Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Oct 1.
Published in final edited form as: J Mol Cell Cardiol. 2011 Jun 28;51(4):501–505. doi: 10.1016/j.yjmcc.2011.06.016

Neuregulin-1β for the treatment of systolic heart failure

Douglas B Sawyer 1, Anthony Caggiano 1
PMCID: PMC3179893  NIHMSID: NIHMS312517  PMID: 21729703

Introduction

Systolic heart failure (HF) is a disease characterized by progressive cardiac dysfunction leading to increased morbidity and early mortality. The vast majority of pharmacologic strategies for HF developed over recent decades have focused on inhibiting processes that have been demonstrated to contribute to progression of myocardial remodeling. Unfortunately, drug therapies in HF do not produce “reverse remodeling” or improved systolic function via non-inotropic mechanisms in a uniform or predictable manner.

Biological therapeutics in the form of growth factors and cell therapies have been studied extensively in animal models of heart failure, and have been pursued as potential strategies to induce myocardial repair by design. Recombinant growth hormone (GH) was promising in preclinical models but did not show reproducible benefits in randomized clinical trials of HF [1, 2]. IGF-1 has similarly appeared promising in preclinical studies [3], but has not been pursued vigorously to clinical trials, perhaps due to the concern of IGF-1 mediated effects on other organs including tumor growth promotion.

In the last decade direct and indirect manipulation of cell populations to induce myocardial regeneration have been attempted, with mixed results as far as efficacy and improvement in LV function. Ongoing trials, including those funded by the National Heart Lung and Blood Institute (NHLBI) will help to address whether these strategies can be refined to provide consistent benefit. A consistent finding in preclinical models where cell therapies are being developed suggests that a major mechanism for the beneficial effects of cell-based therapies may be that cells provide a source of paracrine factors that help myocardial repair. Neuregulin-1β (Nrg-1β) is one of these paracrine factors with interesting biological properties, and potential therapeutic value.

Neuregulins – paracrine regulators of tissue form and function

Nrgs belong to a family of growth factors which are encoded by one of four known genes (Nrg-1 through -4) and act through receptor tyrosine kinases in the epidermal growth factor receptor family (EGFR). Each of the Nrg genes can be expressed as multiple distinct protein isoforms due to alternative splicing of the transcripts [4]. Nrg’s actions are mediated through a set of ErbB tyrosine kinase receptors (ErbB2, ErbB2, ErbB4) which stimulate cellular proliferation, differentiation, and survival of cells in many tissues including the heart. (Table 1).

Table 1.

Adult tissues where NRG has been implicated including possible therapeutics

Tissue Biological role Potential therapeutic Reference
CNS Myelination Multiple sclerosis [51]
Neuron protection & recovery Stroke [52, 53]
Development Schizophrenia [54, 55]
Heart Development and maintenance Heart Failure see text for references
Peripheral nerve Myelination Nerve injury [56] [57]
Skeletal muscle Synapse formation Nerve and muscle injury [58]
Myoblast proliferation Muscular dystrophy [59]
Glucose uptake Metabolic syndromes [60]
Proliferation & Differentiation Muscle injury [61, 62]
Wasting Syndromes

Since their initial identification Nrgs have been named according to their function in various tissues (e.g. skeletal muscle: ARIA – Acetylcholine Receptor Inducing Activity; Her2/neu expressing tumor cells: NDF – Neu Differentiation Factor; central nervous system: GGF - Glial Growth Factor). Nrgs are also named according to the splice variants, distinguishing alpha or beta variants of the EGF-like domain, as well as indicating the linker sequence before the transmembrane domain (e.g. the type II neuregulin GGF2 is also known as Nrg-1β3). Frequently in the literature one of the several recombinant EGF-like domains is used in isolation and referred to as Nrg-1 although the EGF-like domains are not expressed on their own in nature.

The first suggestion that Nrg-1 might have some role in the heart came from studies in mice genetically manipulated to remove functional Nrg-1 and its receptors. Mice with disrupted Nrg-1, ErbB 2, 3 and 4 receptors demonstrate the critical role of this signaling system in the development of ventricular trabeculation as well as the endocardial cushion required for formation of heart valves [57]. There are several interesting points that come from these studies. First is that while there are four Nrg genes, Nrg-2, 3 and 4 cannot supplant the requirement for Nrg-1 during cardiac development. It is also interesting that knockouts targeting specific exons of Nrg-1 demonstrate that a transmembrane Nrg-1β is required for the heart to develop beyond this critical stage [7, 8]. The ErbB2 and ErbB4 knockout mice give the same phenotype as the Nrg-1 knockout [5, 9]. Thus collectively it appears that Nrg-1β/ErbB2/ErbB4 represents a signaling cassette required for ventricular trabeculation during cardiac development. This work was quickly followed with studies of Nrg-1 and its receptors in the adult heart. Investigators found that this same cassette persists in the adult heart[10], where Nrg-1β is expressed in the microvascular endothelial cell and activates both ErbB2 and ErbB4 on cardiac myocytes, leading to downstream activation of kinase cascades that can be demonstrated to modulate myocyte cell biology including cell growth, survival, as well as glucose uptake (see Figure 1, Table 2). Microvascular endothelial cells isolated from adult rat heart express the mRNA of at least 11 isoforms [11]. Both Nrg-1α and Nrg-1β areexpressed, with the β isoforms being the most potent as activators of cardiac myocyte signaling and biological responses [11]. The majority of cardiac isoforms are expressed as transmembrane proteins that appear to require proteolytic activation, which has been observed in the setting of physiologic and pathologic stress [12, 13]. In some vascular beds Nrg-1 may have direct effects on endothelial cells inducing a proliferative response [14]. Thus cardiac Nrg/erbB signaling is a paracrine and juxtacrine system that regulates the interaction between microvascular endothelial cells and cardiac myocytes.

Figure 1.

Figure 1

Simplified schematic for actions of neuregulin and the erbB2 and erbB4 receptors in the heart where signaling are understood. Neuregulins are expressed in the adult heart in the microvascular endothelial cells, and act on adjacent myocytes by proteolytic release by a matrix metalloproteinase. Activation of erbB2 and erbB4 receptor tyrosine kinases leads to downstream activation of signals that regulate myocyte biology. While many of the details remain to be further elucidated, it appears that recombinant Nrg-1β induces improved cardiac function via one or more of these biological responses.

Table 2.

Biological effects of NRG in the heart

Cell type Response Signaling Pathway Reference
Cardiac Myocyte Survival, mitochondrial function PI3-kinase/Akt [10, 23, 63, 64]
Glucose uptake, SR Calcium uptake PI3-kinase/Akt [11, 40]
Growth MEK/Erk [65]
Focal adhesion formation Src/FAK [66]
Muscarinic Activation ? [38]
Cell Division ? [33]
Stem cell Myocyte differentiation ? [67]
Endothelial Cell Proliferation ? [14]
Conduction System Formation ? [68]
Endothelial Progenitor Cells survival PI3kinase/Akt [69]

The early interest in understanding the role of Nrg-1 and its receptors in the adult heart was augmented by the observation that women with breast cancer treated with the erbB2-targeted antibody trastuzumab have increased risk of developing cardiac dysfunction and heart failure [15]. This finding provided important evidence for a role for erbB2 in the adult human heart, and motivated a number of laboratories to explore the role of neuregulin and its receptors in the heart. Subsequent studies demonstrated that ErbB2 and ErbB4 receptor signaling are required for maintenance of myocardial function in the adult heart, as cardiac specific deletion of functional receptors in mice leads to a dilated cardiomyopathy phenotype [16, 17]. Moreover, mice with conditional ErbB2 deletion or heterologous Nrg-1 deficiency have increased susceptibility to anthracycline cardiotoxicity [17, 18]. In adult myocytes in culture, anthracyline-induced sarcopenia is exacerbated by exposure to antibodies against erbB2 and reduced by recombinant Nrg-1β [19]. These observations demonstrate the critical role of Nrg-1β/erbB signaling in the response of the heart to injury as well as the maintenance of normal myocardial structure and function.

Studies of Neuregulin/ErbB activity in human subjects

Using an enzyme-linked antibody assay, Nrg-1β has been detected in human serum. In healthy subjects levels of Nrg-1β correlate with fitness [20], which can be interpreted based upon animal work to be an indicator of physical activity, given that exercise is a potent activator of Nrg/erbB signaling in skeletal muscle [21]. In the setting of advanced heart failure, however, circulating Nrg-1β is elevated in proportion to disease severity, particularly in those with ischemic cardiomyopathy [22]. While there are several interpretations of this observation, one possibility comes from the finding in animals that myocardial Nrg-1β is released in response to ischemic injury [23] and neurohormonal activation [24]. This is supported by recent evidence for increased activity of Nrg-1β/erbB signaling in the myocardium of animal models of heart failure [25]. Receptor down-regulation, a common feedback mechanism in biology, is also observed in chronic heart failure [26, 27], suggesting that heart failure could be a state of reduced Nrg-1β responsiveness. Other evidence suggests that Nrg’s play an important role in vascular health. Nrg suppresses macrophage migration into atherosclerotic placques, and may be a negative regulator of atherosclerosis [28], perhaps accounding for the finding in the Cardiovascular Risk in Young Finns Study where a Nrg-1 polymorphism has been associated with atherosclerosis when coupled with stress [29].

Recombinant Neuregulin improves cardiac function in animals

Work with recombinant Nrg-1β in the setting of animal models of chronic heart failure supports pursuing this as a therapy for systolic heart failure [30]. In rats with cardiac dysfunction induced by coronary artery ligation, a series of intravenous doses of a small fragment of Nrg-1β (10 μg/kg, IV, daily for 5 days) improved cardiac function (reduced left ventricular end systolic dimensions, improved fractional shortening, and ejection fraction). Similar improvements in cardiac function were seen in models of anthracycline and virally induced cardiac injury. In the anthracycline model Nrg-1β was shown to act at least in part by protecting cardiac myocytes as evidenced by maintenance of cardiac troponin [31]. In dogs [30] and primates [32] with rapid-pacing induced heart failure Nrg-1β induced improved cardiac contractility and relaxation.

As discussed, there are many effects of Nrg-1β on the biology of cardiac myocytes and endothelial cells in the heart that could be contributing to the beneficial effects of Nrg-1β on cardiac function (Table 2). In addition to protecting myocytes from stimuli that lead to loss of myocytes and progressive myocardial dysfunction, there are effects of Nrg-1β on sarcomere gene expression, and sarcomere stability that implicate Nrg-1β as a ‘cardiac repair factor’, improving cardiac function by allowing for repairing stressed cardiac myocytes. Alternatively, Nrg-1β has also been reported to induce myocyte proliferation [33], implicating Nrg-1β as a ‘cardiac regeneration factor’. Whether Nrg-1β-induced cardiomyocyte re-entry into the cell cycle is mechanistic in the improvement in cardiac function remains unknown. Still other potential mechanisms of the therapeutic Nrg-1β action in animals are being defined, such as alterations in myocardial metabolism. Overall the data suggest that the effects of Nrg-1β on the heart in disease models are pleiotropic including protection of contractile proteins, regulation of energy utilization, promotion of cell survival as well as cell division.

As will be discussed below, to our knowledge there are two forms of Nrg-1β under development for the treatment of systolic heart failure. The ‘EGF-domain only’ fragment of Nrg-1β used by some investigators is not a natural product of the Nrg-1 gene, and lacks domains that are highly conserved in this family of proteins [4]. In particular, the Ig domain is expressed in Nrg-1 gene products, and is thought to mediate localization of ligand activity through interaction with matrix molecules. In in vitro assays of myocyte response to recombinant Nrg-1β, we find that small EGF-domain only forms of Nrg-1β have equal potency at activating intracellular signaling as the larger GGF2, but that the effects of GGF2 are longer-lasting [34]. In in vivo comparisons between these two peptides, GGF2 had more potent effects on improvement of cardiac function requiring less frequent dosing (Caggiano and colleagues, unpublished observation). Other studies have demonstrated that the Ig-like domain containing neuregulins are important for development and selective disruption of the Ig-like domain results in embryonic lethality with both cardiac and brain defects [35]. This may be explained in part by the differences in receptor binding and cellular signal transduction when comparing recombinant neuregulins with and without the Ig domains [36]. The potential relative benefits of the different Nrg-1β isoforms as heart failure therapies have not been completely explored.

While recombinant Nrg-1β improves cardiac systolic function after injury, there is no evidence that Nrg-1β has a direct inotropic effect, increasing myocardial contractility through changes in calcium handling, excitation-contraction coupling, or myofilament calcium sensitivity. There have been no acute or delayed effects of recombinant Nrg-1β on blood pressure or heart rate observed in numerous safety studies ([30]; our own unpublished data). In fact it appears that recombinant Nrg-1β suppresses the increased fractional shortening induced by beta-adrenergic receptor activation [37, 38]. However there are effects of Nrg-1β on calcium transients that suggest an effect on myocyte relaxation [39, 40]. Thus current data would suggest that Nrg-1β acts via alterations in myocardial structure, and not via acute changes in calcium handling or EC coupling.

Early studies of recombinant Neuregulin in humans

How these multiple actions will translate into a potential human therapeutic is beginning to be explored. One Nrg-1β fragment (a 61 amino acid EGF-domain only peptide common to all NRG-1β) has advanced to clinical studies in Australia and China and another Nrg-1β has begun early phase human trials in the United States. Preliminary results from a dose-ranging tolerability study with Nrg-1β EGF domain peptide were recently reported [41]. Patients with systolic heart failure were treated for ten days with this Nrg-1β, and after 90 days showed a trend towards improvement in cardiac structure and function as assessed by magnetic resonance imaging, at least at one dose studied. An analysis of acute hemodynamic effects of this peptide demonstrate that there is an acute increase in cardiac output that accompanies a vasodilator effect [42]. Of note, there was considerable nausea experienced by subjects receiving this peptide. Another issue that presents a challenge is the dosing schedule used in these trials, which has involved prolonged (multi-hour) infusion, daily, for multiple days. Larger Phase 2 and Phase 3 studies are ongoing in China and Australia [43, 44], and are registered but not yet activated in the United States [45]. The Phase 1 studies in patients with heart failure using a full-length glycosylated recombinant human Nrg-1β3 (also known as GGF2) have recently started [46]. Whether this longer acting Nrg-1β will have any advantages in terms of safety, tolerability, and ultimately efficacy over the EGF-domain only peptide remains to be determined.

Questions and Problems for future research

As Nrg-1β moves forward as a potential therapeutic for HF, it will be important to identify the mechanism for its action, and the extent to which systemic delivery of such a ligand with near ubiquitous receptor expression activates responses in other tissues. A concern in the development of Nrg-1β as a therapeutic is the possibility that it will stimulate growth of other tissue, including cancers [47]. Published data demonstrate the complexity of this issue with evidence of both the potential for tumor suppression as well as tumor growth acceleration[48, 49]. Another concern is that increased Nrg-1β activity may have adverse effects on the central nervous system, as Nrg-1β overexpression in the brain has been associated with behavioral changes in mice [50]. Thus it will be important to determine what dose/regimen is required to maintain any positive effects seen on cardiac function relative to the potential for risk of adverse effects. Ultimately the therapeutic benefit of Nrg-1β must be established in well-controlled and sufficiently powered human trials reviewed and approved by Food and Drug Administration.

It is interesting to consider the many factors that have helped propel Nrg-1β down the translational path. We consider three of these to be interesting biology, diverse opportunity, and inverse evidence. As highlighted in Table 2, interesting biology in the heart as well as other organs has attracted young scientists to this field, looking for worthwhile questions to investigate. There are still many important aspects of Nrg/erbB biology that remain to be answered; one can only hope that this is recognized by the many young scientists looking for areas to focus their efforts, and that funding agencies continue to support fundamental biological investigation. The diverse opportunities for developing Nrg’s as therapeutics, as highlighted in Table 1, has provided an obvious stimulus for industry investment in moving Nrg’s forward. Even if Nrg-1β does not develop into a successful heart failure therapy, the lessons learned will certainly inform development of Nrg’s for other disorders. Finally, progress in cancer therapeutics led to unexpected inverse evidence of the importance of erbB-signaling for maintenance of cardiac structure and function. This trastuzumab experience, and the collaborative investigations this stimulated focused on understanding the underlying mechanisms, have obviously helped to propel the development of erbB agonists as cardiovascular therapies forward. The rapid pace of oncology drug development is sure to create other such opportunities. Of course each of these factors has the attribute of stimulating interactions between people with different experiences and expertise, an excellent path to progress in any walk of life.

Highlights.

  • Neuregulin-1β(Nrg-1β) is a growth and survival factor in the heart and other organs.

  • Nrg-1β works in cardiac myocytes via erbB2 and erbB4 receptor tyrosine kinases.

  • Nrg-1β/erbB signaling in the heart regulates myocardial adaptation to stress.

  • Recombinant Nrg-1β improves heart function in animal models of heart failure (HF).

  • Recombinant human Nrg-1β is now being tested in humans as a possible HF therapy.

Acknowledgments

This work was supported by the NIH/NHLBI 1 P20 HL101425-01, R01 HL068144, the American Heart Association, and Acorda Therapeutics, Inc.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Jose VJ, Zechariah TU, George P, Jonathan V. Growth hormone therapy in patients with dilated cardiomyopathy: preliminary observations of a pilot study. Indian Heart J. 1999 Mar-Apr;51(2):183–5. [PubMed] [Google Scholar]
  • 2.Acevedo M, Corbalan R, Chamorro G, Jalil J, Nazzal C, Campusano C, et al. Administration of growth hormone to patients with advanced cardiac heart failure: effects upon left ventricular function, exercise capacity, and neurohormonal status. Int J Cardiol. 2003 Feb;87(2–3):185–91. doi: 10.1016/s0167-5273(02)00249-8. [DOI] [PubMed] [Google Scholar]
  • 3.Cittadini A, Stromer H, Katz SE, Clark R, Moses AC, Morgan JP, et al. Differential cardiac effects of growth hormone and insulin-like growth factor-1 in the rat. A combined in vivo and in vitro evaluation. Circulation. 1996 Feb 15;93(4):800–9. doi: 10.1161/01.cir.93.4.800. [DOI] [PubMed] [Google Scholar]
  • 4.Falls DL. Neuregulins: functions, forms, and signaling strategies. Exp Cell Res. 2003 Mar 10;284(1):14–30. doi: 10.1016/s0014-4827(02)00102-7. [DOI] [PubMed] [Google Scholar]
  • 5.Gassmann M, Casagranda F, Orioli D, Simon H, Lai C, Klein R, et al. Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor [see comments] Nature. 1995;378(6555):390–4. doi: 10.1038/378390a0. [DOI] [PubMed] [Google Scholar]
  • 6.Lee AD, Hansen PA, Holloszy JO. Wortmannin inhibits insulin-stimulated but not contraction-stimulated glucose transport activity in skeletal muscle. FEBS Lett. 1995 Mar 13;361(1):51–4. doi: 10.1016/0014-5793(95)00147-2. [DOI] [PubMed] [Google Scholar]
  • 7.Meyer D, Birchmeier C. Multiple essential functions of neuregulin in development. Nature. 1995;378(6555):386–90. doi: 10.1038/378386a0. [DOI] [PubMed] [Google Scholar]
  • 8.Liu X, Hwang H, Cao L, Buckland M, Cunningham A, Chen J, et al. Domain-specific gene disruption reveals critical regulation of neuregulin signaling by its cytoplasmic tail. Proc Natl Acad Sci U S A. 1998 Oct 27;95(22):13024–9. doi: 10.1073/pnas.95.22.13024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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(6555):394–8. doi: 10.1038/378394a0. [DOI] [PubMed] [Google Scholar]
  • 10.Zhao YY, Sawyer DR, Baliga RR, Opel DJ, Han X, Marchionni MA, et al. Neuregulins promote survival and growth of cardiac myocytes. Persistence of ErbB2 and ErbB4 expression in neonatal and adult ventricular myocytes. JBiolChem. 1998;273(17):10261–9. doi: 10.1074/jbc.273.17.10261. [DOI] [PubMed] [Google Scholar]
  • 11.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 Nov 15;311(1):135–46. doi: 10.1016/j.yexcr.2005.08.017. [DOI] [PubMed] [Google Scholar]
  • 12.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 Mar;300(3):H931–42. doi: 10.1152/ajpheart.00385.2010. [DOI] [PubMed] [Google Scholar]
  • 13.Kuramochi Y, Cote GM, Guo X, LeBrasseur NK, Cui L, Liao R, et al. Cardiac endothelial cells regulate ROS-induced cardiomyocyte apoptosis through neuregulin-1beta /erbB4 signaling. J Biol Chem. 2004 Sep 21; doi: 10.1074/jbc.M408662200. [DOI] [PubMed] [Google Scholar]
  • 14.Russell KS, Stern DF, Polverini PJ, Bender JR. Neuregulin activation of ErbB receptors in vascular endothelium leads to angiogenesis. Am J Physiol Heart Circ Physiol. 1999;277(6 Pt 2):H2205–H11. doi: 10.1152/ajpheart.1999.277.6.H2205. [DOI] [PubMed] [Google Scholar]
  • 15.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344(11):783–92. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
  • 16.Ozcelik C, Erdmann B, Pilz B, Wettschureck N, Britsch S, Hubner N, et al. Conditional mutation of the ErbB2 (HER2) receptor in cardiomyocytes leads to dilated cardiomyopathy. Proc Natl Acad Sci U S A. 2002 Jun 25;99(13):8880–5. doi: 10.1073/pnas.122249299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Crone SA, Zhao YY, Fan L, Gu Y, Minamisawa S, Liu Y, et al. ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat Med. 2002 May;8(5):459–65. doi: 10.1038/nm0502-459. [DOI] [PubMed] [Google Scholar]
  • 18.Liu FF, Stone JR, Schuldt AJ, Okoshi K, Okoshi MP, Nakayama M, et al. Heterozygous knockout of neuregulin-1 gene in mice exacerbates doxorubicin-induced heart failure. Am J Physiol Heart Circ Physiol. 2005 Aug;289(2):H660–6. doi: 10.1152/ajpheart.00268.2005. [DOI] [PubMed] [Google Scholar]
  • 19.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 Apr 2;105(13):1551–4. doi: 10.1161/01.cir.0000013839.41224.1c. [DOI] [PubMed] [Google Scholar]
  • 20.Moondra V, Sarma S, Buxton T, Safa R, Cote G, Storer T, et al. Serum Neuregulin-1β as a Biomarker of Cardiovascular Fitness. Open Biomarkers Journal. 2009;2:5. doi: 10.2174/1875318300902010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lebrasseur NK, Cote GM, Miller TA, Fielding RA, Sawyer DB. Regulation of neuregulin/ErbB signaling by contractile activity in skeletal muscle. Am J Physiol Cell Physiol. 2003 May;284(5):C1149–55. doi: 10.1152/ajpcell.00487.2002. [DOI] [PubMed] [Google Scholar]
  • 22.Ky B, Kimmel SE, Safa RN, Putt ME, Sweitzer NK, Fang JC, et al. Neuregulin-1 beta is associated with disease severity and adverse outcomes in chronic heart failure. Circulation. 2009 Jul 28;120(4):310–7. doi: 10.1161/CIRCULATIONAHA.109.856310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kuramochi Y, Cote GM, Guo X, Lebrasseur NK, Cui L, Liao R, et al. Cardiac endothelial cells regulate reactive oxygen species-induced cardiomyocyte apoptosis through neuregulin-1beta/erbB4 signaling. J Biol Chem. 2004 Dec 3;279(49):51141–7. doi: 10.1074/jbc.M408662200. [DOI] [PubMed] [Google Scholar]
  • 24.Lemmens K, Doggen K, De Keulenaer GW. Role of neuregulin-1/ErbB signaling in cardiovascular physiology and disease: implications for therapy of heart failure. Circulation. 2007 Aug 21;116(8):954–60. doi: 10.1161/CIRCULATIONAHA.107.690487. [DOI] [PubMed] [Google Scholar]
  • 25.Doggen K, Ray L, Mathieu M, Mc Entee K, Lemmens K, De Keulenaer GW. Ventricular ErbB2/ErbB4 activation and downstream signaling in pacing-induced heart failure. J Mol Cell Cardiol. 2009 Jan;46(1):33–8. doi: 10.1016/j.yjmcc.2008.10.010. [DOI] [PubMed] [Google Scholar]
  • 26.Rohrbach S, Yan X, Weinberg EO, Hasan F, Bartunek J, Marchionni MA, et al. Neuregulin in cardiac hypertrophy in rats with aortic stenosis. Differential expression of erbB2 and erbB4 receptors. Circulation. 1999 Jul 27;100(4):407–12. doi: 10.1161/01.cir.100.4.407. [DOI] [PubMed] [Google Scholar]
  • 27.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 May;100(3):240–9. doi: 10.1007/s00395-005-0514-4. [DOI] [PubMed] [Google Scholar]
  • 28.Xu G, Watanabe T, Iso Y, Koba S, Sakai T, Nagashima M, et al. Preventive effects of heregulin-beta1 on macrophage foam cell formation and atherosclerosis. Circ Res. 2009 Aug 28;105(5):500–10. doi: 10.1161/CIRCRESAHA.109.193870. [DOI] [PubMed] [Google Scholar]
  • 29.Hintsanen M, Elovainio M, Puttonen S, Kivimaki M, Raitakari OT, Lehtimaki T, et al. Neuregulin-1 genotype moderates the association between job strain and early atherosclerosis in young men. Ann Behav Med. 2007 Apr;33(2):148–55. doi: 10.1007/BF02879896. [DOI] [PubMed] [Google Scholar]
  • 30.Liu X, Gu X, Li Z, Li X, Li H, Chang J, et al. Neuregulin-1/erbB-activation improves cardiac function and survival in models of ischemic, dilated, and viral cardiomyopathy. J Am Coll Cardiol. 2006 Oct 3;48(7):1438–47. doi: 10.1016/j.jacc.2006.05.057. [DOI] [PubMed] [Google Scholar]
  • 31.Bian Y, Sun M, Silver M, Ho KK, Marchionni MA, Caggiano AO, et al. Neuregulin-1 attenuated doxorubicin-induced decrease in cardiac troponins. Am J Physiol Heart Circ Physiol. 2009 Dec;297(6):H1974–83. doi: 10.1152/ajpheart.01010.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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 Jan;38(1):105–8. [PubMed] [Google Scholar]
  • 33.Bersell K, Arab S, Haring B, Kuhn B. Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell. 2009 Jul 23;138(2):257–70. doi: 10.1016/j.cell.2009.04.060. [DOI] [PubMed] [Google Scholar]
  • 34.Ganguly A, iaci JF, Srinivas M, Vecchione AV, Hackett CS, Kasperbauer S, et al. Neuregulin Treatment of Heart Failure in Rat. Circ Res. 2008;103:e51. [Google Scholar]
  • 35.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 U S A. 1996;93(10):4833–8. doi: 10.1073/pnas.93.10.4833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Eto K, Eda K, Kanemoto S, Abe S. The immunoglobulin-like domain is involved in interaction of Neuregulin1 with ErbB. Biochem Biophys Res Commun. 2006 Nov 17;350(2):263–71. doi: 10.1016/j.bbrc.2006.09.028. [DOI] [PubMed] [Google Scholar]
  • 37.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 Jan 27;109(3):324–6. doi: 10.1161/01.CIR.0000114521.88547.5E. [DOI] [PubMed] [Google Scholar]
  • 38.Okoshi K, Nakayama M, Yan X, Okoshi MP, Schuldt AJ, Marchionni MA, et al. Neuregulins regulate cardiac parasympathetic activity: muscarinic modulation of beta-adrenergic activity in myocytes from mice with neuregulin-1 gene deletion. Circulation. 2004 Aug 10;110(6):713–7. doi: 10.1161/01.CIR.0000138109.32748.80. [DOI] [PubMed] [Google Scholar]
  • 39.Timolati F, Ott D, Pentassuglia L, Giraud MN, Perriard JC, Suter TM, et al. Neuregulin-1 beta attenuates doxorubicin-induced alterations of excitation-contraction coupling and reduces oxidative stress in adult rat cardiomyocytes. J Mol Cell Cardiol. 2006 Nov;41(5):845–54. doi: 10.1016/j.yjmcc.2006.08.002. [DOI] [PubMed] [Google Scholar]
  • 40.Brero A, Ramella R, Fitou A, Dati C, Alloatti G, Gallo MP, et al. Neuregulin-1beta1 rapidly modulates nitric oxide synthesis and calcium handling in rat cardiomyocytes. Cardiovasc Res. 2010 Dec 1;88(3):443–52. doi: 10.1093/cvr/cvq238. [DOI] [PubMed] [Google Scholar]
  • 41.Gao R, Zhang J, Cheng L, Wu X, Dong W, Yang X, et al. 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 May 4;55(18):1907–14. doi: 10.1016/j.jacc.2009.12.044. [DOI] [PubMed] [Google Scholar]
  • 42.Jabbour A, Hayward CS, Keogh AM, Kotlyar E, McCrohon JA, England JF, et al. 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 Jan;13(1):83–92. doi: 10.1093/eurjhf/hfq152. [DOI] [PubMed] [Google Scholar]
  • 43.Zensun Sci & Tech CoL. Clinical Trial to Evaluate the Efficacy and Safety of Recombinant Human Neuregulin-1 for Subcutaneous Administration in Patients With Chronic Systolic Heart Failure. ClinicalTrialsgov [Google Scholar]
  • 44.Zensun Sci. & Tech. Co. L. Study of the Survival of Recombinant Human Neuregulin-1ß in Chronic Heart Failure (CHF) Patients. ClinicalTrialsgov [Google Scholar]
  • 45.Zensun Sci. & Tech. Co. L. Efficacy and Safety of Human Neuregulin-1 to Treat Stable Chronic Heart Failure (ZS-01-210) ClinicalTrialsgov [Google Scholar]
  • 46.Therapeutics. A. Single Ascending Doses of GGF2 in Patients With Left Ventricular Dysfunction and Symptomatic Heart Failure (GGF2-1101-1) ClinicalTrialsgov [Google Scholar]
  • 47.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127–37. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
  • 48.Chua YL, Ito Y, Pole JC, Newman S, Chin SF, Stein RC, et al. The NRG1 gene is frequently silenced by methylation in breast cancers and is a strong candidate for the 8p tumour suppressor gene. Oncogene. 2009 Nov 19;28(46):4041–52. doi: 10.1038/onc.2009.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.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 Jan;1(3):165–75. [PubMed] [Google Scholar]
  • 50.Kato T, Kasai A, Mizuno M, Fengyi L, Shintani N, Maeda S, et al. Phenotypic characterization of transgenic mice overexpressing neuregulin-1. PLoS One. 2010;5(12):e14185. doi: 10.1371/journal.pone.0014185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Marchionni MA, Cannella B, Hoban C, Gao YL, Garcia-Arenas R, Lawson D, et al. Neuregulin in neuron/glial interactions in the central nervous system. GGF2 diminishes autoimmune demyelination, promotes oligodendrocyte progenitor expansion, and enhances remyelination. Adv Expt Med Biol. 1999;468:283–95. [PubMed] [Google Scholar]
  • 52.Xu Z, Jiang J, Ford G, Ford BD. Neuregulin-1 is neuroprotective and attenuates inflammatory responses induced by ischemic stroke. Biochem Biophys Res Commun. 2004 Sep 17;322(2):440–6. doi: 10.1016/j.bbrc.2004.07.149. [DOI] [PubMed] [Google Scholar]
  • 53.Iaci JF, Ganguly A, Finklestein SP, Parry TJ, Ren J, Saha S, et al. Glial growth factor 2 promotes functional recovery with treatment initiated up to 7 days after permanent focal ischemic stroke. Neuropharmacology. 2010 Dec;59(7–8):640–9. doi: 10.1016/j.neuropharm.2010.07.017. [DOI] [PubMed] [Google Scholar]
  • 54.Stefansson H, Sigurdsson E, Steinthorsdottir V, Bjornsdottir S, Sigmundsson T, Ghosh S, et al. Neuregulin 1 and susceptibility to schizophrenia. Am J Hum Genet. 2002 Oct;71(4):877–92. doi: 10.1086/342734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Moises HW, Zoega T, Gottesman The glial growth factors deficiency and synaptic destabilization hypothesis of schizophrenia. BMC Psychiatry. 2002 Jul 3;2:8. doi: 10.1186/1471-244X-2-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.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 May 1;69(2):247–58. doi: 10.1002/jbm.a.20119. [DOI] [PubMed] [Google Scholar]
  • 57.ter Laak MP, Hamers FP, Kirk CJ, Gispen WH. rhGGF2 protects against cisplatin-induced neuropathy in the rat. J Neurosci Res. 2000 Apr 15;60(2):237–44. doi: 10.1002/(SICI)1097-4547(20000415)60:2<237::AID-JNR13>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
  • 58.Jo SA, Zhu X, Marchionni MA, Burden SJ. Neuregulins are concentrated at nerve-muscle synapses and activate ACh- receptor gene expression. Nature. 1995;373(6510):158–61. doi: 10.1038/373158a0. [DOI] [PubMed] [Google Scholar]
  • 59.Ford BD, Han B, Fischbach GD. Differentiation-dependent regulation of skeletal myogenesis by neuregulin-1. Biochem Biophys Res Commun. 2003 Jun 20;306(1):276–81. doi: 10.1016/s0006-291x(03)00964-1. [DOI] [PubMed] [Google Scholar]
  • 60.Suarez E, Bach D, Cadefau J, Palacin M, Zorzano A, Guma 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 May 25;276(21):18257–64. doi: 10.1074/jbc.M008100200. [DOI] [PubMed] [Google Scholar]
  • 61.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 May 31;271(22):12699–702. doi: 10.1074/jbc.271.22.12699. [DOI] [PubMed] [Google Scholar]
  • 62.Zorzano A, Kaliman P, Guma A, Palacin M. Intracellular signals involved in the effects of insulin-like growth factors and neuregulins on myofibre formation. Cell Signal. 2003 Feb;15(2):141–9. doi: 10.1016/s0898-6568(02)00081-5. [DOI] [PubMed] [Google Scholar]
  • 63.Fukazawa R, Miller TA, Kuramochi Y, Frantz S, Kim YD, Marchionni MA, et al. Neuregulin-1 protects ventricular myocytes from anthracycline-induced apoptosis via erbB4-dependent activation of PI3-kinase/Akt. J Mol Cell Cardiol. 2003 Dec;35(12):1473–9. doi: 10.1016/j.yjmcc.2003.09.012. [DOI] [PubMed] [Google Scholar]
  • 64.Grazette LP, Boecker W, Matsui T, Semigran M, Force TL, Hajjar RJ, et al. Inhibition of ErbB2 causes mitochondrial dysfunction in cardiomyocytes: implications for herceptin-induced cardiomyopathy. J Am Coll Cardiol. 2004 Dec 7;44(11):2231–8. doi: 10.1016/j.jacc.2004.08.066. [DOI] [PubMed] [Google Scholar]
  • 65.Baliga RR, Pimental DR, Zhao YY, Simmons WW, Marchionni MA, Sawyer DB, et al. NRG-1-induced cardiomyocyte hypertrophy. Role of PI-3-kinase, p70(S6K), and MEK-MAPK-RSK. Am J Physiol Heart Circ Physiol. 1999;277(5 Pt 2):H2026–H37. doi: 10.1152/ajpheart.1999.277.5.H2026. [DOI] [PubMed] [Google Scholar]
  • 66.Kuramochi Y, Guo X, Sawyer DB. Neuregulin activates erbB2-dependent src/FAK signaling and cytoskeletal remodeling in isolated adult rat cardiac myocytes. J Mol Cell Cardiol. 2006 Aug;41(2):228–35. doi: 10.1016/j.yjmcc.2006.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zhu WZ, Xie Y, Moyes KW, Gold JD, Askari B, Laflamme MA. Neuregulin/ErbB signaling regulates cardiac subtype specification in differentiating human embryonic stem cells. Circ Res. 2010 Sep 17;107(6):776–86. doi: 10.1161/CIRCRESAHA.110.223917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rentschler S, Zander J, Meyers K, France D, Levine R, Porter G, et al. Neuregulin-1 promotes formation of the murine cardiac conduction system. Proc Natl Acad Sci U S A. 2002 Aug 6;99(16):10464–9. doi: 10.1073/pnas.162301699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Safa R, Peng XY, Pentassuglia L, Lim CC, Lamparter M, Silverstein C, et al. Neuregulin-1{beta} Regulation of Embryonic Endothelial Progenitor Cell Survival. Am J Physiol Heart Circ Physiol. 2011 Jan 14; doi: 10.1152/ajpheart.01104.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES