1
|
Novacescu D, Nesiu A, Bardan R, Latcu SC, Dema VF, Croitor A, Raica M, Cut TG, Walter J, Cumpanas AA. Rats, Neuregulins and Radical Prostatectomy: A Conceptual Overview. J Clin Med 2023; 12:jcm12062208. [PMID: 36983210 PMCID: PMC10051646 DOI: 10.3390/jcm12062208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
In the contemporary era of early detection, with mostly curative initial treatment for prostate cancer (PC), mortality rates have significantly diminished. In addition, mean age at initial PC diagnosis has decreased. Despite technical advancements, the probability of erectile function (EF) recovery post radical prostatectomy (RP) has not significantly changed throughout the last decade. Due to virtually unavoidable intraoperative cavernous nerve (CN) lesions and operations with younger patients, post-RP erectile dysfunction (ED) has now begun affecting these younger patients. To address this pervasive limitation, a plethora of CN lesion animal model investigations have analyzed the use of systemic/local treatments for EF recovery post-RP. Most promisingly, neuregulins (NRGs) have demonstrated neurotrophic effects in both neurodegenerative disease and peripheral nerve injury models. Recently, glial growth factor 2 (GGF2) has demonstrated far superior, dose-dependent, neuroprotective/restorative effects in the CN injury rat model, as compared to previous therapeutic counterparts. Although potentially impactful, these initial findings remain limited and under-investigated. In an effort to aid clinicians, our paper reviews post-RP ED pathogenesis and currently available therapeutic tools. To stimulate further experimentation, a standardized preparation protocol and in-depth analysis of applications for the CN injury rat model is provided. Lastly, we report on NRGs, such as GGF2, and their potentially revolutionary clinical applications, in hopes of identifying relevant future research directions.
Collapse
Affiliation(s)
- Dorin Novacescu
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Alexandru Nesiu
- Department Medicine, Discipline of Urology, Vasile Goldiş Western University, Liviu Rebreanu Boulevard, Nr. 86, 310414 Arad, Romania
- Correspondence: ; Tel.: +40-753521488
| | - Razvan Bardan
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Silviu Constantin Latcu
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Vlad Filodel Dema
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Alexei Croitor
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Marius Raica
- Department II, Discipline of Histology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - Talida Georgiana Cut
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Center for Ethics in Human Genetic Identifications, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| | - James Walter
- Emeritus, Department of Urology, Loyola Medical Center, Maywood, IL 60153, USA
| | - Alin Adrian Cumpanas
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
| |
Collapse
|
2
|
Asker H, Yilmaz-Oral D, Oztekin CV, Gur S. An update on the current status and future prospects of erectile dysfunction following radical prostatectomy. Prostate 2022; 82:1135-1161. [PMID: 35579053 DOI: 10.1002/pros.24366] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/30/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Radical prostatectomy (RP) and radiation treatment are standard options for localized prostate cancer. Even though nerve-sparing techniques have been increasingly utilized in RP, erectile dysfunction (ED) due to neuropraxia remains a frequent complication. Erectile function recovery rates after RP remain unsatisfactory, and many men still suffer despite the availability of various therapies. OBJECTIVE This systematic review aims to summarize the current treatments for post-RP-ED, assess the underlying pathological mechanisms, and emphasize promising therapeutic strategies based on the evidence from basic research. METHOD Evaluation and review of articles on the relevant topic published between 2010 and 2021, which are indexed and listed in the PubMed database. RESULTS Phosphodiesterase type 5 inhibitors, intracavernosal and intraurethral injections, vacuum erection devices, pelvic muscle training, and surgical procedures are utilized for penile rehabilitation. Clinical trials evaluating the efficacy of erectogenic drugs in this setting are conflicting and far from being conclusive. The use of androgen deprivation therapy in certain scenarios after RP further exacerbates the already problematic situation and emphasizes the need for effective treatment strategies. CONCLUSION This article is a detailed overview focusing on the pathophysiology and mechanism of the nerve injury developed during RP and a compilation of various strategies to induce cavernous nerve regeneration to improve erectile function (EF). These strategies include stem cell therapy, gene therapy, growth factors, low-intensity extracorporeal shockwave therapy, immunophilins, and various pharmacological approaches that have induced improvements in EF in experimental models of cavernous nerve injury. Many of the mentioned strategies can improve EF following RP if transformed into clinically applicable safe, and effective techniques with reproducible outcomes.
Collapse
Affiliation(s)
- Heba Asker
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
- Department of Medical Pharmacology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Didem Yilmaz-Oral
- Department of Pharmacology, Faculty of Pharmacy, Cukurova University, Adana, Turkey
| | - Cetin Volkan Oztekin
- Department of Urology, Faculty of Medicine, University of Kyrenia, Girne, Turkey
| | - Serap Gur
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
3
|
Collyer J, Xu F, Munkhsaikhan U, Alberson NF, Orgil BO, Zhang W, Czosek RJ, Lu L, Jefferies JL, Towbin JA, Purevjav E. Combining whole exome sequencing with in silico analysis and clinical data to identify candidate variants in pediatric left ventricular noncompaction. Int J Cardiol 2022; 347:29-37. [PMID: 34752814 DOI: 10.1016/j.ijcard.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Understanding the overall variant burden in pediatric patients with left ventricular noncompaction (LVNC) has clinical implications. Whole exome sequencing (WES) allows detection of coding variants in both candidate cardiomyopathy genes and those included on commercial panels. Other lines of evidence, including in silico analysis, are necessary to reduce the overwhelming number of variants to those most likely having a phenotypic impact. METHODS Five families, including five pediatric probands with LVNC, 5 other affected, and 10 unaffected family members, had WES performed, followed by bioinformatics filtering and Sanger sequencing. Review of the HGMD, variant classification by ACMG guidelines, and clinical information were used to further refine complex genotypes. RESULTS One nonsense and eleven missense variants were identified. In Family 1, affected siblings carried digenic heterozygous variants: E1350K-MYH7 and A276V-ANKRD1. The proband also carried heterozygous W143X-NRG1. Four affected members of Family 2 carried K184Q-MYH7 while unaffected members did not. In Family 3, homozygous A161T-MYH7 and heterozygous P4935T-OBSCN variants were identified in the proband with the latter being absent in his unaffected brother. In Family 4, proband's father and half-sibling have mild hypertrabeculation and carry T3796I-PLEC. The proband, carrying T3796I-PLEC and V2878A-OBSCN, demonstrated higher trabeculation burden. The proband in Family 5 carried four variants, R3247W-PLEC, C92Y-ERG, T1233M-NCOR2, and E54K-HIST1H4B. Application of ACMG criteria and clinical data revealed that W143X-NRG1, P4935T-OBSCN, and V2878A-OBSCN likely have no phenotypic role. CONCLUSIONS We report nine variants, including novel T3796I-PLEC and biallelic A161T-MYH7, likely contributing to phenotypes ranging from asymptomatic hypertrabeculation to severe LVNC with heart failure.
Collapse
Affiliation(s)
- John Collyer
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America; Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States of America
| | - Fuyi Xu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America; School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Undral Munkhsaikhan
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America; Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States of America
| | - Neely F Alberson
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America; Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States of America
| | - Buyan-Ochir Orgil
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America; Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States of America
| | - Wenying Zhang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America; Laboratory of Genetics and Genomics, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Richard J Czosek
- Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - John L Jefferies
- Division of Adult Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, United States of America; Pediatric Cardiology, Le Bonheur Children's Hospital, Memphis, TN, United States of America; Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, TN, United States of America
| | - Jeffrey A Towbin
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America; Pediatric Cardiology, Le Bonheur Children's Hospital, Memphis, TN, United States of America; Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, TN, United States of America
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America; Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States of America.
| |
Collapse
|
4
|
Shraim BA, Moursi MO, Benter IF, Habib AM, Akhtar S. The Role of Epidermal Growth Factor Receptor Family of Receptor Tyrosine Kinases in Mediating Diabetes-Induced Cardiovascular Complications. Front Pharmacol 2021; 12:701390. [PMID: 34408653 PMCID: PMC8365470 DOI: 10.3389/fphar.2021.701390] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Bara A Shraim
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Moaz O Moursi
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
5
|
Neuregulins: protective and reparative growth factors in multiple forms of cardiovascular disease. Clin Sci (Lond) 2021; 134:2623-2643. [PMID: 33063822 PMCID: PMC7557502 DOI: 10.1042/cs20200230] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Neuregulins (NRGs) are protein ligands that act through ErbB receptor tyrosine kinases to regulate tissue morphogenesis, plasticity, and adaptive responses to physiologic needs in multiple tissues, including the heart and circulatory system. The role of NRG/ErbB signaling in cardiovascular biology, and how it responds to physiologic and pathologic stresses is a rapidly evolving field. While initial concepts focused on the role that NRG may play in regulating cardiac myocyte responses, including cell survival, growth, adaptation to stress, and proliferation, emerging data support a broader role for NRGs in the regulation of metabolism, inflammation, and fibrosis in response to injury. The constellation of effects modulated by NRGs may account for the findings that two distinct forms of recombinant NRG-1 have beneficial effects on cardiac function in humans with systolic heart failure. NRG-4 has recently emerged as an adipokine with similar potential to regulate cardiovascular responses to inflammation and injury. Beyond systolic heart failure, NRGs appear to have beneficial effects in diastolic heart failure, prevention of atherosclerosis, preventing adverse effects on diabetes on the heart and vasculature, including atherosclerosis, as well as the cardiac dysfunction associated with sepsis. Collectively, this literature supports the further examination of how this developmentally critical signaling system functions and how it might be leveraged to treat cardiovascular disease.
Collapse
|
6
|
Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases. Cancers (Basel) 2020; 13:cancers13010084. [PMID: 33396739 PMCID: PMC7795854 DOI: 10.3390/cancers13010084] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The aim of this review is to provide an overview of the current scientific evidence concerning the role played by exosomes in the pathogenesis, diagnosis and treatment of diseases. The potential use of exosomes as delivery vectors for small-molecule therapeutic agents will be discussed. In addition, a special emphasis will be placed on the involvement of exosomes in oncological diseases, as well as to their potential therapeutic application as liquid biopsy tools mainly in cancer diagnosis. A better understanding of exosome biology could improve the results of clinical interventions using exosomes as therapeutic agents. Abstract Exosomes are lipid bilayer particles released from cells into their surrounding environment. These vesicles are mediators of near and long-distance intercellular communication and affect various aspects of cell biology. In addition to their biological function, they play an increasingly important role both in diagnosis and as therapeutic agents. In this paper, we review recent literature related to the molecular composition of exosomes, paying special attention to their role in pathogenesis, along with their application as biomarkers and as therapeutic tools. In this context, we analyze the potential use of exosomes in biomedicine, as well as the limitations that preclude their wider application.
Collapse
|
7
|
Rao P, Liu Z, Duan H, Dang S, Li H, Zhong L, Wang X, Wang L, Wang X. Pretreatment with neuregulin-1 improves cardiac electrophysiological properties in a rat model of myocardial infarction. Exp Ther Med 2019; 17:3141-3149. [PMID: 30936986 PMCID: PMC6434250 DOI: 10.3892/etm.2019.7306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022] Open
Abstract
Neuregulin-1 (NRG-1) is considered to be a potential therapeutic agent for cardiovascular diseases due to its diverse protective effects. The aim of the present study was to investigate the effect of NRG-1 on cardiac electrophysiology in rats with myocardial infarction (MI). The rats were randomly divided into three groups: The sham operation group (SO; n=8); MI group (n=8); and the MI with recombinant human NRG (rhNRG)-1 administration group (NRG-1 group; 10 µg/kg; n=8). A rat MI model was established via ligation of the left anterior descending coronary artery. The rats in the NRG-1 group received a 10 µg/kg rhNRG-1 injection through the tail vein 30 min prior to ligation. Following 24 h of intervention, the field potential (FP) parameters, including the interspike interval (ISI), field potential duration (FPD), FPrise, FPmin, FPmax and conduction velocity (CV), were measured using microelectrode array technology. Subsequently, burst pacing was performed to assess ventricular arrhythmia (VA) susceptibility in the left ventricle. FP parameters in the MI group were significantly different when compared with those observed in the SO group. ISI, FPD, FPrise and FPmax in the infarct, peri-infarct and normal zones, as well as the CV of the infarct and peri-infarct zones, were all significantly decreased, and FPmin in the normal zone was increased (P<0.05). However, when compared with the MI group, NRG-1 prolonged the ISI and FPD in the 3 zones, and increased FPrise in the infarct zone, FPmax in the normal zone and CV in the peri-infarct zone; it also decreased FPmin in the normal zone (P<0.05). Furthermore, the incidence of VA was significantly reduced in the NRG-1 group when compared with the MI group (P<0.05). In conclusion, NRG-1 improved cardiac electrophysiological properties and reduced VA susceptibility in acute MI.
Collapse
Affiliation(s)
- Panpan Rao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan Pulmonary Hospital, Wuhan, Hubei 430060, P.R. China
| | - Ziqiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan Pulmonary Hospital, Wuhan, Hubei 430060, P.R. China
| | - Huinan Duan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Department of Cardiology, Wuhan Pulmonary Hospital, Wuhan, Hubei 430060, P.R. China
| | - Song Dang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan Pulmonary Hospital, Wuhan, Hubei 430060, P.R. China
| | - Haitao Li
- Department of Cardiology, Hainan General Hospital, Haikou, Hainan 570100, P.R. China
| | - Liang Zhong
- Department of Anesthesiology, Wuhan Medical and Healthcare Center for Women and Children, Wuhan, Hubei 430015, P.R. China
| | - Xin Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan Pulmonary Hospital, Wuhan, Hubei 430060, P.R. China
| | - Long Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan Pulmonary Hospital, Wuhan, Hubei 430060, P.R. China.,Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan Pulmonary Hospital, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
8
|
Shoop S, Maria Z, Campolo A, Rashdan N, Martin D, Lovern P, Lacombe VA. Glial Growth Factor 2 Regulates Glucose Transport in Healthy Cardiac Myocytes and During Myocardial Infarction via an Akt-Dependent Pathway. Front Physiol 2019; 10:189. [PMID: 30971932 PMCID: PMC6445869 DOI: 10.3389/fphys.2019.00189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 02/14/2019] [Indexed: 12/23/2022] Open
Abstract
Neuregulin (NRG), a paracrine factor in myocytes, promotes cardiac development via the ErbB receptors. NRG-1β also improves cardiac function and cell survival after myocardial infarction (MI), although the mechanisms underlying these cardioprotective effects are not well elucidated. Increased glucose uptake has been shown to be cardio-protective during MI. We hypothesized that treatment with a recombinant version of NRG-1β, glial growth factor 2 (GGF2), will enhance glucose transport in the healthy myocardium and during MI. Cardiac myocytes were isolated from MI and healthy adult rats, and subsequently incubated with or without insulin or GGF2. Glucose uptake was measured using a fluorescent D-glucose analog. The translocation of glucose transporter (GLUT) 4 to the cell surface, the rate-limiting step in glucose uptake, was measured using a photolabeled biotinylation assay in isolated myocytes. Similar to insulin, acute in vitro GGF2 treatment increased glucose uptake in healthy cardiac myocytes (by 40 and 49%, respectively, P = 0.002). GGF2 treatment also increased GLUT4 translocation in healthy myocytes by 184% (P < 0.01), while ErbB 2/4 receptor blockade (by afatinib) abolished these effects. In addition, GGF2 treatment enhanced Akt phosphorylation (at both threonine and serine sites, by 75 and 139%, respectively, P = 0.029 and P = 0.01), which was blunted by ErbB 2/4 receptor blockade. GGF2 treatment increased the phosphorylation of AS160 (an Akt effector) by 72% (P < 0.05), as well as the phosphorylation of PDK-1 and PKC (by 118 and 92%, respectively, P < 0.05). During MI, cardiac GLUT4 translocation was downregulated by 44% (P = 0.004) and was partially rescued by both in vitro insulin and GGF2 treatment. Our data demonstrate that acute GGF2 treatment increased glucose transport in cardiac myocytes by activating the ErbB 2/4 receptors and subsequent key downstream effectors (i.e., PDK-1, Akt, AS160, and PKC). These findings highlight novel mechanisms of action of GGF2, which warrant further investigation in patients with heart failure.
Collapse
Affiliation(s)
- Shanell Shoop
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States.,Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
| | - Zahra Maria
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States.,Harold Hamm Diabetes Center, University of Oklahoma, Oklahoma City, OK, United States
| | - Allison Campolo
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States.,Harold Hamm Diabetes Center, University of Oklahoma, Oklahoma City, OK, United States
| | - Nabil Rashdan
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Dominic Martin
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
| | - Pamela Lovern
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Véronique A Lacombe
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States.,Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States.,Harold Hamm Diabetes Center, University of Oklahoma, Oklahoma City, OK, United States
| |
Collapse
|
9
|
Ferrini A, Stevens MM, Sattler S, Rosenthal N. Toward Regeneration of the Heart: Bioengineering Strategies for Immunomodulation. Front Cardiovasc Med 2019; 6:26. [PMID: 30949485 PMCID: PMC6437044 DOI: 10.3389/fcvm.2019.00026] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/26/2019] [Indexed: 01/10/2023] Open
Abstract
Myocardial Infarction (MI) is the most common cardiovascular disease. An average-sized MI causes the loss of up to 1 billion cardiomyocytes and the adult heart lacks the capacity to replace them. Although post-MI treatment has dramatically improved survival rates over the last few decades, more than 20% of patients affected by MI will subsequently develop heart failure (HF), an incurable condition where the contracting myocardium is transformed into an akinetic, fibrotic scar, unable to meet the body's need for blood supply. Excessive inflammation and persistent immune auto-reactivity have been suggested to contribute to post-MI tissue damage and exacerbate HF development. Two newly emerging fields of biomedical research, immunomodulatory therapies and cardiac bioengineering, provide potential options to target the causative mechanisms underlying HF development. Combining these two fields to develop biomaterials for delivery of immunomodulatory bioactive molecules holds great promise for HF therapy. Specifically, minimally invasive delivery of injectable hydrogels, loaded with bioactive factors with angiogenic, proliferative, anti-apoptotic and immunomodulatory functions, is a promising route for influencing the cascade of immune events post-MI, preventing adverse left ventricular remodeling, and offering protection from early inflammation to fibrosis. Here we provide an updated overview on the main injectable hydrogel systems and bioactive factors that have been tested in animal models with promising results and discuss the challenges to be addressed for accelerating the development of these novel therapeutic strategies.
Collapse
Affiliation(s)
- Arianna Ferrini
- Department of Materials, Imperial College London, London, United Kingdom,National Heart and Lung Institute and BHF Centre for Research Excellence, Imperial College London, London, United Kingdom
| | - Molly M. Stevens
- Department of Materials, Imperial College London, London, United Kingdom,Department of Bioengineering, Imperial College London, London, United Kingdom,Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Susanne Sattler
- National Heart and Lung Institute and BHF Centre for Research Excellence, Imperial College London, London, United Kingdom
| | - Nadia Rosenthal
- National Heart and Lung Institute and BHF Centre for Research Excellence, Imperial College London, London, United Kingdom,The Jackson Laboratory, Bar Harbor, ME, United States,*Correspondence: Nadia Rosenthal
| |
Collapse
|
10
|
Ryzhov S, Robich MP, Roberts DJ, Favreau-Lessard AJ, Peterson SM, Jachimowicz E, Rath R, Vary CPH, Quinn R, Kramer RS, Sawyer DB. ErbB2 promotes endothelial phenotype of human left ventricular epicardial highly proliferative cells (eHiPC). J Mol Cell Cardiol 2018; 115:39-50. [PMID: 29291395 PMCID: PMC5926239 DOI: 10.1016/j.yjmcc.2017.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/20/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022]
Abstract
The adult human heart contains a subpopulation of highly proliferative cells. The role of ErbB receptors in these cells has not been studied. From human left ventricular (LV) epicardial biopsies, we isolated highly proliferative cells (eHiPC) to characterize the cell surface expression and function of ErbB receptors in the regulation of cell proliferation and phenotype. We found that human LV eHiPC express all four ErbB receptor subtypes. However, the expression of ErbB receptors varied widely among eHiPC isolated from different subjects. eHiPC with higher cell surface expression of ErbB2 reproduced the phenotype of endothelial cells and were characterized by endothelial cell-like functional properties. We also found that EGF/ErbB1 induces VEGFR2 expression, while ligands for both ErbB1 and ErbB3/4 induce expression of Tie2. The number of CD31posCD45neg endothelial cells is higher in LV biopsies from subjects with high ErbB2 (ErbB2high) eHiPC compared to low ErbB2 (ErbB2low) eHiPC. These findings have important implications for potential strategies to increase the efficacy of cell-based revascularization of the injured heart, through promotion of an endothelial phenotype in cardiac highly proliferative cells.
Collapse
Affiliation(s)
- Sergey Ryzhov
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Michael P Robich
- Maine Medical Center Research Institute, Scarborough, ME, United States; Maine Medical Center, Portland, ME, United States
| | - Daniel J Roberts
- Maine Medical Center Research Institute, Scarborough, ME, United States; Maine Medical Center, Portland, ME, United States
| | | | - Sarah M Peterson
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | | | - Rutwik Rath
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Calvin P H Vary
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Reed Quinn
- Maine Medical Center, Portland, ME, United States
| | | | - Douglas B Sawyer
- Maine Medical Center Research Institute, Scarborough, ME, United States; Maine Medical Center, Portland, ME, United States.
| |
Collapse
|
11
|
Huang Z, Sawyer DB, Troy EL, McEwen C, Cleator JH, Murphy A, Caggiano AO, Eisen A, Parry TJ. Species-specific effects of neuregulin-1β (cimaglermin alfa) on glucose handling in animal models and humans with heart failure. Toxicol Appl Pharmacol 2017; 332:92-99. [PMID: 28780372 DOI: 10.1016/j.taap.2017.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/05/2017] [Accepted: 08/01/2017] [Indexed: 10/19/2022]
Abstract
Neuregulin-1β is a member of the neuregulin family of growth factors and is critically important for normal development and functioning of the heart and brain. A recombinant version of neuregulin-1β, cimaglermin alfa (also known as glial growth factor 2 or GGF2) is being investigated as a possible therapy for heart failure. Previous studies suggest that neuregulin-1β stimulation of skeletal muscle increases glucose uptake and, specifically, sufficient doses of cimaglermin alfa acutely produce hypoglycemia in pigs. Since acute hypoglycemia could be a safety concern, blood glucose changes in the above pig study were further investigated. In addition, basal glucose and glucose disposal were investigated in mice. Finally, as part of standard clinical chemistry profiling in a single ascending-dose human safety study, blood glucose levels were evaluated in patients with heart failure after cimaglermin alfa treatment. A single intravenous injection of cimaglermin alfa at doses of 0.8mg/kg and 2.6mg/kg in mice resulted in a transient reduction of blood glucose concentrations of approximately 20% and 34%, respectively, at 2h after the treatment compared to pre-treatment levels. Similar results were observed in diabetic mice. Treatment with cimaglermin alfa also increased blood glucose disposal following oral challenge in mice. However, no significant alterations in blood glucose concentrations were found in human heart failure patients at 0.5 and 2h after treatment with cimaglermin alfa over an equivalent human dose range, based on body surface area. Taken together, these data indicate strong species differences in blood glucose handling after cimaglermin alfa treatment, and particularly do not indicate that this phenomenon should affect human subjects.
Collapse
Affiliation(s)
- Zhihong Huang
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Douglas B Sawyer
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Erika L Troy
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA
| | - Corissa McEwen
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA
| | - John H Cleator
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Abigail Murphy
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Anthony O Caggiano
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA
| | - Andrew Eisen
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA
| | - Tom J Parry
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| |
Collapse
|
12
|
Gupte M, Lal H, Ahmad F, Sawyer DB, Hill MF. Chronic Neuregulin-1β Treatment Mitigates the Progression of Postmyocardial Infarction Heart Failure in the Setting of Type 1 Diabetes Mellitus by Suppressing Myocardial Apoptosis, Fibrosis, and Key Oxidant-Producing Enzymes. J Card Fail 2017; 23:887-899. [PMID: 28870731 DOI: 10.1016/j.cardfail.2017.08.456] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 08/11/2017] [Accepted: 08/18/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Type 1 diabetes mellitus (DM) patients surviving myocardial infarction (MI) have substantially higher cardiovascular morbidity and mortality compared to their nondiabetic counterparts owing to the more frequent development of subsequent heart failure (HF). Neuregulin (NRG)-1β is released from cardiac microvascular endothelial cells and acts as a paracrine factor via the ErbB family of tyrosine kinase receptors expressed in cardiac myocytes to regulate cardiac development and stress responses. Because myocardial NRG-1/ErbB signaling has been documented to be impaired during HF associated with type 1 DM, we examined whether enhancement of NRG-1β signaling via exogenous administration of recombinant NRG-1β could exert beneficial effects against post-MI HF in the type 1 diabetic heart. METHODS AND RESULTS Type 1 DM was induced in male Sprague Dawley rats by a single injection of streptozotocin (STZ) (65 mg/kg). Two weeks after induction of type 1 DM, rats underwent left coronary artery ligation to induce MI. STZ-diabetic rats were treated with saline or NRG-1β (100 µg/kg) twice per week for 7 weeks, starting 2 weeks before experimental MI. Residual left ventricular function was significantly greater in the NRG-1β-treated STZ-diabetic MI group compared with the vehicle-treated STZ-diabetic MI group 5 weeks after MI as assessed by high-resolution echocardiography. NRG-1β treatment of STZ-diabetic MI rats was associated with reduced myocardial fibrosis and apoptosis as well as decreased gene expression of key oxidant-producing enzymes. CONCLUSIONS These results suggest that recombinant NRG-1β may be a promising therapeutic for HF post-MI in the setting of type 1 DM.
Collapse
Affiliation(s)
- Manisha Gupte
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hind Lal
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Firdos Ahmad
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas B Sawyer
- Department of Cardiac Services, Maine Medical Center, Portland, Maine
| | - Michael F Hill
- Department of Professional and Medical Education, Meharry Medical College, Nashville, Tennessee.
| |
Collapse
|
13
|
Kirabo A, Ryzhov S, Gupte M, Sengsayadeth S, Gumina RJ, Sawyer DB, Galindo CL. Neuregulin-1β induces proliferation, survival and paracrine signaling in normal human cardiac ventricular fibroblasts. J Mol Cell Cardiol 2017; 105:59-69. [PMID: 28263756 PMCID: PMC5715731 DOI: 10.1016/j.yjmcc.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/14/2017] [Accepted: 03/01/2017] [Indexed: 01/27/2023]
Abstract
Neuregulin-1β (NRG-1β) is critical for cardiac development and repair, and recombinant forms are currently being assessed as possible therapeutics for systolic heart failure. We previously demonstrated that recombinant NRG-1β reduces cardiac fibrosis in an animal model of cardiac remodeling and heart failure, suggesting that there may be direct effects on cardiac fibroblasts. Here we show that NRG-1β receptors (ErbB2, ErbB3, and ErbB4) are expressed in normal human cardiac ventricular (NHCV) fibroblast cell lines. Treatment of NHCV fibroblasts with recombinant NRG-1β induced activation of the AKT pathway, which was phosphoinositide 3-kinase (PI3K)-dependent. Moreover, the NRG-1β-induced PI3K/AKT signaling in these cells required phosphorylation of both ErbB2 and ErbB3 receptors at tyrosine (Tyr)1248 and Tyr1289 respectively. RNASeq analysis of NRG-1β-treated cardiac fibroblasts obtained from three different individuals revealed a global gene expression signature consistent with cell growth and survival. We confirmed enhanced cellular proliferation and viability in NHCV fibroblasts in response to NRG-1β, which was abrogated by PI3K, ErbB2, and ErbB3 inhibitors. NRG-1β also induced production and secretion of cytokines (interleukin-1α and interferon-γ) and pro-reparative factors (angiopoietin-2, brain-derived neurotrophic factor, and crypto-1), suggesting a role in cardiac repair through the activation of paracrine signaling.
Collapse
Affiliation(s)
- Annet Kirabo
- Department of Pharmacology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Sergey Ryzhov
- Maine Medical Research Institute, 81 Research Drive, Scarborough, ME 04074, United States.
| | - Manisha Gupte
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Seng Sengsayadeth
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States; Department of Pathology, Immunology, and Microbiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Douglas B Sawyer
- Maine Medical Research Institute, 81 Research Drive, Scarborough, ME 04074, United States.
| | - Cristi L Galindo
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| |
Collapse
|
14
|
Morano M, Angotti C, Tullio F, Gambarotta G, Penna C, Pagliaro P, Geuna S. Myocardial ischemia/reperfusion upregulates the transcription of the Neuregulin1 receptor ErbB3, but only postconditioning preserves protein translation: Role in oxidative stress. Int J Cardiol 2017; 233:73-79. [PMID: 28162790 DOI: 10.1016/j.ijcard.2017.01.122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 12/28/2016] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Neuregulin1 (Nrg1) and its receptors ErbB are crucial for heart development and for adult heart structural maintenance and function and Nrg1 has been proposed for heart failure treatment. Infarct size is the major determinant of heart failure and the mechanism of action and the role of each ErbB receptor remain obscure, especially in the post-ischemic myocardium. We hypothesized that Nrg1 and ErbB are affected at transcriptional level early after ischemia/reperfusion (I/R) injury, and that the protective postconditioning procedure (PostC, brief cycles of ischemia/reperfusion carried out after a sustained ischemia) can influence this pathway. METHODS AND RESULTS The Langendorff's heart was used as an ex-vivo model to mimic an I/R injury in the whole rat heart; after 30min of ischemia and 2h of reperfusion, with or without PostC, Nrg1 and ErbB expression were analysed by quantitative real-time PCR and Western blot. While no changes occur for ErbB2, ErbB4 and Nrg1, an increase of ErbB3 expression occurs after I/R injury, with and without PostC. However, I/R reduces ErbB3 protein, whereas PostC preserves it. An in vitro analysis with H9c2 cells exposed to redox-stress indicated that the transient over-expression of ErbB3 alone is able to increase cell survival (MTT assay), limiting mitochondrial dysfunction (JC-1 probe) and apoptotic signals (Bax/Bcl-2 ratio). CONCLUSIONS This study suggests ErbB3 as a protective factor against death pathways activated by redox stress and supports an involvement of this receptor in the pro-survival responses.
Collapse
Affiliation(s)
- Michela Morano
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Carmelina Angotti
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy.
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy; Neuroscience Institute Cavalieri Ottolenghi (NICO), Torino, Italy
| |
Collapse
|
15
|
Parry TJ, Ganguly A, Troy EL, Luis Guerrero J, Iaci JF, Srinivas M, Vecchione AM, Button DC, Hackett CS, Zolty R, Sawyer DB, Caggiano AO. Effects of neuregulin GGF2 (cimaglermin alfa) dose and treatment frequency on left ventricular function in rats following myocardial infarction. Eur J Pharmacol 2016; 796:76-89. [PMID: 27993643 DOI: 10.1016/j.ejphar.2016.12.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 11/17/2022]
Abstract
Neuregulins are important growth factors involved in cardiac development and response to stress. Certain isoforms and fragments of neuregulin have been found to be cardioprotective. The effects of a full-length neuregulin-1β isoform, glial growth factor 2 (GGF2; USAN/INN; also called cimaglermin) were investigated in vitro. Various dosing regimens were then evaluated for their effects on left ventricular (LV) function in rats with surgically-induced myocardial infarction. In vitro, GGF2 bound with high affinity to erythroblastic leukemia viral oncogene (ErbB) 4 receptors, potently promoted Akt phosphorylation, as well as reduced cell death following doxorubicin exposure in HL1 cells. Daily GGF2 treatment beginning 7-14 days after left anterior descending coronary artery ligation produced improvements in LV ejection fraction and other measures of LV function and morphology. The improvements in LV function (e.g. 10% point increase in absolute LV ejection fraction) with GGF2 were dose-dependent. LV performance was substantially improved when GGF2 treatment was delivered infrequently, despite a serum half-life of less than 2h and could be maintained for more than 10 months with treatment once weekly or once every 2 weeks. These studies confirm previous findings that GGF2 may improve contractile performance in the failing rat heart and that infrequent exposure to GGF2 may improve LV function and impact remodeling in the failing myocardium. GGF2 is now being developed for the treatment of heart failure in humans.
Collapse
Affiliation(s)
- Tom J Parry
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Anindita Ganguly
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Erika L Troy
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - J Luis Guerrero
- Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA.
| | - Jennifer F Iaci
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Maya Srinivas
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Andrea M Vecchione
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Donald C Button
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Craig S Hackett
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| | - Ronald Zolty
- University of Nebraska Medical Center, 982265 S 42nd St & Emile St, Omaha, NE 68198, USA.
| | | | - Anthony O Caggiano
- Acorda Therapeutics, Inc., 420 Saw Mill River Rd, Ardsley, NY 10502, USA.
| |
Collapse
|
16
|
Lenihan DJ, Anderson SA, Lenneman CG, Brittain E, Muldowney JA, Mendes L, Zhao PZ, Iaci J, Frohwein S, Zolty R, Eisen A, Sawyer DB, Caggiano AO. A Phase I, Single Ascending Dose Study of Cimaglermin Alfa (Neuregulin 1β3) in Patients With Systolic Dysfunction and Heart Failure. JACC Basic Transl Sci 2016; 1:576-586. [PMID: 30167542 PMCID: PMC6113538 DOI: 10.1016/j.jacbts.2016.09.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 01/03/2023]
Abstract
A first-in-human, phase 1, double blind, placebo-controlled, single ascending dose study examined the safety, tolerability, and exploratory efficacy of intravenous infusion of a recombinant growth factor, cimaglermin alfa, in patients with heart failure and left ventricular systolic dysfunction (LVSD). In these patients on optimal guideline-directed medical therapy, cimaglermin treatment was generally tolerated except for transient nausea and headache and a dose-limiting toxicity was noted at the highest planned dose. There was a dose-dependent improvement in left ventricular ejection fraction lasting 90 days following infusion. Thus, cimaglermin is a potential therapy to enhance cardiac function in LVSD and warrants further investigation.
Collapse
Key Words
- AE, adverse event
- AUC, area under the curve
- DLT, dose-limiting toxicity
- GGF, glial growth factor
- HF, heart failure
- LVEF, left ventricular ejection fraction
- LVSD, left ventricular systolic dysfunction
- NRG, neuregulin
- NYHA, New York Heart Association functional class
- TEAE, treatment-emergent adverse event
- cardiac repair
- growth factor
- neuregulin
- systolic dysfunction
Collapse
Affiliation(s)
- Daniel J. Lenihan
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee
| | - Sarah A. Anderson
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee
| | | | - Evan Brittain
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee
| | - James A.S. Muldowney
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee
| | - Lisa Mendes
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee
| | | | | | - Stephen Frohwein
- Emory Heart and Vascular Center at Emory Saint Joseph’s, Atlanta, Georgia
| | - Ronald Zolty
- University of Nebraska Medical Center, Omaha, Nebraska
| | | | | | | |
Collapse
|
17
|
Samsa LA, Ito CE, Brown DR, Qian L, Liu J. IgG-Containing Isoforms of Neuregulin-1 Are Dispensable for Cardiac Trabeculation in Zebrafish. PLoS One 2016; 11:e0166734. [PMID: 27846271 PMCID: PMC5112773 DOI: 10.1371/journal.pone.0166734] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/02/2016] [Indexed: 12/25/2022] Open
Abstract
The Neuregulin-1 (Nrg1) signaling pathway has been widely implicated in many aspects of heart development including cardiac trabeculation. Cardiac trabeculation is an important morphogenetic process where clusters of ventricular cardiomyocytes extrude and expand into the lumen of the ventricular chambers. In mouse, Nrg1 isoforms containing an immunoglobulin-like (IgG) domain are essential for cardiac trabeculation through interaction with heterodimers of the epidermal growth factor-like (EGF-like) receptors ErbB2/ErbB4. Recent reports have underscored the importance of Nrg1 signaling in cardiac homeostasis and disease, however, placental development has precluded refined evaluation of the role of this pathway in mammals. ErbB2 has been shown to have a developmentally conserved role in cardiac trabeculation in zebrafish, a vertebrate model organism with completely external development, but the requirement for Nrg1 has not been examined. We found that among the multiple Nrg1 isoforms, the IgG domain-containing, type I Nrg1 (nrg1-I) is the only isoform detectable in the heart. Then, using CRISPR/Cas9 gene editing, we targeted the IgG domain of Nrg1 to produce novel alleles, nrg1nc28 and nrg1nc29, encoding nrg1-I and nrg1-II truncations. Our results indicated that zebrafish deficient for nrg1-I developed trabeculae in an ErbB2-dependent manner. Further, these mutants survive to reproductive adulthood with no overt cardiovascular defects. We also found that additional EGF-like ligands were expressed in the zebrafish heart during development of trabeculae. Together, these results suggest that Nrg1 is not the primary effector of trabeculation and/or that other EGF-like ligand(s) activates the ErbB2/ErbB4 pathway, either through functioning as the primary ligand or acting in a redundant manner. Overall, our work provides an example of cross-species differences in EGF family member requirements for an evolutionary conserved process.
Collapse
Affiliation(s)
- Leigh Ann Samsa
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America.,McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Cade Ellis Ito
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Daniel Ross Brown
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Li Qian
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
18
|
Favreau-Lessard AJ, Ryzhov S, Sawyer DB. Novel Biological Therapies Targeting Heart Failure: Myocardial Rejuvenation. Heart Fail Clin 2016; 12:461-71. [PMID: 27371521 DOI: 10.1016/j.hfc.2016.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recovery of ventricular function occurs in a subset of patients with advanced heart failure treated with medical and/or mechanical therapy. Finding strategies that induce ventricular recovery through induction of repair, regeneration, or "rejuvenation" is a long-sought goal of research programs. Cell-based strategies, use of recombinant growth and survival factors, and gene delivery are under investigation. In this brief article we highlight a few of the biological approaches in development to treat heart failure.
Collapse
Affiliation(s)
- Amanda J Favreau-Lessard
- Center for Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME 04074, USA
| | - Sergey Ryzhov
- Center for Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME 04074, USA
| | - Douglas B Sawyer
- Center for Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME 04074, USA.
| |
Collapse
|
19
|
Pentassuglia L, Heim P, Lebboukh S, Morandi C, Xu L, Brink M. Neuregulin-1β promotes glucose uptake via PI3K/Akt in neonatal rat cardiomyocytes. Am J Physiol Endocrinol Metab 2016; 310:E782-94. [PMID: 26979522 DOI: 10.1152/ajpendo.00259.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 03/02/2016] [Indexed: 12/21/2022]
Abstract
Nrg1β is critically involved in cardiac development and also maintains function of the adult heart. Studies conducted in animal models showed that it improves cardiac performance under a range of pathological conditions, which led to its introduction in clinical trials to treat heart failure. Recent work also implicated Nrg1β in the regenerative potential of neonatal and adult hearts. The molecular mechanisms whereby Nrg1β acts in cardiac cells are still poorly understood. In the present study, we analyzed the effects of Nrg1β on glucose uptake in neonatal rat ventricular myocytes and investigated to what extent mTOR/Akt signaling pathways are implicated. We show that Nrg1β enhances glucose uptake in cardiomyocytes as efficiently as IGF-I and insulin. Nrg1β causes phosphorylation of ErbB2 and ErbB4 and rapidly induces the phosphorylation of FAK (Tyr(861)), Akt (Thr(308) and Ser(473)), and its effector AS160 (Thr(642)). Knockdown of ErbB2 or ErbB4 reduces Akt phosphorylation and blocks the glucose uptake. The Akt inhibitor VIII and the PI3K inhibitors LY-294002 and Byl-719 abolish Nrg1β-induced phosphorylation and glucose uptake. Finally, specific mTORC2 inactivation after knockdown of rictor blocks the Nrg1β-induced increases in Akt-p-Ser(473) but does not modify AS160-p-Thr(642) or the glucose uptake responses to Nrg1β. In conclusion, our study demonstrates that Nrg1β enhances glucose uptake in cardiomyocytes via ErbB2/ErbB4 heterodimers, PI3Kα, and Akt. Furthermore, although Nrg1β activates mTORC2, the resulting Akt-Ser(473) phosphorylation is not essential for glucose uptake induction. These new insights into pathways whereby Nrg1β regulates glucose uptake in cardiomyocytes may contribute to the understanding of its regenerative capacity and protective function in heart failure.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Blotting, Western
- Gene Knockdown Techniques
- Glucose/metabolism
- Heart Ventricles/cytology
- Hypoglycemic Agents/pharmacology
- Immunoprecipitation
- Insulin/pharmacology
- Insulin-Like Growth Factor I/pharmacology
- Mechanistic Target of Rapamycin Complex 2
- Mice
- Mice, Inbred C57BL
- Multiprotein Complexes/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Neuregulin-1/pharmacology
- Phosphatidylinositol 3-Kinases/drug effects
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation/drug effects
- Protein Biosynthesis/drug effects
- Proto-Oncogene Proteins c-akt/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering
- Rats
- Receptor, ErbB-2/drug effects
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-4/drug effects
- Receptor, ErbB-4/genetics
- Receptor, ErbB-4/metabolism
- TOR Serine-Threonine Kinases/metabolism
Collapse
Affiliation(s)
- Laura Pentassuglia
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Philippe Heim
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Sonia Lebboukh
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christian Morandi
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Lifen Xu
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Marijke Brink
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
20
|
López-Soldado I, Niisuke K, Veiga C, Adrover A, Manzano A, Martínez-Redondo V, Camps M, Bartrons R, Zorzano A, Gumà A. Neuregulin improves response to glucose tolerance test in control and diabetic rats. Am J Physiol Endocrinol Metab 2016; 310:E440-51. [PMID: 26714846 DOI: 10.1152/ajpendo.00226.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 12/20/2015] [Indexed: 12/31/2022]
Abstract
Neuregulin (NRG) is an EGF-related growth factor that binds to the tyrosine kinase receptors ErbB3 and ErbB4, thus inducing tissue development and muscle glucose utilization during contraction. Here, we analyzed whether NRG has systemic effects regulating glycemia in control and type 2 diabetic rats. To this end, recombinant NRG (rNRG) was injected into Zucker diabetic fatty (ZDF) rats and their respective lean littermates 15 min before a glucose tolerance test (GTT) was performed. rNRG enhanced glucose tolerance without promoting the activation of the insulin receptor (IR) or insulin receptor substrates (IRS) in muscle and liver. However, in control rats, rNRG induced the phosphorylation of protein kinase B (PKB) and glycogen synthase kinase-3 (GSK-3) in liver but not in muscle. In liver, rNRG increased ErbB3 tyrosine phosphorylation and its binding to phosphatidylinositol 3-kinase (PI3K), thus indicating that rNRG activates the ErbB3/PI3K/PKB signaling pathway. rNRG increased glycogen content in liver but not in muscle. rNRG also increased the content of fructose-2,6-bisphosphate (Fru-2,6-P2), an activator of hepatic glycolysis, and lactate in liver but not in muscle. Increases in lactate were abrogated by wortmannin, a PI3K inhibitor, in incubated hepatocytes. The liver of ZDF rats showed a reduced content of ErbB3 receptors, entailing a minor stimulation of the rNRG-induced PKB/GSK-3 cascade and resulting in unaltered hepatic glycogen content. Nonetheless, rNRG increased hepatic Fru-2,6-P2 and augmented lactate both in liver and in plasma of diabetic rats. As a whole, rNRG improved response to the GTT in both control and diabetic rats by enhancing hepatic glucose utilization.
Collapse
Affiliation(s)
- Iliana López-Soldado
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Center for Biomedical Investigation in Net of Diabetes and Associated Metabolic Pathologies, Madrid, Spain
| | - Katrin Niisuke
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Catarina Veiga
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Anna Adrover
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Anna Manzano
- Department of Physiological Sciences II, Faculty of Medicine, University of Barcelona-IDIBELL: Bellvitge Institute for Biomedical Research, Barcelona, Spain; and
| | - Vicente Martínez-Redondo
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Marta Camps
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain; Center for Biomedical Investigation in Net of Diabetes and Associated Metabolic Pathologies, Madrid, Spain; Institute of Biomedicine from the University of Barcelona, Barcelona, Spain
| | - Ramon Bartrons
- Department of Physiological Sciences II, Faculty of Medicine, University of Barcelona-IDIBELL: Bellvitge Institute for Biomedical Research, Barcelona, Spain; and
| | - Antonio Zorzano
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Center for Biomedical Investigation in Net of Diabetes and Associated Metabolic Pathologies, Madrid, Spain
| | - Anna Gumà
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain; Center for Biomedical Investigation in Net of Diabetes and Associated Metabolic Pathologies, Madrid, Spain; Institute of Biomedicine from the University of Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Ferguson SW, Nguyen J. Exosomes as therapeutics: The implications of molecular composition and exosomal heterogeneity. J Control Release 2016; 228:179-190. [PMID: 26941033 DOI: 10.1016/j.jconrel.2016.02.037] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 12/20/2022]
Abstract
Harnessing exosomes as therapeutic drug delivery vehicles requires a better understanding of exosomal composition and their mode of action. A full appreciation of all the exosomal components (proteins, lipids, and RNA content) will be important for the design of effective exosome-based or exosome-mimicking drug carriers. In this review we describe the presence of rarely studied, non-coding RNAs that exist in high numbers in exosomes. We discuss the implications of the molecular composition and heterogeneity of exosomes on their biological and therapeutic effects. Finally, we highlight outstanding questions with regard to RNA loading into exosomes, analytical methods to sort exosomes and their sub-populations, and the effects of exosomal proteins and lipids on recipient cells. Investigations into these facets of exosome biology will further advance the field, could lead to the clinical translation of exosome-based therapeutics, and aid in the reverse-engineering of synthetic exosomes. Although synthetic exosomes are still an underexplored area, they could offer researchers a way to manufacture exosomes with highly defined structure, composition, and function.
Collapse
Affiliation(s)
- Scott W Ferguson
- Department of Pharmaceutical Sciences, School of Pharmacy, The State University of New York at Buffalo, United States
| | - Juliane Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy, The State University of New York at Buffalo, United States.
| |
Collapse
|
22
|
Iaci JF, Parry TJ, Huang Z, Pavlopoulos E, Finklestein SP, Ren J, Caggiano A. An optimized dosing regimen of cimaglermin (neuregulin 1β3, glial growth factor 2) enhances molecular markers of neuroplasticity and functional recovery after permanent ischemic stroke in rats. J Neurosci Res 2015; 94:253-65. [PMID: 26660233 PMCID: PMC4737294 DOI: 10.1002/jnr.23699] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/16/2015] [Accepted: 11/09/2015] [Indexed: 12/14/2022]
Abstract
Cimaglermin (neuregulin 1β3, glial growth factor 2) is a neuregulin growth factor family member in clinical development for chronic heart failure. Previously, in a permanent middle cerebral artery occlusion (pMCAO) rat stroke model, systemic cimaglermin treatment initiated up to 7 days after ischemia onset promoted recovery without reduced lesion volume. Presented here to extend the evidence are two studies that use a rat stroke model to evaluate the effects of cimaglermin dose level and dose frequency initiated 24 hr after pMCAO. Forelimb‐ and hindlimb‐placing scores (proprioceptive behavioral tests), body‐swing symmetry, and infarct volume were compared between treatment groups (n = 12/group). Possible mechanisms underlying cimaglermin‐mediated neurologic recovery were examined through axonal growth and synapse formation histological markers. Cimaglermin was evaluated over a wider dose range (0.02, 0.1, or 1.0 mg/kg) than doses previously shown to be effective but used the same dosing regimen (2 weeks of daily intravenous administration, then 1 week without treatment). The dose‐frequency study used the dose‐ranging study's most effective dose (1.0 mg/kg) to compare daily, once per week, and twice per week dosing for 3 weeks (then 1 week without treatment). Dose‐ and frequency‐dependent functional improvements were observed with cimaglermin without reduced lesion volume. Cimaglermin treatment significantly increased growth‐associated protein 43 expression in both hemispheres (particularly somatosensory and motor cortices) and also increased synaptophysin expression. These data indicate that cimaglermin enhances recovery after stroke. Immunohistochemical changes were consistent with axonal sprouting and synapse formation but not acute neuroprotection. Cimaglermin represents a potential clinical development candidate for ischemic stroke treatment. © 2015 The Authors. Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Tom J Parry
- Acorda Therapeutics, Inc., Ardsley, New York
| | | | | | | | | | | |
Collapse
|
23
|
Mendes-Ferreira P, Maia-Rocha C, Adão R, Mendes MJ, Santos-Ribeiro D, Alves BS, Cerqueira RJ, Castro-Chaves P, Lourenço AP, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Neuregulin-1 improves right ventricular function and attenuates experimental pulmonary arterial hypertension. Cardiovasc Res 2015; 109:44-54. [PMID: 26503987 DOI: 10.1093/cvr/cvv244] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/15/2015] [Indexed: 12/24/2022] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) is a serious disease that affects both the pulmonary vasculature and the right ventricle (RV). Current treatment options are insufficient. The cardiac neuregulin (NRG)-1/ErbB system is deregulated during heart failure, and treatment with recombinant human NRG-1 (rhNRG-1) has been shown to be beneficial in animal models and in patients with left ventricular (LV) dysfunction. This study aimed to evaluate the effects of rhNRG-1 in RV function and pulmonary vasculature in monocrotaline (MCT)-induced PAH and RV hypertrophy (RVH). METHODS AND RESULTS Male wistar rats (7- to 8-weeks old, n = 78) were injected with MCT (60 mg/kg, s.c.) or saline and treated with rhNRG-1 (40 µg/kg/day) or vehicle for 1 week, starting 2 weeks after MCT administration. Another set of animals was submitted to pulmonary artery banding (PAB) or sham surgery, and followed the same protocol. MCT administration resulted in the development of PAH, pulmonary arterial and RV remodelling, and dysfunction, and increased RV markers of cardiac damage. Treatment with rhNRG-1 attenuated RVH, improved RV function, and decreased RV expression of disease markers. Moreover, rhNRG-1 decreased pulmonary vascular remodelling and attenuated MCT-induced endothelial dysfunction. The anti-remodelling effects of rhNRG-1 were confirmed in the PAB model, where rhNRG-1 treatment was able to attenuate PAB-induced RVH. CONCLUSION rhNRG-1 treatment attenuates pulmonary arterial and RV remodelling, and dysfunction in a rat model of MCT-induced PAH and has direct anti-remodelling effects on the pressure-overloaded RV.
Collapse
Affiliation(s)
- Pedro Mendes-Ferreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carolina Maia-Rocha
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui Adão
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Maria José Mendes
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Diana Santos-Ribeiro
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Bárbara Silvana Alves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui João Cerqueira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Paulo Castro-Chaves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - André Pedro Lourenço
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | | | - Adelino Ferreira Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Carmen Brás-Silva
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, Cardiovascular Research and Development Centre, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
24
|
Abstract
Heart failure (HF) can rightfully be called the epidemic of the 21(st) century. Historically, the only available medical treatment options for HF have been diuretics and digoxin, but the capacity of these agents to alter outcomes has been brought into question by the scrutiny of modern clinical trials. In the past 4 decades, neurohormonal blockers have been introduced into clinical practice, leading to marked reductions in morbidity and mortality in chronic HF with reduced left ventricular ejection fraction (LVEF). Despite these major advances in pharmacotherapy, our understanding of the underlying disease mechanisms of HF from epidemiological, clinical, pathophysiological, molecular, and genetic standpoints remains incomplete. This knowledge gap is particularly evident with respect to acute decompensated HF and HF with normal (preserved) LVEF. For these clinical phenotypes, no drug has been shown to reduce long-term clinical event rates substantially. Ongoing developments in the pharmacotherapy of HF are likely to challenge our current best-practice algorithms. Novel agents for HF therapy include dual-acting neurohormonal modulators, contractility-enhancing agents, vasoactive and anti-inflammatory peptides, and myocardial protectants. These novel compounds have the potential to enhance our armamentarium of HF therapeutics.
Collapse
Affiliation(s)
- Thomas G von Lueder
- Department of Cardiology, Oslo University Hospital Ullevål, 0407 Oslo, Norway
| | - Henry Krum
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Alfred Hospital, Melbourne, VIC 3004, Australia
| |
Collapse
|
25
|
Gu A, Jie Y, Sun L, Zhao S, E M, You Q. RhNRG-1β Protects the Myocardium against Irradiation-Induced Damage via the ErbB2-ERK-SIRT1 Signaling Pathway. PLoS One 2015; 10:e0137337. [PMID: 26332771 PMCID: PMC4558028 DOI: 10.1371/journal.pone.0137337] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 08/15/2015] [Indexed: 12/17/2022] Open
Abstract
Radiation-induced heart disease (RIHD), which is a serious side effect of the radiotherapy applied for various tumors due to the inevitable irradiation of the heart, cannot be treated effectively using current clinical therapies. Here, we demonstrated that rhNRG-1β, an epidermal growth factor (EGF)-like protein, protects myocardium tissue against irradiation-induced damage and preserves cardiac function. rhNRG-1β effectively ameliorated irradiation-induced myocardial nuclear damage in both cultured adult rat-derived cardiomyocytes and rat myocardium tissue via NRG/ErbB2 signaling. By activating ErbB2, rhNRG-1β maintained mitochondrial integrity, ATP production, respiratory chain function and the Krebs cycle status in irradiated cardiomyocytes. Moreover, the protection of irradiated cardiomyocytes and myocardium tissue by rhNRG-1β was at least partly mediated by the activation of the ErbB2-ERK-SIRT1 signaling pathway. Long-term observations further showed that rhNRG-1β administered in the peri-irradiation period exerts continuous protective effects on cardiac pump function, the myocardial energy metabolism, cardiomyocyte volume and interstitial fibrosis in the rats receiving radiation via NRG/ErbB2 signaling. Our findings indicate that rhNRG-1β can protect the myocardium against irradiation-induced damage and preserve cardiac function via the ErbB2-ERK-SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Anxin Gu
- Department of Radiotherapy, the Affiliated Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yamin Jie
- Department of Radiotherapy, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Liang Sun
- Department of Human Anatomy, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuping Zhao
- Department of Radiotherapy, the Affiliated Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Mingyan E
- Department of Radiotherapy, the Affiliated Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- * E-mail: (QY); (ME)
| | - Qingshan You
- Department of Radiotherapy, the Affiliated Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- * E-mail: (QY); (ME)
| |
Collapse
|
26
|
Yin HK, Li XY, Jiang ZG, Zhou MD. Progress in neuregulin/ErbB signaling and chronic heart failure. World J Hypertens 2015; 5:63-73. [DOI: 10.5494/wjh.v5.i2.63] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 03/10/2015] [Accepted: 04/20/2015] [Indexed: 02/06/2023] Open
Abstract
Heart failure is one of the leading causes of death today. It is a complex clinical syndrome in which the heart has a reduced contraction ability and decreased viable myocytes. Novel approaches to the clinical management of heart failure have been achieved through an understanding of the molecular pathways necessary for normal heart development. Neuregulin-1 (NRG-1) has emerged as a potential therapeutic target based on the fact that mice null for NRG-1 or receptors mediating its activity, ErbB2 and ErbB4, are embryonic lethal and exhibit severe cardiac defects. Preclinical studies performed with animal models of heart failure demonstrate that treatment with NRG-1 significantly improves heart function and survival. Clinical data further support NRG-1 as a promising drug candidate for the treatment of cardiac dysfunction in patients. Recent studies have revealed the mechanism underlying the therapeutic effects of NRG-1/ErbB signaling in the treatment of heart failure. Through activation of upstream signaling molecules such as phosphoinositide 3-kinase, mitogen-activated protein kinase, and focal adhesion kinase, NRG-1/ErbB pathway activation results in increased cMLCK expression and enhanced intracellular calcium cycling. The former is a regulator of the contractile machinery, and the latter triggers cell contraction and relaxation. In addition, NRG-1/ErbB signaling also influences energy metabolism and induces epigenetic modification in cardiac myocytes in a way that more closely resembles healthy heart. These observations reveal potentially new treatment options for heart failure.
Collapse
|
27
|
Rupert CE, Coulombe KL. The roles of neuregulin-1 in cardiac development, homeostasis, and disease. Biomark Insights 2015; 10:1-9. [PMID: 25922571 PMCID: PMC4395047 DOI: 10.4137/bmi.s20061] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/01/2015] [Accepted: 03/04/2015] [Indexed: 02/07/2023] Open
Abstract
Neuregulin-1 (NRG-1) and its signaling receptors, erythroblastic leukemia viral oncogene homologs (ErbB) 2, 3, and 4, have been implicated in both cardiomyocyte development and disease, as well as in homeostatic cardiac function. NRG-1/ErbB signaling is involved in a multitude of cardiac processes ranging from myocardial and cardiac conduction system development to angiogenic support of cardiomyocytes, to cardioprotective effects upon injury. Numerous studies of NRG-1 employ a variety of platforms, including in vitro assays, animal models, and human clinical trials, with equally varying and, sometimes, contradictory outcomes. NRG-1 has the potential to be used as a therapeutic tool in stem cell therapies, tissue engineering applications, and clinical diagnostics and treatment. This review presents a concise summary of the growing body of literature to highlight the temporally persistent significance of NRG-1/ErbB signaling throughout development, homeostasis, and disease in the heart, specifically in cardiomyocytes.
Collapse
Affiliation(s)
- Cassady E Rupert
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen Lk Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA. ; Department of Molecular Pharmacology, Physiology and Biotechnology, Division of Biology and Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
28
|
Burnett AL, Sezen SF, Hoke A, Caggiano AO, Iaci J, Lagoda G, Musicki B, Bella AJ. GGF2 is neuroprotective in a rat model of cavernous nerve injury-induced erectile dysfunction. J Sex Med 2015; 12:897-905. [PMID: 25639458 DOI: 10.1111/jsm.12834] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Erectile dysfunction is a major complication of radical prostatectomy, commonly associated with penile neuropathy. In animal models of peripheral nerve injury, glial growth factor-2 (GGF2), a member of the neuregulin family of growth factors, has neuroprotective and neurorestorative properties, but this potential has not been established after cavernous nerve (CN) injury. AIMS The effectiveness of GGF2 in preserving axonal integrity and recovering erectile function in a rat model of radical prostatectomy-associated CN injury. METHODS Adult male Sprague-Dawley rats underwent bilateral CN crush injury (BCNI) or sham surgery. Rats were administered GGF2 (0.5, 5, or 15 mg/kg) or vehicle subcutaneously 24 hour pre and 24-hour post-BCNI, and once weekly for 5 weeks. Erectile function was assessed in response to electrical stimulation of the CN. CN survival was assessed by fluorogold retrograde axonal tracing in major pelvic ganglia (MPG). Unmyelinated axons in the CNs were quantitated by electron microscopy. MAIN OUTCOME MEASURES Erectile function recovery, CN survival, and unmyelinated CN axon preservation in response to GGF2 treatment following BCNI. RESULTS Erectile function was decreased (P < 0.05) after BCNI, and it was improved (P < 0.05) by all doses of GGF2. The number of fluorogold-labeled cells in the MPG was reduced (P < 0.05) by BCNI and was increased (P < 0.05) by GGF2 (0.5 and 5 mg/kg). The percentage of denervated Schwann cells in the BCNI group was higher (P < 0.05) than that in the sham-treated group and was decreased (P < 0.05) in the GGF2-treated (5 mg/kg) BCNI group. In the BCNI + GGF2 (5 mg/kg) group, the unmyelinated fiber histogram demonstrated a rightward shift, indicating an increased number of unmyelinated axons per Schwann cell compared with the BCNI group. CONCLUSIONS GGF2 promotes erectile function recovery following CN injury in conjunction with preserving unmyelinated CN fibers. Our findings suggest the clinical opportunity to develop GGF2 as a neuroprotective therapy for radical prostatectomy.
Collapse
Affiliation(s)
- Arthur L Burnett
- Department of Urology, The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Galindo CL, Kasasbeh E, Murphy A, Ryzhov S, Lenihan S, Ahmad FA, Williams P, Nunnally A, Adcock J, Song Y, Harrell FE, Tran TL, Parry TJ, Iaci J, Ganguly A, Feoktistov I, Stephenson MK, Caggiano AO, Sawyer DB, Cleator JH. Anti-remodeling and anti-fibrotic effects of the neuregulin-1β glial growth factor 2 in a large animal model of heart failure. J Am Heart Assoc 2014; 3:e000773. [PMID: 25341890 PMCID: PMC4323814 DOI: 10.1161/jaha.113.000773] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Neuregulin-1β (NRG-1β) is a growth factor critical for cardiac development and repair with therapeutic potential for heart failure. We previously showed that the glial growth factor 2 (GGF2) isoform of NRG-1β improves cardiac function in rodents after myocardial infarction (MI), but its efficacy in a large animal model of cardiac injury has not been examined. We therefore sought to examine the effects of GGF2 on ventricular remodeling, cardiac function, and global transcription in post-MI swine, as well as potential mechanisms for anti-remodeling effects. METHODS AND RESULTS MI was induced in anesthetized swine (n=23) by intracoronary balloon occlusion. At 1 week post-MI, survivors (n=13) received GGF2 treatment (intravenous, biweekly for 4 weeks; n=8) or were untreated (n=5). At 5 weeks post-MI, fractional shortening was higher (32.8% versus 25.3%, P=0.019), and left ventricular (LV) end-diastolic dimension lower (4.5 versus 5.3 cm, P=0.003) in GGF2-treated animals. Treatment altered expression of 528 genes, as measured by microarrays, including collagens, basal lamina components, and matricellular proteins. GGF2-treated pigs exhibited improvements in LV cardiomyocyte mitochondria and intercalated disk structures and showed less fibrosis, altered matrix structure, and fewer myofibroblasts (myoFbs), based on trichrome staining, electron microscopy, and immunostaining. In vitro experiments with isolated murine and rat cardiac fibroblasts demonstrate that NRG-1β reduces myoFbs, and suppresses TGFβ-induced phospho-SMAD3 as well as αSMA expression. CONCLUSIONS These results suggest that GGF2/NRG-1β prevents adverse remodeling after injury in part via anti-fibrotic effects in the heart.
Collapse
Affiliation(s)
- Cristi L Galindo
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Ehab Kasasbeh
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Abigail Murphy
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Sergey Ryzhov
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Sean Lenihan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Farhaan A Ahmad
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Philip Williams
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Amy Nunnally
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Jamie Adcock
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Yanna Song
- Department of Biostatistics, Vanderbilt University, Nashville, TN (Y.S., F.E.H.)
| | - Frank E Harrell
- Department of Biostatistics, Vanderbilt University, Nashville, TN (Y.S., F.E.H.)
| | - Truc-Linh Tran
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - Tom J Parry
- Acorda Therapeutics, Ardsley, NY (T.J.P., J.I., A.G., A.O.C.)
| | - Jen Iaci
- Acorda Therapeutics, Ardsley, NY (T.J.P., J.I., A.G., A.O.C.)
| | | | - Igor Feoktistov
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | | | | | - Douglas B Sawyer
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.L.G., E.K., A.M., S.R., S.L., F.A.A., P.W., A.N., J.A., T.L.T., I.F., D.B.S.)
| | - John H Cleator
- Department of Pharmacology, Vanderbilt University, Nashville, TN (J.H.C.)
| |
Collapse
|
30
|
Abstract
The beta isoform of Neuregulin-1 (NRG-1β), along with its receptors (ErbB2-4), is required for cardiac development. NRG-1β, as well as the ErbB2 and ErbB4 receptors, is also essential for maintenance of adult heart function. These observations have led to its evaluation as a therapeutic for heart failure. Animal studies and ongoing clinical trials have demonstrated beneficial effects of two forms of recombinant NRG-1β on cardiac function. In addition to the possible role for recombinant NRG-1βs as heart failure therapies, endogenous NRG-1β/ErbB signaling appears to play a role in restoring cardiac function after injury. The potential mechanisms by which NRG-1β may act as both a therapy and a mediator of reverse remodeling remain incompletely understood. In addition to direct effects on cardiac myocytes NRG-1β acts on the vasculature, interstitium, cardiac fibroblasts, and hematopoietic and immune cells, which, collectively, may contribute to NRG-1β's role in maintaining cardiac structure and function, as well as mediating reverse remodeling.
Collapse
|
31
|
Parodi EM, Kuhn B. Signalling between microvascular endothelium and cardiomyocytes through neuregulin. Cardiovasc Res 2014; 102:194-204. [PMID: 24477642 PMCID: PMC3989448 DOI: 10.1093/cvr/cvu021] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/23/2013] [Accepted: 01/10/2014] [Indexed: 12/26/2022] Open
Abstract
Heterocellular communication in the heart is an important mechanism for matching circulatory demands with cardiac structure and function, and neuregulins (Nrgs) play an important role in transducing this signal between the hearts' vasculature and musculature. Here, we review the current knowledge regarding Nrgs, explaining their roles in transducing signals between the heart's microvasculature and cardiomyocytes. We highlight intriguing areas being investigated for developing new, Nrg-mediated strategies to heal the heart in acquired and congenital heart diseases, and note avenues for future research.
Collapse
Affiliation(s)
| | - Bernhard Kuhn
- Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Enders Building, Room 1212, Brookline, MA 02115, USA
| |
Collapse
|
32
|
Abstract
A number of new and innovative approaches for repairing damaged myocardium are currently undergoing investigation, with several encouraging results. In addition to the progression of stem cell-based approaches and gene therapy/silencing methods, evidence continues to emerge that protein therapeutics may be used to directly promote cardiac repair and even regeneration. However, proteins are often limited in their therapeutic potential by short local half-lives and insufficient bioavailability and bioactivity, and many academic laboratories studying cardiovascular diseases are more comfortable with molecular and cellular biology than with protein biochemistry. Protein engineering has been used broadly to overcome weaknesses traditionally associated with protein therapeutics and has the potential to specifically enhance the efficacy of molecules for cardiac repair. However, protein engineering as a strategy has not yet been used in the development of cardiovascular therapeutics to the degree that it has been used in other fields. In this review, we discuss the role of engineered proteins in cardiovascular therapies to date. Further, we address the promise of applying emerging protein engineering technologies to cardiovascular medicine and the barriers that must be overcome to enable the ultimate success of this approach.
Collapse
Affiliation(s)
- Steven M Jay
- From the Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|