondrial function in animals treated with NRG-1β.82 Collectively, this work suggests that the effects of NRG-1β on cardiac contractile function are pleiotropic.
The promising preclinical animal studies have led to ongoing clinical trials examining whether these rhNRG-1βs have similar effects in humans. A phase II, double-blinded, placebo-controlled study to evaluate the efficacy and safety of recombinant EGF-domain rhNRG-1β in patients with chronic heart failure has been completed.83 Patients with New York Heart Association functional class II or III systolic heart failure received placebo or EGF-domain rhNRG-1β (0.3, 0.6, or 1.2 μg/kg per day) by intravenous infusion over a period of hours, daily for 10 days. Three months later, improved cardiac function as measured by magnetic resonance imaging was observed in the intermediate dose group. The acute and chronic responses to EGF-domain rhNRG-1β also have been reported in patients with stable congestive heart failure.84 Patients receiving an EGF-domain rhNRG-1β infusion daily for 11 days showed an acute increase in cardiac output that was accompanied by a vasodilator effect. At 12 weeks after treatment, there was a 12% increase in ventricular systolic function from baseline. The authors concluded that treatment with EGF-domain rhNRG-1β in patients with stable congestive heart failure on optimal medical therapy produced favorable acute and chronic hemodynamic effects. Larger trials are ongoing in China and Australia and are registered to begin in the United States (ZS-01-210, www.ClinicalTrials.gov). The phase I studies in patients with heart failure using a full-length glycosylated GGF2 are ongoing (GGF2-1101-1, www.ClinicalTrials.gov).
It seems unlikely that the improvement in cardiac function in response to recombinant NRG treatment is because of a direct inotropic effect. In fact, it seems that recombinant NRG-1β suppresses the increased fractional shortening induced by β-adrenergic receptor activation.35,37 There are effects of NRG-1β on calcium transients that may improve myocyte relaxation.33,85 Acute inodilator effects of rhNRG-1β have been observed,84 although it seems more likely that any long-term beneficial effects of rhNRG-1β are occurring through improvements in myocardial structure and function.
Preclinical data support exploration of rhNRG-1β in other areas of cardiovascular medicine (Figure 4). In addition to reducing the magnitude of ischemic heart and brain injury, there are effects of NRG-1β on vascular ErbB receptor expression after carotid balloon injury. Intravenous NRG-1β reduces neointimal hyperplasia after injury and prevents mitogen-stimulated vascular smooth muscle cell proliferation and migration.86 In human samples, NRG-1β expression was amplified in atherosclerotic lesions in coronary artery disease87 and inhibits atherogenesis via suppression of macrophage foam cell formation.88
Figure 4. Potential therapeutic effects of augmented neuregulin-1 signaling.
Augmentation of NRG-1/ErbB signaling has been explored as potential therapy in several cardiovascular pathological conditions (see text for details).
As rhNRG-1β moves forward as a potential therapeutic for heart failure, it will be important to consider the potential for adverse effects of systemic rhNRG-1β. One obvious concern is its potential to stimulate tumor growth. However, oncological studies have demonstrated that NRG-1 is both a tumor growth factor and a tumor suppressor, preventing generalization of what will be the effects of rhNRG-1β on tumor growth.89,90 Also concerning is that NRG/ErbB signaling has been implicated in the pathogenesis of schizophrenia, and behavioral effects of augmented NRG activity in the brain have been observed in mice.91 Efforts to localize delivery to specific target tissues and minimizing duration of exposure may help in this regard.
Role of NRG as a Potential Biomarker
As discussed, NRG-1β is activated in skeletal muscle by exercise50 and in the heart by injury27 and by changes in hemodynamic load.25 These findings have prompted measurements of NRG-1β in the serum and plasma. In healthy subjects, NRG-1β levels correlate with fitness.70 In contrast, levels correlate with disease prognosis in the subjects with chronic heart failure.92 In this latter study, higher-serum NRG-1β levels were particularly associated with poorer prognosis in ischemic heart disease.92 In patients with stable coronary disease without ischemia or heart failure, NRG-1β levels were inversely associated with coronary arterial disease severity.93 Interestingly, NRG-1β levels were higher in patients with stress-induced ischemia. These results are consistent with the concept that myocardial NRG-1β is activated in response to ischemia as observed in the isolated heart.27 The possibility that blood NRG-1β levels may have use as a clinical indicator of health and cardiovascular disease warrants further investigation.
Conclusion
Although a great deal has been learned about the role of NRG-1 and ErbB receptors in the cardiovascular system, there are many important unanswered questions. Knowledge in this field has been stimulated by effects of ErbB2-targeted therapies on the heart, demonstrating the continued importance of interdisciplinary research efforts in both basic and clinical research. Development of recombinant NRGs or small-molecule activators of ErbB receptor signaling in the cardiovascular system as a possible therapy for heart failure is ongoing. However, the mechanisms by which ErbB activation improves cardiac function are not clear and warrant further investigation. The question of whether cardiac myocyte proliferation in response to NRG treatment plays any role in the improved cardiac function with NRG treatment remains to be answered. There seem to be many determinants of myocardial ErbB receptor expression and, therefore, NRG-responsiveness. Understanding these will enhance the likelihood that NRG development as a therapy for heart failure will succeed. Furthermore, the potential for adverse effects of recombinant ErbB activators should prompt continued interdisciplinary team approaches. Our understanding of what regulates the expression and activity of endogenous myocardial NRGs is rudimentary at best. Perhaps, with further investigation, we will discover strategies to augment these signals in a clinically useful way.
Acknowledgments
The authors thank Katie Ris-Vicari, who created the figures according to the authors’ specifications, and Christopher Wells, MD, PhD for critical reading of the article.
Sources of Funding
This work was funded by P20 HL101425 and R01 HL068144 (to D.B.S.), R01 HL089385-02 (to M.F.H.), and 3R01HL089385-02S1 (to O.O.) from the National Heart, Lung, and Blood Institute.
Non-standard Abbreviations and Acronyms
- Akt
protein kinase B
- EGF
epidermal growth factor
- ErbB
erythroblastic leukemia viral oncogene homolog
- GGF2
glial growth factor 2
- NRG-1
neuregulin-1
- PI3K
phosphatidylinositol-3-kinase
- rhNRG-1
recombinant human neuregulin-1
Footnotes
Disclosures
D.B. Sawyer has received grant support from and has consulted for Acorda Therapeutics, Inc, a developer of the recombinant human neuregulin-1β glial growth factor 2 for cardiovascular and neurological disorders. The other authors have no conflicts to report.
References
- 1.Marchionni MA, Goodearl AD, Chen MS, Bermingham-McDonogh O, Kirk C, Hendricks M, Danehy F, Misumi D, Sudhalter J, Kobayashi K. Glial growth factors are alternatively spliced erbB2 ligands expressed in the nervous system. Nature. 1993;362:312–318. doi: 10.1038/362312a0. [DOI] [PubMed] [Google Scholar]
- 2.Meyer D, Birchmeier C. Multiple essential functions of neuregulin in development. Nature. 1995;378:386–390. doi: 10.1038/378386a0. [DOI] [PubMed] [Google Scholar]
- 3.Falls DL. Neuregulins: functions, forms, and signaling strategies. Exp Cell Res. 2003;284:14–30. doi: 10.1016/s0014-4827(02)00102-7. [DOI] [PubMed] [Google Scholar]
- 4.Fuller SJ, Sivarajah K, Sugden PH. ErbB receptors, their ligands, and the consequences of their activation and inhibition in the myocardium. J Mol Cell Cardiol. 2008;44:831–854. doi: 10.1016/j.yjmcc.2008.02.278. [DOI] [PubMed] [Google Scholar]
- 5.Montero JC, Yuste L, Díaz-Rodríguez E, Esparís-Ogando A, Pandiella A. Differential shedding of transmembrane neuregulin isoforms by the tumor necrosis factor-alpha-converting enzyme. Mol Cell Neurosci. 2000;16:631–648. doi: 10.1006/mcne.2000.0896. [DOI] [PubMed] [Google Scholar]
- 6.Hu X, Hicks CW, He W, Wong P, Macklin WB, Trapp BD, Yan R. Bace1 modulates myelination in the central and peripheral nervous system. Nat Neurosci. 2006;9:1520–1525. doi: 10.1038/nn1797. [DOI] [PubMed] [Google Scholar]
- 7.Yokozeki T, Wakatsuki S, Hatsuzawa K, Black RA, Wada I, Sehara-Fujisawa A. Meltrin beta (ADAM19) mediates ectodomain shedding of Neuregulin beta1 in the Golgi apparatus: fluorescence correlation spectroscopic observation of the dynamics of ectodomain shedding in living cells. Genes Cells. 2007;12:329–343. doi: 10.1111/j.1365-2443.2007.01060.x. [DOI] [PubMed] [Google Scholar]
- 8.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–137. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
- 9.Gassmann M, Casagranda F, Orioli D, Simon H, Lai C, Klein R, Lemke G. Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor. Nature. 1995;378:390–394. doi: 10.1038/378390a0. [DOI] [PubMed] [Google Scholar]
- 10.Lee KF, Simon H, Chen H, Bates B, Hung MC, Hauser C. Requirement for neuregulin receptor erbB2 in neural and cardiac development. Nature. 1995;378:394–398. doi: 10.1038/378394a0. [DOI] [PubMed] [Google Scholar]
- 11.Chan R, Hardy WR, Laing MA, Hardy SE, Muller WJ. The catalytic activity of the ErbB-2 receptor tyrosine kinase is essential for embryonic development. Mol Cell Biol. 2002;22:1073–1078. doi: 10.1128/MCB.22.4.1073-1078.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Erickson SL, O’Shea KS, Ghaboosi N, Loverro L, Frantz G, Bauer M, Lu LH, Moore MW. ErbB3 is required for normal cerebellar and cardiac development: a comparison with ErbB2-and heregulin-deficient mice. Development. 1997;124:4999–5011. doi: 10.1242/dev.124.24.4999. [DOI] [PubMed] [Google Scholar]
- 13.Zhao YY, Sawyer DR, Baliga RR, Opel DJ, Han X, Marchionni MA, Kelly RA. Neuregulins promote survival and growth of cardiac myocytes. Persistence of ErbB2 and ErbB4 expression in neonatal and adult ventricular myocytes. J Biol Chem. 1998;273:10261–10269. doi: 10.1074/jbc.273.17.10261. [DOI] [PubMed] [Google Scholar]
- 14.Campreciós G, Lorita J, Pardina E, Peinado-Onsurbe J, Soley M, Ramírez I. Expression, localization, and regulation of the neuregulin receptor ErbB3 in mouse heart. J Cell Physiol. 2011;226:450–455. doi: 10.1002/jcp.22354. [DOI] [PubMed] [Google Scholar]
- 15.Liu X, Hwang H, Cao L, Buckland M, Cunningham A, Chen J, Chien KR, Graham RM, Zhou M. Domain-specific gene disruption reveals critical regulation of neuregulin signaling by its cytoplasmic tail. Proc Natl Acad Sci USA. 1998;95:13024–13029. doi: 10.1073/pnas.95.22.13024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kramer R, Bucay N, Kane DJ, Martin LE, Tarpley JE, Theill LE. Neuregulins with an Ig-like domain are essential for mouse myocardial and neuronal development. Proc Natl Acad Sci USA. 1996;93:4833–4838. doi: 10.1073/pnas.93.10.4833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kim HS, Cho JW, Hidaka K, Morisaki T. Activation of MEK-ERK by heregulin-beta1 promotes the development of cardiomyocytes derived from ES cells. Biochem Biophys Res Commun. 2007;361:732–738. doi: 10.1016/j.bbrc.2007.07.045. [DOI] [PubMed] [Google Scholar]
- 18.Wang Z, Xu G, Wu Y, Guan Y, Cui L, Lei X, Zhang J, Mou L, Sun B, Dai Q. Neuregulin-1 enhances differentiation of cardiomyocytes from embryonic stem cells. Med Biol Eng Comput. 2009;47:41–48. doi: 10.1007/s11517-008-0383-2. [DOI] [PubMed] [Google Scholar]
- 19.Lai D, Liu X, Forrai A, Wolstein O, Michalicek J, Ahmed I, Garratt AN, Birchmeier C, Zhou M, Hartley L, Robb L, Feneley MP, Fatkin D, Harvey RP. Neuregulin 1 sustains the gene regulatory network in both trabecular and nontrabecular myocardium. Circ Res. 2010;107:715–727. doi: 10.1161/CIRCRESAHA.110.218693. [DOI] [PubMed] [Google Scholar]
- 20.Rentschler S, Zander J, Meyers K, France D, Levine R, Porter G, Rivkees SA, Morley GE, Fishman GI. Neuregulin-1 promotes formation of the murine cardiac conduction system. Proc Natl Acad Sci USA. 2002;99:10464–10469. doi: 10.1073/pnas.162301699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
- 22.Ozcelik C, Erdmann B, Pilz B, Wettschureck N, Britsch S, Hübner N, Chien KR, Birchmeier C, Garratt AN. Conditional mutation of the ErbB2 (HER2) receptor in cardiomyocytes leads to dilated cardiomyopathy. Proc Natl Acad Sci USA. 2002;99:8880–8885. doi: 10.1073/pnas.122249299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Crone SA, Zhao YY, Fan L, Gu Y, Minamisawa S, Liu Y, Peterson KL, Chen J, Kahn R, Condorelli G, Ross J, Jr, Chien KR, Lee KF. ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat Med. 2002;8:459–465. doi: 10.1038/nm0502-459. [DOI] [PubMed] [Google Scholar]
- 24.García-Rivello H, Taranda J, Said M, Cabeza-Meckert P, Vila-Petroff M, Scaglione J, Ghio S, Chen J, Lai C, Laguens RP, Lloyd KC, Hertig CM. Dilated cardiomyopathy in Erb-b4-deficient ventricular muscle. Am J Physiol Heart Circ Physiol. 2005;289:H1153–H1160. doi: 10.1152/ajpheart.00048.2005. [DOI] [PubMed] [Google Scholar]
- 25.Lemmens K, Doggen K, De Keulenaer GW. Activation of the neuregulin/ErbB system during physiological ventricular remodeling in pregnancy. Am J Physiol Heart Circ Physiol. 2011;300:H931–H942. doi: 10.1152/ajpheart.00385.2010. [DOI] [PubMed] [Google Scholar]
- 26.Cote GM, Miller TA, Lebrasseur NK, Kuramochi Y, Sawyer DB. Neuregulin-1alpha and beta isoform expression in cardiac microvascular endothelial cells and function in cardiac myocytes in vitro. Exp Cell Res. 2005;311:135–146. doi: 10.1016/j.yexcr.2005.08.017. [DOI] [PubMed] [Google Scholar]
- 27.Kuramochi Y, Cote GM, Guo X, Lebrasseur NK, Cui L, Liao R, Sawyer DB. Cardiac endothelial cells regulate reactive oxygen species-induced cardiomyocyte apoptosis through neuregulin-1beta/erbB4 signaling. J Biol Chem. 2004;279:51141–51147. doi: 10.1074/jbc.M408662200. [DOI] [PubMed] [Google Scholar]
- 28.Lemmens K, Segers VF, Demolder M, De Keulenaer GW. Role of neuregulin-1/ErbB2 signaling in endothelium-cardiomyocyte cross-talk. J Biol Chem. 2006;281:19469–19477. doi: 10.1074/jbc.M600399200. [DOI] [PubMed] [Google Scholar]
- 29.Fukazawa R, Miller TA, Kuramochi Y, Frantz S, Kim YD, Marchionni MA, Kelly RA, Sawyer DB. Neuregulin-1 protects ventricular myocytes from anthracycline-induced apoptosis via erbB4-dependent activation of PI3-kinase/Akt. J Mol Cell Cardiol. 2003;35:1473–1479. doi: 10.1016/j.yjmcc.2003.09.012. [DOI] [PubMed] [Google Scholar]
- 30.Grazette LP, Boecker W, Matsui T, Semigran M, Force TL, Hajjar RJ, Rosenzweig A. Inhibition of ErbB2 causes mitochondrial dysfunction in cardiomyocytes: implications for herceptin-induced cardiomyopathy. J Am Coll Cardiol. 2004;44:2231–2238. doi: 10.1016/j.jacc.2004.08.066. [DOI] [PubMed] [Google Scholar]
- 31.Bersell K, Arab S, Haring B, Kühn B. Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell. 2009;138:257–270. doi: 10.1016/j.cell.2009.04.060. [DOI] [PubMed] [Google Scholar]
- 32.Baliga RR, Pimental DR, Zhao YY, Simmons WW, Marchionni MA, Sawyer DB, Kelly RA. NRG-1-induced cardiomyocyte hypertrophy. Role of PI-3-kinase, p70(S6K), and MEK-MAPK-RSK. Am J Physiol. 1999;277:H2026–H2037. doi: 10.1152/ajpheart.1999.277.5.H2026. [DOI] [PubMed] [Google Scholar]
- 33.Brero A, Ramella R, Fitou A, Dati C, Alloatti G, Gallo MP, Levi R. Neuregulin-1beta1 rapidly modulates nitric oxide synthesis and calcium handling in rat cardiomyocytes. Cardiovasc Res. 2010;88:443–452. doi: 10.1093/cvr/cvq238. [DOI] [PubMed] [Google Scholar]
- 34.Kuramochi Y, Guo X, Sawyer DB. Neuregulin activates erbB2-dependnet src/FAK signaling and cytoskeletal remodeling in isolated adult rat cardiac myocytes. J Mol Cell Cardiol. 2006;41:228–235. doi: 10.1016/j.yjmcc.2006.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lemmens K, Fransen P, Sys SU, Brutsaert DL, De Keulenaer GW. Neuregulin-1 induces a negative inotropic effect in cardiac muscle: role of nitric oxide synthase. Circulation. 2004;109:324–326. doi: 10.1161/01.CIR.0000114521.88547.5E. [DOI] [PubMed] [Google Scholar]
- 36.Kuramochi Y, Lim CC, Guo X, Colucci WS, Liao R, Sawyer DB. Myocyte contractile activity modulates norepinephrine cytotoxicity and survival effects of neuregulin-1beta. Am J Physiol, Cell Physiol. 2004;286:C222–C229. doi: 10.1152/ajpcell.00312.2003. [DOI] [PubMed] [Google Scholar]
- 37.Okoshi K, Nakayama M, Yan X, Okoshi MP, Schuldt AJ, Marchionni MA, Lorell BH. Neuregulins regulate cardiac parasympathetic activity: muscarinic modulation of beta-adrenergic activity in myocytes from mice with neuregulin-1 gene deletion. Circulation. 2004;110:713–717. doi: 10.1161/01.CIR.0000138109.32748.80. [DOI] [PubMed] [Google Scholar]
- 38.Sawyer DB, Zuppinger C, Miller TA, Eppenberger HM, Suter TM. Modulation of anthracycline-induced myofibrillar disarray in rat ventricular myocytes by neuregulin-1beta and anti-erbB2: potential mechanism for trastuzumab-induced cardiotoxicity. Circulation. 2002;105:1551–1554. doi: 10.1161/01.cir.0000013839.41224.1c. [DOI] [PubMed] [Google Scholar]
- 39.Miller TA, Icli B, Cote GM, Lebrasseur NK, Borkan SC, Pimentel DR, Peng X, Sawyer DB. Palmitate alters neuregulin signaling and biology in cardiac myocytes. Biochem Biophys Res Commun. 2009;379:32–37. doi: 10.1016/j.bbrc.2008.11.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chiosis G, Timaul MN, Lucas B, Munster PN, Zheng FF, Sepp-Lorenzino L, Rosen N. A small molecule designed to bind to the adenine nucleotide pocket of Hsp90 causes Her2 degradation and the growth arrest and differentiation of breast cancer cells. Chem Biol. 2001;8:289–299. doi: 10.1016/s1074-5521(01)00015-1. [DOI] [PubMed] [Google Scholar]
- 41.Peng X, Guo X, Borkan SC, Bharti A, Kuramochi Y, Calderwood S, Sawyer DB. Heat shock protein 90 stabilization of ErbB2 expression is disrupted by ATP depletion in myocytes. J Biol Chem. 2005;280:13148–13152. doi: 10.1074/jbc.M410838200. [DOI] [PubMed] [Google Scholar]
- 42.Liu X, Gu X, Li Z, Li X, Li H, Chang J, Chen P, Jin J, Xi B, Chen D, Lai D, Graham RM, Zhou M. Neuregulin-1/erbB-activation improves cardiac function and survival in models of ischemic, dilated, and viral cardiomyopathy. J Am Coll Cardiol. 2006;48:1438–1447. doi: 10.1016/j.jacc.2006.05.057. [DOI] [PubMed] [Google Scholar]
- 43.Russell KS, Stern DF, Polverini PJ, Bender JR. Neuregulin activation of ErbB receptors in vascular endothelium leads to angiogenesis. Am J Physiol. 1999;277:H2205–H2211. doi: 10.1152/ajpheart.1999.277.6.H2205. [DOI] [PubMed] [Google Scholar]
- 44.Bagheri-Yarmand R, Vadlamudi RK, Wang RA, Mendelsohn J, Kumar R. Vascular endothelial growth factor up-regulation via p21-activated kinase-1 signaling regulates heregulin-beta1-mediated angiogenesis. J Biol Chem. 2000;275:39451–39457. doi: 10.1074/jbc.M006150200. [DOI] [PubMed] [Google Scholar]
- 45.Shin HS, Lee HJ, Nishida M, Lee MS, Tamura R, Yamashita S, Matsuzawa Y, Lee IK, Koh GY. Betacellulin and amphiregulin induce upregulation of cyclin D1 and DNA synthesis activity through differential signaling pathways in vascular smooth muscle cells. Circ Res. 2003;93:302–310. doi: 10.1161/01.RES.0000086803.64109.9E. [DOI] [PubMed] [Google Scholar]
- 46.Kim HS, Shin HS, Kwak HJ, Cho CH, Lee CO, Koh GY. Betacellulin induces angiogenesis through activation of mitogen-activated protein kinase and phosphatidylinositol 3′-kinase in endothelial cell. FASEB J. 2003;17:318–320. doi: 10.1096/fj.02-0570fje. [DOI] [PubMed] [Google Scholar]
- 47.Lange T, Nentwich MF, Lüth M, Yekebas E, Schumacher U. Trastuzumab has anti-metastatic and anti-angiogenic activity in a spontaneous metastasis xenograft model of esophageal adenocarcinoma. Cancer Lett. 2011;308:54–61. doi: 10.1016/j.canlet.2011.04.013. [DOI] [PubMed] [Google Scholar]
- 48.Hedhli N, Dobrucki LW, Kalinowski A, Zhuang ZW, Wu X, Russell RR, 3rd, Sinusas AJ, Russell KS. Endothelial-derived neuregulin is an important mediator of ischaemia-induced angiogenesis and arteriogenesis. Cardiovasc Res. 2012;93:516–524. doi: 10.1093/cvr/cvr352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ii M, Nishimura H, Iwakura A, Wecker A, Eaton E, Asahara T, Losordo DW. Endothelial progenitor cells are rapidly recruited to myocardium and mediate protective effect of ischemic preconditioning via “imported” nitric oxide synthase activity. Circulation. 2005;111:1114–1120. doi: 10.1161/01.CIR.0000157144.24888.7E. [DOI] [PubMed] [Google Scholar]
- 50.Lebrasseur NK, Coté GM, Miller TA, Fielding RA, Sawyer DB. Regulation of neuregulin/ErbB signaling by contractile activity in skeletal muscle. Am J Physiol, Cell Physiol. 2003;284:C1149–C1155. doi: 10.1152/ajpcell.00487.2002. [DOI] [PubMed] [Google Scholar]
- 51.Rehman J, Li J, Parvathaneni L, Karlsson G, Panchal VR, Temm CJ, Mahenthiran J, March KL. Exercise acutely increases circulating endothelial progenitor cells and monocyte-/macrophage-derived angiogenic MA, cells. J Am Coll Cardiol. 2004;43:2314–2318. doi: 10.1016/j.jacc.2004.02.049. [DOI] [PubMed] [Google Scholar]
- 52.Safa RN, Peng XY, Pentassuglia L, Lim CC, Lamparter M, Silverstein C, Walker J, Chen B, Geisberg C, Hatzopoulos AK, Sawyer DB. Neuregulin-1β regulation of embryonic endothelial progenitor cell survival. Am J Physiol Heart Circ Physiol. 2011;300:H1311–H1319. doi: 10.1152/ajpheart.01104.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Buonanno A, Fischbach GD. Neuregulin and ErbB receptor signaling pathways in the nervous system. Curr Opin Neurobiol. 2001;11:287–296. doi: 10.1016/s0959-4388(00)00210-5. [DOI] [PubMed] [Google Scholar]
- 54.Thompson M, Lauderdale S, Webster MJ, Chong VZ, McClintock B, Saunders R, Weickert CS. Widespread expression of ErbB2, ErbB3 and ErbB4 in non-human primate brain. Brain Res. 2007;1139:95–109. doi: 10.1016/j.brainres.2006.11.047. [DOI] [PubMed] [Google Scholar]
- 55.Xu Z, Ford GD, Croslan DR, Jiang J, Gates A, Allen R, Ford BD. Neuroprotection by neuregulin-1 following focal stroke is associated with the attenuation of ischemia-induced pro-inflammatory and stress DB. gene expression. Neurobiol Dis. 2005;19:461–470. doi: 10.1016/j.nbd.2005.01.027. [DOI] [PubMed] [Google Scholar]
- 56.Li Y, Xu Z, Ford GD, Croslan DR, Cairobe T, Li Z, Ford BD. Neuroprotection by neuregulin-1 in a rat model of permanent focal cerebral ischemia. Brain Res. 2007;1184:277–283. doi: 10.1016/j.brainres.2007.09.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Iaci JF, Ganguly A, Finklestein SP, Parry TJ, Ren J, Saha S, Sietsma DK, Srinivas M, Vecchione AM, Caggiano AO. Glial growth factor 2 promotes functional recovery with treatment initiated up to 7 days after permanent focal ischemic stroke. Neuropharmacology. 2010;59:TM. 640–649. doi: 10.1016/j.neuropharm.2010.07.017. [DOI] [PubMed] [Google Scholar]
- 58.Tokita Y, Keino H, Matsui F, Aono S, Ishiguro H, Higashiyama S, Oohira A. Regulation of neuregulin expression in the injured rat brain and cultured astrocytes. J Neurosci. 2001;21:1257–1264. doi: 10.1523/JNEUROSCI.21-04-01257.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lok J, Wang H, Murata Y, Zhu HH, Qin T, Whalen MJ, Lo EH. Effect of neuregulin-1 on histopathological and functional outcome after controlled cortical impact in mice. J Neurotrauma. 2007;24:1817–1822. doi: 10.1089/neu.2007.0372. [DOI] [PubMed] [Google Scholar]
- 60.Lok J, Sardi SP, Guo S, Besancon E, Ha DM, Rosell A, Kim WJ, Corfas G, Lo EH. Neuregulin-1 signaling in brain endothelial cells. J Cereb Blood Flow Metab. 2009;29:39–43. doi: 10.1038/jcbfm.2008.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Matsukawa R, Hirooka Y, Nishihara M, Ito K, Sunagawa K. Neuregulin-1/ErbB signaling in rostral ventrolateral medulla is involved in blood pressure regulation as an antihypertensive system. J Hypertens. 2011;29:1735–1742. doi: 10.1097/HJH.0b013e32834937d6. [DOI] [PubMed] [Google Scholar]
- 62.Florini JR, Samuel DS, Ewton DZ, Kirk C, Sklar RM. Stimulation of myogenic differentiation by a neuregulin, glial growth factor 2. Are neuregulins the long-sought muscle trophic factors secreted by nerves? J Biol Chem. 1996;271:12699–12702. doi: 10.1074/jbc.271.22.12699. [DOI] [PubMed] [Google Scholar]
- 63.Kim D, Chi S, Lee KH, Rhee S, Kwon YK, Chung CH, Kwon H, Kang MS. Neuregulin stimulates myogenic differentiation in an autocrine manner. J Biol Chem. 1999;274:15395–15400. doi: 10.1074/jbc.274.22.15395. [DOI] [PubMed] [Google Scholar]
- 64.Martinou JC, Falls DL, Fischbach GD, Merlie JP. Acetylcholine receptor-inducing activity stimulates expression of the epsilon-subunit gene of the muscle acetylcholine receptor. Proc Natl Acad Sci USA. 1991;88:7669–7673. doi: 10.1073/pnas.88.17.7669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tansey MG, Chu GC, Merlie JP. ARIA/HRG regulates AChR epsilon subunit gene expression at the neuromuscular synapse via activation of phosphatidylinositol 3-kinase and Ras/MAPK pathway. J Cell Biol. 1996;134:465–476. doi: 10.1083/jcb.134.2.465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Si J, Luo Z, Mei L. Induction of acetylcholine receptor gene expression by ARIA requires activation of mitogen-activated protein kinase. J Biol Chem. 1996;271:19752–19759. doi: 10.1074/jbc.271.33.19752. [DOI] [PubMed] [Google Scholar]
- 67.Andrechek ER, Hardy WR, Girgis-Gabardo AA, Perry RL, Butler R, Graham FL, Kahn RC, Rudnicki MA, Muller WJ. ErbB2 is required for muscle spindle and myoblast cell survival. Mol Cell Biol. 2002;22:4714–4722. doi: 10.1128/MCB.22.13.4714-4722.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Suárez E, Bach D, Cadefau J, Palacin M, Zorzano A, Gumá A. A novel role of neuregulin in skeletal muscle. Neuregulin stimulates glucose uptake, glucose transporter translocation, and transporter expression in muscle cells. J Biol Chem. 2001;276:18257–18264. doi: 10.1074/jbc.M008100200. [DOI] [PubMed] [Google Scholar]
- 69.LeBrasseur NK, Mizer KC, Parkington JD, Sawyer DB, Fielding RA. The expression of neuregulin and erbB receptors in human skeletal muscle: effects of progressive resistance training. Eur J Appl Physiol. 2005;94:371–375. doi: 10.1007/s00421-005-1333-4. [DOI] [PubMed] [Google Scholar]
- 70.Moondra V, Sarma S, Buxton T, Safa R, Cote G, Storer T, Lebrasseur NK, Sawyer DB. Serum Neuregulin-1beta as a Biomarker of Cardiovascular Fitness. Open Biomark J. 2009;2:1–5. doi: 10.2174/1875318300902010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Rohrbach S, Yan X, Weinberg EO, Hasan F, Bartunek J, Marchionni MA, Lorell BH. Neuregulin in cardiac hypertrophy in rats with aortic stenosis. Differential expression of erbB2 and erbB4 receptors. Circulation. 1999;100:407–412. doi: 10.1161/01.cir.100.4.407. [DOI] [PubMed] [Google Scholar]
- 72.Gui C, Zhu L, Hu M, Lei L, Long Q. Neuregulin-1/ErbB signaling is impaired in the rat model of diabetic cardiomyopathy. Cardiovasc Pathol. 2012;21:414–420. doi: 10.1016/j.carpath.2011.12.006. [DOI] [PubMed] [Google Scholar]
- 73.Rohrbach S, Niemann B, Silber RE, Holtz J. Neuregulin receptors erbB2 and erbB4 in failing human myocardium—depressed expression and attenuated activation. Basic Res Cardiol. 2005;100:240–249. doi: 10.1007/s00395-005-0514-4. [DOI] [PubMed] [Google Scholar]
- 74.Uray IP, Connelly JH, Thomázy V, Shipley GL, Vaughn WK, Frazier OH, Taegtmeyer H, Davies PJ. Left ventricular unloading alters receptor tyrosine kinase expression in the failing human heart. J Heart Lung Transplant. 2002;21:771–782. doi: 10.1016/s1053-2498(02)00390-x. [DOI] [PubMed] [Google Scholar]
- 75.Hedhli N, Huang Q, Kalinowski A, Palmeri M, Hu X, Russell RR, Russell KS. Endothelium-derived neuregulin protects the heart against ischemic injury. Circulation. 2011;123:2254–2262. doi: 10.1161/CIRCULATIONAHA.110.991125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Pentassuglia L, Graf M, Lane H, Kuramochi Y, Cote G, Timolati F, Sawyer DB, Zuppinger C, Suter TM. Inhibition of ErbB2 by receptor tyrosine kinase inhibitors causes myofibrillar structural damage without cell death in adult rat cardiomyocytes. Exp Cell Res. 2009;315:1302–1312. doi: 10.1016/j.yexcr.2009.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Fang SJ, Wu XS, Han ZH, Zhang XX, Wang CM, Li XY, Lu LQ, Zhank JL. Neuregulin-1 preconditioning protects the heart against ischemia/reperfusion injury through a PI3K/Akt-dependent mechanism. Chin Med J. 2010;123:3597–3604. [PubMed] [Google Scholar]
- 78.Li J, Gu XH, Duan JC, Zeng L, Li Y, Wang L. Effects of recombined human neuregulin on the contractibility of cardiac muscles of rhesus monkeys with pacing-induced heart failure. Sichuan Da Xue Xue Bao Yi Xue Ban. 2007;38:105–108. [PubMed] [Google Scholar]
- 79.Ganguly A, Iaci JF, Srinivas M, Vecchione AV, Hackett CS, Kasperbauer S, Caggiano AO. Neuregulin treatment of heart failure in rat. Circ Res. 2008;103:Abstract e51. [Google Scholar]
- 80.Hill MF, Patel AV, Smith HM, Pentassuglia L, Peng X, Srinivas M, Ganguly A, Iaci JF, Parry TJ, Caggiano AO, Sawyer DB. Neuregulin GGF2 retains cardioprotective effects in the presence of high fat diet in post-myocardial infarcted rats. Circulation. 2010;122:Abstract 20699. [Google Scholar]
- 81.Kasasbeh E, Murphy A, Ahmad FA, Yu E, Williams P, Nunnally A, Adcock J, Caggiano AO, Parry T, Ganguly A, Sawyer DB, Cleator JH. Neuregulin-1β improves cardiac remodeling after myocardial infarction in swine. Circulation. 2011;124:Abstract 15531. [Google Scholar]
- 82.Guo YF, Zhang XX, Liu Y, Duan HY, Jie BZ, Wu XS. Neuregulin-1 atenuates mitochondrial dysfunction in a rat model of heart failure. Chin Med J. 2012;125:807–814. [PubMed] [Google Scholar]
- 83.Gao R, Zhang J, Cheng L, Wu X, Dong W, Yang X, Li T, Liu X, Xu Y, Li X, Zhou M. A Phase II, randomized, double-blind, multicenter, based on standard therapy, placebo-controlled study of the efficacy and safety of recombinant human neuregulin-1 in patients with chronic heart failure. J Am Coll Cardiol. 2010;55:1907–1914. doi: 10.1016/j.jacc.2009.12.044. [DOI] [PubMed] [Google Scholar]
- 84.Jabbour A, Hayward CS, Keogh AM, Kotlyar E, McCrohon JA, England JF, Amor R, Liu X, Li XY, Zhou MD, Graham RM, Macdonald PS. Parenteral administration of recombinant human neuregulin-1 to patients with stable chronic heart failure produces favourable acute and chronic haemodynamic responses. Eur J Heart Fail. 2011;13:83–92. doi: 10.1093/eurjhf/hfq152. [DOI] [PubMed] [Google Scholar]
- 85.Timolati F, Ott D, Pentassuglia L, Giraud MN, Perriard JC, Suter TM, Zuppinger C. Neuregulin-1 beta attenuates doxorubicin-induced alterations of excitation-contraction coupling and reduces oxidative stress in adult rat cardiomyocytes. J Mol Cell Cardiol. 2006;41:845–854. doi: 10.1016/j.yjmcc.2006.08.002. [DOI] [PubMed] [Google Scholar]
- 86.Clement CM, Thomas LK, Mou Y, Croslan DR, Gibbons GH, Ford BD. Neuregulin-1 attenuates neointimal formation following vascular injury and inhibits the proliferation of vascular smooth muscle cells. J Vasc Res. 2007;44:303–312. doi: 10.1159/000101776. [DOI] [PubMed] [Google Scholar]
- 87.Panutsopulos D, Arvanitis DL, Tsatsanis C, Papalambros E, Sigala F, Spandidos DA. Expression of heregulin in human coronary atherosclerotic lesions. J Vasc Res. 2005;42:463–474. doi: 10.1159/000088100. [DOI] [PubMed] [Google Scholar]
- 88.Xu G, Watanabe T, Iso Y, Koba S, Sakai T, Nagashima M, Arita S, Hongo S, Ota H, Kobayashi Y, Miyazaki A, Hirano T. Preventive effects of heregulin-beta1 on macrophage foam cell formation and atherosclerosis. Circ Res. 2009;105:500–510. doi: 10.1161/CIRCRESAHA.109.193870. [DOI] [PubMed] [Google Scholar]
- 89.Atlas E, Cardillo M, Mehmi I, Zahedkargaran H, Tang C, Lupu R. Heregulin is sufficient for the promotion of tumorigenicity and metastasis of breast cancer cells in vivo. Mol Cancer Res. 2003;1:165–175. [PubMed] [Google Scholar]
- 90.Chua YL, Ito Y, Pole JC, Newman S, Chin SF, Stein RC, Ellis IO, Caldas C, O’Hare MJ, Murrell A, Edwards PA. The NRG1 gene is frequently silenced by methylation in breast cancers and is a strong candidate for the 8p tumour suppressor gene. Oncogene. 2009;28:4041–4052. doi: 10.1038/onc.2009.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kato T, Kasai A, Mizuno M, Fengyi L, Shintani N, Maeda S, Yokoyama M, Ozaki M, Nawa H. Phenotypic characterization of transgenic mice overexpressing neuregulin-1. PLoS ONE. 2010;5:e14185. doi: 10.1371/journal.pone.0014185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ky B, Kimmel SE, Safa RN, Putt ME, Sweitzer NK, Fang JC, Sawyer DB, Cappola TP. Neuregulin-1 beta is associated with disease severity and adverse outcomes in chronic heart failure. Circulation. 2009;120:310–317. doi: 10.1161/CIRCULATIONAHA.109.856310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Geisberg CA, Wang G, Safa RN, Smith HM, Anderson B, Peng XY, Veerkamp B, Zhao DX, Blakemore D, Yu C, Sawyer DB. Circulating neuregulin-1β levels vary according to the angiographic severity of coronary artery disease and ischemia. Coron Artery Dis. 2011;22:577–582. doi: 10.1097/MCA.0b013e32834d3346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Marchionni MA, Cannella B, Hoban C, Gao YL, Garcia-Arenas R, Lawson D, Happel E, Noel F, Tofilon P, Gwynne D, Raine CS. Neuregulin in neuron/glial interactions in the central nervous system. GGF2 diminishes autoimmune demyelination, promotes oligodendrocyte progenitor expansion, and enhances remyelination. Adv Exp Med Biol. 1999;468:283–295. [PubMed] [Google Scholar]
- 95.Xu Z, Jiang J, Ford G, Ford BD. Neuregulin-1 is neuroprotective and attenuates inflammatory responses induced by ischemic stroke. Biochem M, Biophys Res Commun. 2004;322:440–446. doi: 10.1016/j.bbrc.2004.07.149. [DOI] [PubMed] [Google Scholar]
- 96.Cai J, Peng X, Nelson KD, Eberhart R, Smith GM. Synergistic improvements in cell and axonal migration across sciatic nerve lesion gaps using bioresorbable filaments and heregulin-beta1. J Biomed Mater Res A. 2004;69:247–258. doi: 10.1002/jbm.a.20119. [DOI] [PubMed] [Google Scholar]
- 97.Krag TO, Bogdanovich S, Jensen CJ, Fischer MD, Hansen-Schwartz J, Javazon EH, Flake AW, Edvinsson L, Khurana TS. Heregulin ameliorates the dystrophic phenotype in mdx mice. Proc Natl Acad Sci USA. 2004;101:13856–13860. doi: 10.1073/pnas.0405972101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gu X, Liu X, Xu D, Li X, Yan M, Qi Y, Yan W, Wang W, Pan J, Xu Y, Xi B, Cheng L, Jia J, Wang K, Ge J, Zhou M. Cardiac functional improvement in rats with myocardial infarction by up-regulating cardiac myosin light chain kinase with neuregulin. Cardiovasc Res. 2010;88:334–343. doi: 10.1093/cvr/cvq223. [DOI] [PubMed] [Google Scholar]
- 99.Bian Y, Sun M, Silver M, Ho KK, Marchionni MA, Caggiano AO, Stone JR, Amende I, Hampton TG, Morgan JP, Yan X. Neuregulin-1 attenuated doxorubicin-induced decrease in cardiac troponins. Am J Physiol Heart Circ Physiol. 2009;297:H1974–H1983. doi: 10.1152/ajpheart.01010.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]




