Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Exp Neurol ; 345: 113817, 2021 11.
Article in English | MEDLINE | ID: mdl-34314724

ABSTRACT

Spinal cord injury (SCI) causes sensorimotor and autonomic impairment that partly reflects extensive, permanent loss of neurons at the epicenter and penumbra of the injury. Strategies aimed at enhancing neuronal protection are critical to attenuate neurodegeneration and improve neurological recovery after SCI. In rat SCI, we previously uncovered that the tissue levels of neuregulin-1beta 1 (Nrg-1ß1) are acutely and persistently downregulated in the injured spinal cord. Nrg-1ß1 is well-known for its critical roles in the development, maintenance and physiology of neurons and glia in the developing and adult spinal cord. However, despite this pivotal role, Nrg-1ß1 specific effects and mechanisms of action on neuronal injury remain largely unknown in SCI. In the present study, using a clinically-relevant model of compressive/contusive SCI in rats and an in vitro model of glutamate toxicity in primary neurons, we demonstrate Nrg-1ß1 provides early neuroprotection through attenuation of reactive oxygen species, lipid peroxidation, necrosis and apoptosis in acute and subacute stages of SCI. Mechanistically, availability of Nrg-1ß1 following glutamate challenge protects neurons from caspase-dependent and independent cell death that is mediated by modulation of mitochondria associated apoptotic cascades and MAP kinase and AKT signaling pathways. Altogether, our work provides novel insights into the role and mechanisms of Nrg-1ß1 in neuronal injury after SCI and introduces its potential as a new neuroprotective target for this debilitating neurological condition.


Subject(s)
Caspases/metabolism , Glutamic Acid/toxicity , Neuregulin-1/metabolism , Neuroprotection/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/prevention & control , Animals , Cells, Cultured , Female , Neuregulin-1/administration & dosage , Neuroprotection/drug effects , Rats , Rats, Sprague-Dawley
2.
PLoS One ; 14(9): e0222587, 2019.
Article in English | MEDLINE | ID: mdl-31560696

ABSTRACT

Adipose-derived mesenchymal stem cells (AdMSCs) have been reported to ameliorate neurological deficits after acute ischemic stroke. As neuregulin 1 (NRG1, or heregulin 1), a growth factor with versatile functions in the central nervous system, has demonstrated protective effects against ischemic brain injuries, we have generated NRG1-overexpressing AdMSCs in order to investigate whether NRG1-AdMSCs could enhance therapeutic benefits of AdMSCs in ischemic stroke. After AdMSCs were infected with adenoviral NRG1, increased NRG1 secretion in NRG1-AdMSCs was confirmed with ELISA. At 1 d after ischemic stroke that was induced by the occlusion of middle cerebral artery (MCAo) for 60 min in Sprague Dawley (SD) rats, adenoviral NRG1, AdMSCs, NRG1-AdMSCs, or PBS were injected into the striatum and serial neurologic examinations were performed. Administration of NRG1-AdMSCs resulted in significant improvement of functional outcome following stroke compared to AdMSCs- or adenoviral NRG1-treated group, in addition to the reduction in the infarct size evaluated by hematoxylin and eosin staining. When NRG1 expression in the brain was examined by double immunofluorescence to human nuclei (HuNu)/NRG1 and ELISA, NRG1-AdMSCs demonstrated marked increase in NRG1 expression. Moreover, western blot analysis further showed that transplantation of NRG1-AdMSCs significantly increased both endogenous and adenoviral NRG1 expression compared to AdMSCs-treated group. To elucidate molecular mechanisms, NRG1-associated downstream molecules were evaluated by western blot analysis. Expression of ErbB4, a receptor for NRG1, was markedly increased by NRG1-AdMSCs administration, in addition to pMAPK and pAkt, crucial molecules of NRG1-ErbB4 signaling. Taken together, our data suggest that NRG1-AdMSCs can provide excellent therapeutic potential in ischemic stroke by activating NRG1-ErbB4 signaling network.


Subject(s)
Adipose Tissue/cytology , Brain Ischemia/therapy , Mesenchymal Stem Cell Transplantation/methods , Neuregulin-1/therapeutic use , Stroke/therapy , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , MAP Kinase Signaling System , Male , Neuregulin-1/administration & dosage , Neuregulin-1/metabolism , Rats , Rats, Sprague-Dawley
3.
Pharmazie ; 74(4): 231-234, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30940307

ABSTRACT

Aims: Neuregulin-1 (NRG-1) is a member of the epidermal growth factor family, and has an important role in cardiomyocyte development and myocardial regeneration. The aim of this study was to determine the protective effect of NRG-1 on cardiac electrical conduction in a rat myocardial infarction (MI) model. Methods: Thirty-three adult male SPF SD rats were randomized into three groups: sham-operated (n=9), acute myocardial infarction (AMI, n=12), and the NRG-1-treated (NRG-1, n=12) groups. All rats were sacrificed on day 8 after inducing MI. The 6-lead electrocardiograms (ECG) were recorded pre-operatively and eight days after operation, and analyzed. The expression levels of matrix metalloproteinase (MMP)-9 and gap junction protein connexin 43 (Cx43) in the infarcted myocardium were measured by Western blotting, and its in-situ distribution was evaluated using immunohistochemistry. Results: The PR, QRS and QT intervals were significantly prolonged in the AMI group compared to the sham operated animals (P<0.05, P<0.01 and P<0.01 respectively), and the PR and QRS intervals were partially restored in the NRG-1-treated rats (P<0.01 and P<0.01 compared to AMI group). Similarly, the increased levels of MMP-9 in the AMI group was restored upon NRG-1 treatment. The myocardial expression of Cx43 was decreased significantly in the AMI group, and was upregulated by NRG-1 treatment. Conclusions: NRG-1 attenuates MI-induced dysfunctional cardiac electrical conduction by downregulating MMP-9 and upregulating Cx43.


Subject(s)
Connexin 43/genetics , Matrix Metalloproteinase 9/genetics , Myocardial Infarction/physiopathology , Neuregulin-1/metabolism , Animals , Blotting, Western , Disease Models, Animal , Down-Regulation , Electrocardiography , Male , Neuregulin-1/administration & dosage , Rats , Rats, Sprague-Dawley , Up-Regulation
4.
J Drug Target ; 27(5-6): 573-581, 2019.
Article in English | MEDLINE | ID: mdl-30289743

ABSTRACT

Neuregulin-1 loaded poly(lactic-co-glycolic acid) (PLGA) microparticles hold great promise for treating acute myocardial infarction, as they have been proved to recover heart function and induce positive heart remodelling in preclinical studies. More recently, the inflammatory response of the heart after acute myocardial infarction (AMI) has been identified as one of the major mechanisms in cardiac tissue remodelling and repair. However, the connection between neuregulin-1 PLGA microparticles and inflammation is still not well characterised. In the present study we assessed this relationship in a mouse AMI model. First, in vitro evidence indicated that neuregulin-1 PLGA microparticles induced a macrophage polarisation toward a regenerative phenotype (CD206+ cells), preventing macrophages from evolving toward the inflammatory phenotype (B7-2+ cells). This correlated with in vivo experiments, where neuregulin-1 PLGA microparticles locally improved the CD206+/B7-2+ ratio. Moreover, neuregulin-1 PLGA microparticles were administered at different time points (15 min, 24, 72 and 168 h) after infarction induction without causing secondary inflammatory issues. The time of treatment administration did not alter the inflammatory response. Taken together, these results suggest that neuregulin-1 PLGA microparticles can be administered depending on the therapeutic window of the encapsulated drug and that they enhance the heart's reparative inflammatory response after acute myocardial infarction, helping cardiac tissue repair.


Subject(s)
Drug Carriers/chemistry , Heart/physiology , Macrophages/drug effects , Myocardial Infarction/drug therapy , Neuregulin-1/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Regeneration , Animals , Cell Line , Disease Models, Animal , Heart/drug effects , Heart/physiopathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neuregulin-1/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use
5.
JCI Insight ; 3(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29515030

ABSTRACT

Neuregulins (NRGs) are emerging as an important family of signaling ligands that regulate glucose and lipid homeostasis. NRG1 lowers blood glucose levels in obese mice, whereas the brown fat-enriched secreted factor NRG4 protects mice from high-fat diet-induced insulin resistance and hepatic steatosis. However, the therapeutic potential of NRGs remains elusive, given the poor plasma half-life of the native ligands. Here, we engineered a fusion protein using human NRG1 and the Fc domain of human IgG1 (NRG1-Fc) that exhibited extended half-life in circulation and improved potency in receptor signaling. We evaluated its efficacy in improving metabolic parameters and dissected the mechanisms of action. NRG1-Fc treatment triggered potent AKT activation in the liver, lowered blood glucose, improved insulin sensitivity, and suppressed food intake in obese mice. NRG1-Fc acted as a potent secretagogue for the metabolic hormone FGF21; however, the latter was largely dispensable for its metabolic effects. NRG1-Fc directly targeted the hypothalamic POMC neurons to promote membrane depolarization and increase firing rate. Together, NRG1-Fc exhibits improved pharmacokinetic properties and exerts metabolic benefits through dual inhibition of hepatic gluconeogenesis and caloric intake.


Subject(s)
Energy Intake/drug effects , Fatty Liver/drug therapy , Gluconeogenesis/drug effects , Neuregulin-1/administration & dosage , Obesity/drug therapy , Recombinant Fusion Proteins/administration & dosage , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/metabolism , Half-Life , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/administration & dosage , Immunoglobulin G/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Neuregulin-1/genetics , Neuregulin-1/pharmacokinetics , Obesity/etiology , Obesity/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Treatment Outcome
6.
J Neuroinflammation ; 15(1): 53, 2018 Feb 21.
Article in English | MEDLINE | ID: mdl-29467001

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) triggers a robust neuroinflammatory response that governs secondary injury mechanisms with both degenerative and pro-regenerative effects. Identifying new immunomodulatory therapies to promote the supportive aspect of immune response is critically needed for the treatment of SCI. We previously demonstrated that SCI results in acute and permanent depletion of the neuronally derived Neuregulin-1 (Nrg-1) in the spinal cord. Increasing the dysregulated level of Nrg-1 through acute intrathecal Nrg-1 treatment enhanced endogenous cell replacement and promoted white matter preservation and functional recovery in rat SCI. Moreover, we identified a neuroprotective role for Nrg-1 in moderating the activity of resident astrocytes and microglia following injury. To date, the impact of Nrg-1 on immune response in SCI has not yet been investigated. In this study, we elucidated the effect of systemic Nrg-1 therapy on the recruitment and function of macrophages, T cells, and B cells, three major leukocyte populations involved in neuroinflammatory processes following SCI. METHODS: We utilized a clinically relevant model of moderately severe compressive SCI in female Sprague-Dawley rats. Nrg-1 (2 µg/day) or saline was delivered subcutaneously through osmotic mini-pumps starting 30 min after SCI. We conducted flow cytometry, quantitative real-time PCR, and immunohistochemistry at acute, subacute, and chronic stages of SCI to investigate the effects of Nrg-1 treatment on systemic and spinal cord immune response as well as cytokine, chemokine, and antibody production. RESULTS: We provide novel evidence that Nrg-1 promotes a pro-regenerative immune response after SCI. Bioavailability of Nrg-1 stimulated a regulatory phenotype in T and B cells and augmented the population of M2 macrophages in the spinal cord and blood during the acute and chronic stages of SCI. Importantly, Nrg-1 fostered a more balanced microenvironment in the injured spinal cord by attenuating antibody deposition and expression of pro-inflammatory cytokines and chemokines while upregulating pro-regenerative mediators. CONCLUSION: We provide the first evidence of a significant regulatory role for Nrg-1 in neuroinflammation after SCI. Importantly, the present study establishes the promise of systemic Nrg-1 treatment as a candidate immunotherapy for traumatic SCI and other CNS neuroinflammatory conditions.


Subject(s)
Immunity, Cellular/drug effects , Neuregulin-1/administration & dosage , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/physiology , Female , Immunity, Cellular/physiology , Infusions, Subcutaneous , Macrophages/drug effects , Macrophages/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology
7.
Glia ; 66(3): 538-561, 2018 03.
Article in English | MEDLINE | ID: mdl-29148104

ABSTRACT

Oligodendroglial cell death and demyelination are hallmarks of neurotrauma and multiple sclerosis that cause axonal damage and functional impairments. Remyelination remains a challenge as the ability of endogenous precursor cells for oligodendrocyte replacement is hindered in the unfavorable milieu of demyelinating conditions. Here, in a rat model of lysolecithin lysophosphatidyl-choline (LPC)-induced focal demyelination, we report that Neuregulin-1 (Nrg-1), an important factor for oligodendrocytes and myelination, is dysregulated in demyelinating lesions and its bio-availability can promote oligodendrogenesis and remyelination. We delivered recombinant human Nrg-1ß1 (rhNrg-1ß1) intraspinally in the vicinity of LPC demyelinating lesion in a sustained manner using poly lactic-co-glycolic acid microcarriers. Availability of Nrg-1 promoted generation and maturation of new oligodendrocytes, and accelerated endogenous remyelination by both oligodendrocyte and Schwann cell populations in demyelinating foci. Importantly, Nrg-1 enhanced myelin thickness in newly remyelinated spinal cord axons. Our complementary in vitro studies also provided direct evidence that Nrg-1 significantly promotes maturation of new oligodendrocytes and facilitates their transition to a myelinating phenotype. Nrg-1 therapy remarkably attenuated the upregulated expression chondroitin sulfate proteoglycans (CSPGs) specific glycosaminoglycans in the extracellular matrix of demyelinating foci and promoted interleukin-10 (IL-10) production by immune cells. CSPGs and IL-10 are known to negatively and positively regulate remyelination, respectively. We found that Nrg-1 effects are mediated through ErbB2 and ErbB4 receptor activation. Our work provides novel evidence that dysregulated levels of Nrg-1 in demyelinating lesions of the spinal cord pose a challenge to endogenous remyelination, and appear to be an underlying cause of myelin thinning in newly remyelinated axons.


Subject(s)
Demyelinating Diseases/therapy , Immunomodulation , Neuregulin-1/administration & dosage , Neuroprotective Agents/administration & dosage , Remyelination/physiology , Spinal Cord/immunology , Animals , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Drug Carriers , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Humans , Lactic Acid , Male , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Oligodendroglia/immunology , Oligodendroglia/pathology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Spinal Cord/pathology , Spinal Cord Diseases/immunology , Spinal Cord Diseases/pathology , Spinal Cord Diseases/therapy
8.
J Card Fail ; 23(12): 887-899, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28870731

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)-1ß 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-1ß signaling via exogenous administration of recombinant NRG-1ß 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-1ß (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-1ß-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-1ß 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-1ß may be a promising therapeutic for HF post-MI in the setting of type 1 DM.


Subject(s)
Apoptosis/drug effects , Diabetes Mellitus, Type 1/drug therapy , Heart Failure/drug therapy , Myocardial Infarction/drug therapy , Neuregulin-1/administration & dosage , Oxidants/antagonists & inhibitors , Animals , Antioxidants/administration & dosage , Apoptosis/physiology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Progression , Drug Administration Schedule , Fibrosis , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Oxidants/metabolism , Rats , Rats, Sprague-Dawley
9.
J Control Release ; 261: 147-162, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28668379

ABSTRACT

Spinal cord injury (SCI) results in significant tissue damage that underlies functional impairments. Pharmacological interventions to confer neuroprotection and promote cell replacement are essential for SCI repair. We previously reported that Neuregulin-1 (Nrg-1) is acutely and permanently downregulated after SCI. Nrg-1 is a critical growth factor for differentiation of neural precursor cells (NPCs) into myelinating oligodendrocytes. We showed that intrathecal delivery of Nrg-1 enhances oligodendrocyte replacement following SCI. While an effective delivery system, intrathecal and systemic administration of growth factors with diverse biological targets may pose adverse off-target effects. Here, we have developed and optimized an injectable biodegradable poly(lactic-co-glycolic acid) (PLGA) microparticles system for sustained and prolonged intraspinal delivery of Nrg-1 in SCI. Recombinant human Nrg-1ß1 peptide was encapsulated into PLGA microparticles. Optimal Nrg-1 release rate and duration were achieved by manipulating the porosity and size of PLGA particles. Our in vitro analysis showed a direct correlation between particle size and porosity with Nrg-1 release rate, while Nrg-1 loading efficiency in PLGA microparticles was inversely correlated with particle porosity. In SCI, local intraspinal injection of PLGA-Nrg-1 microparticles maintained significantly higher tissue levels of Nrg-1 for a long-term duration compared to Nrg-1 delivered intrathecally by osmotic pumps. Bioactivity of Nrg-1 in PLGA microparticles was verified by promoting oligodendrocyte differentiation of NPCs in vitro, and preservation of oligodendrocytes and axons in SCI. PLGA-Nrg-1 also attenuated neuroinflammation and glial scarring following SCI. We show, for the first time, the feasibility, efficacy and safety of PLGA microparticle system for local and controlled administration of Nrg-1 in SCI.


Subject(s)
Drug Delivery Systems , Lactic Acid/chemistry , Neuregulin-1/administration & dosage , Polyglycolic Acid/chemistry , Spinal Cord Injuries/drug therapy , Animals , Cell Differentiation/drug effects , Delayed-Action Preparations , Disease Models, Animal , Drug Carriers/chemistry , Drug Liberation , Female , Humans , Injections, Spinal , Microspheres , Neuregulin-1/pharmacology , Oligodendroglia/cytology , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
11.
J Mol Neurosci ; 61(4): 607-616, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28265860

ABSTRACT

Neuregulin1ß (NRG1ß), a member of the excitomotor of tyrosine kinase receptor (erbB) family, was recently shown to play a neuroprotective role in cerebral ischemia-reperfusion injury. The present study analyzed the effects and its possible signaling pathway of NRG1ß on brain tissues after cerebral ischemia-reperfusion injury. A focal cerebral ischemic model was established by inserting a monofilament thread to achieve middle cerebral artery occlusion, followed by an NRG1ß injection via the internal carotid artery. NRG1ß injection resulted in significantly improved neurobehavioral activity according to the modified neurological severity score test. Tetrazolium chloridestaining revealed a smaller cerebral infarction volume; hematoxylin-eosin staining and transmission electron microscopy showed significantly alleviated neurodegeneration in the middle cerebral artery occlusion rats. Moreover, expression of phosphorylated MEK5, phosphorylated ERK5, and phosphorylated MEK2C increased after NRG1ß treatment, and the neuroprotective effect of NRG1ß was attenuated by an injection of the MEK5 inhibitor, BIX02189. Results from the present study demonstrate that NRG1ß provides neuroprotection following cerebral ischemia-reperfusion injury via the ERK5-dependent MAPK pathway.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , MAP Kinase Signaling System , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Infarction, Middle Cerebral Artery/metabolism , MAP Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase 5/metabolism , Male , Mitogen-Activated Protein Kinase 7/metabolism , Neuregulin-1/administration & dosage , Neuregulin-1/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar
13.
Biomed Pharmacother ; 85: 418-424, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27887848

ABSTRACT

Overexpression of the HER2 oncogene contributes to tumor angiogenesis, which is an essential hallmark of cancer. Simvastatin has been reported to exhibit antitumor activities in several cancers; however, its roles and molecular mechanismsin the regulation of colorectal angiogenesis remain to be clarified. Here, we show that colon cancer cells express high levels of VEGF, total HER2 and phosphorylated HER2, and simvastatin apparently decreased their expression in HER2-overexpressing colon cancer cells. Simvastatin pretreatment reduced endothelial tube formation in vitro and microvessel density in vivo. Furthermore, simvastatin markedly inhibited tumor angiogenesis even in the presence of heregulin (HRG)-ß1 (a HER2 co-activator) pretreatment in HER2+ tumor cells. Mechanistic investigation showed that simvastatin significantly abrogated HER2-induced tumor angiogenesis by inhibiting VEGF secretion. Together, these results provide a novel mechanism underlying the simvastatin-induced inhibition of tumor angiogenesis through regulating HER2/VEGF axis.


Subject(s)
Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Neovascularization, Pathologic/prevention & control , Receptor, ErbB-2/metabolism , Simvastatin/pharmacology , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Bevacizumab/administration & dosage , Bevacizumab/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Humans , Neuregulin-1/administration & dosage , Neuregulin-1/pharmacology , Receptor, ErbB-2/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
14.
Int J Pharm ; 523(2): 531-533, 2017 May 25.
Article in English | MEDLINE | ID: mdl-27838293

ABSTRACT

Neuregulin (NRG1) and fibroblast growth factor (FGF1) are well known growth factors implicated in cardiomyocyte proliferation and survival, as well as in angiogenesis, the development of adult heart and the maintenance of cardiac function. NRG1 and FGF1 have become promising therapeutic agents to treat myocardial infarction (MI) disorder. Unfortunately, clinical trials performed so far reported negative efficacy results, because growth factors are rapidly degraded and eliminated from the biological tissues once administered. In order to increase their bioavailability and favour their therapeutic effects, they have been combined with poly(lactic-co-glycolic acid) and polyethylene glycol microparticles (PLGA MPs and PEG-PLGA MPs). Here we compare both types of microparticles loaded with NRG1 or FGF1 in terms of efficacy in a rat MI model. Our results showed that intramyocardial injection of NRG1 or FGF1-loaded PLGA and PEG-PLGA MPs brought about similar improvements in the ejection fraction, angiogenesis and arteriogenesis after administration into the infarcted hearts. PEG coating did not add any effect regarding MP efficacy. Both PLGA and PEG-PLGA MPs were equally phagocyted in the heart. To our knowledge, this is the first study analysing the opsonisation process in heart tissue. The results allow us to conclude that the opsonisation process is different in heart tissue compared to blood.


Subject(s)
Fibroblast Growth Factor 1/administration & dosage , Lactic Acid/chemistry , Myocardial Infarction/therapy , Myocardium/pathology , Neuregulin-1/administration & dosage , Polyesters/chemistry , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Animals , Cytokines , Heart/physiopathology , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Regeneration
15.
Peptides ; 88: 196-207, 2017 02.
Article in English | MEDLINE | ID: mdl-27993557

ABSTRACT

Neuregulin-1 (NRG-1), an endogenously produced polypeptide, is the ligand of cardiomyocyte ErbB receptors, with cardiovascular protective effects. In the present study, we explored whether the cardioprotective effect of NRG-1 against I/R injury is mediated by inhibiting myocardial endoplasmic reticulum (ER) stress. In vitro, NRG-1 directly inhibited the upregulation of ER stress markers such as glucose-regulated protein 78, CCAAT/enhancer binding protein homologous protein and cleaved caspase-12 induced by the ER stress inducers tunicamycin or dithiothreitol in both neonatal and adult ventricular myocytes. Attenuating ErbB signals by an ErbB inhibitor AG1478 or ErbB4 knockdown and preincubation with phosphoinositide 3-kinase inhibitors all reversed the effect of NRG-1 inhibiting ER stress in cultured neonatal rat cardiomyocytes. Concurrently, cardiomyocyte ER stress and apoptosis induced by hypoxia-reoxygenation were decreased by NRG-1 treatment in vitro. Furthermore, in an in vivo rat model of myocardium ischemia/reperfusion (I/R), intravenous NRG-1 administration significantly decreased ER stress and myocardial infarct size induced by I/R. NRG-1 could protect the heart against I/R injury by inhibiting myocardial ER stress, which might be mediated by the phosphoinositide 3-kinase/Akt signaling pathway.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Neuregulin-1/genetics , Animals , Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/genetics , Cardiotonic Agents/administration & dosage , Caspase 12/genetics , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/genetics , Humans , Myocardial Infarction/chemically induced , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/chemically induced , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neuregulin-1/administration & dosage , Neuregulin-1/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects , Tunicamycin/toxicity
16.
Eur J Pharmacol ; 796: 76-89, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-27993643

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-1ß 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.


Subject(s)
Heart Ventricles/drug effects , Myocardial Infarction/physiopathology , Neuregulin-1/pharmacology , Ventricular Dysfunction/drug therapy , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Cytoprotection/drug effects , Doxorubicin/adverse effects , Drug Administration Schedule , Heart Failure/complications , Humans , Mice , Myocardial Infarction/complications , Neuregulin-1/administration & dosage , Neuregulin-1/chemistry , Neuregulin-1/metabolism , Rats , Receptor, ErbB-4/metabolism
17.
Sci Rep ; 6: 30467, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27469430

ABSTRACT

Adult hippocampal neurogenesis is associated with learning and affective behavioural regulation. Its diverse functionality is segregated along the septotemporal axis from the dorsal to ventral hippocampus. However, features distinguishing immature neurons in these regions have yet to be characterized. Additionally, although we have shown that administration of the neurotrophic factor neuregulin-1 (NRG1) selectively increases proliferation and overall neurogenesis in the mouse ventral dentate gyrus (DG), likely through ErbB3, NRG1's effects on intermediate neurogenic stages in immature neurons are unknown. We examined whether NRG1 administration increases DG ErbB3 phosphorylation. We labeled adultborn cells using BrdU, then administered NRG1 to examine in vivo neurogenic effects on immature neurons with respect to cell survival, morphology, and synaptogenesis. We also characterized features of immature neurons along the septotemporal axis. We found that neurogenic effects of NRG1 are temporally and subregionally specific to proliferation in the ventral DG. Particular morphological features differentiate immature neurons in the dorsal and ventral DG, and cytogenesis differed between these regions. Finally, we identified synaptic heterogeneity surrounding the granule cell layer. These results indicate neurogenic involvement of NRG1-induced antidepressant-like behaviour is particularly associated with increased ventral DG cell proliferation, and identify novel distinctions between dorsal and ventral hippocampal neurogenic development.


Subject(s)
Aging/metabolism , Cell Differentiation/drug effects , Hippocampus/cytology , Neuregulin-1/administration & dosage , Neuregulin-1/pharmacology , Neurogenesis/drug effects , Neurons/cytology , Animals , Cell Survival/drug effects , Dendrites/drug effects , Dendrites/metabolism , Dentate Gyrus/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Phosphorylation/drug effects , Receptor, ErbB-3/metabolism , Synapses/drug effects , Synapses/metabolism
18.
Neuroscience ; 329: 182-92, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27189883

ABSTRACT

The bed nucleus of the stria terminalis (BNST), a nucleus defined as part of the extended amygdala, is involved in the expression of anxiety disorders. However, the regulatory mechanisms of BNST inhibitory activity that is involved in anxiety are unknown. Here, we showed that blocking neuregulin 1 (NRG1)-ErbB4 signaling in the BNST of mice, by either neutralizing endogenous NRG1 with ecto-Erbb4 or antagonizing the ErbB4 receptor with its specific inhibitor, produced anxiogenic responses. Interestingly, application of exogenous NRG1 into the BNST induced no anxiolytic effects, suggesting saturating activity of endogenous NRG1. While infusion of the GABAA receptor antagonist bicuculline into the BNST also led to anxiety-related behaviors, it did not worsen the anxiogenic effects produced by blocking NRG1-ErbB4 signaling, suggesting possible involvement of GABAergic neurotransmission. Further, in vitro electrophysiological recordings showed that BNST NRG1-ErbB4 signaling regulated the presynaptic GABA release. Together, these results suggest that NRG1-ErbB4 signaling in the BNST may play an important role in regulating anxiety-like behaviors.


Subject(s)
Anxiety/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-4/metabolism , Septal Nuclei/metabolism , Animals , Bicuculline/pharmacology , Central Nervous System Agents/pharmacology , GABA-A Receptor Antagonists/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , Models, Animal , Neuregulin-1/administration & dosage , Neuregulin-1/antagonists & inhibitors , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Pyrimidines/pharmacology , Receptor, ErbB-4/antagonists & inhibitors , Receptors, GABA-A/metabolism , Septal Nuclei/drug effects , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
19.
PLoS One ; 11(5): e0155456, 2016.
Article in English | MEDLINE | ID: mdl-27175488

ABSTRACT

BACKGROUND: We previously developed and validated a strategy for stimulating heart regeneration by administration of recombinant neuregulin (rNRG1), a growth factor, in mice. rNRG1 stimulated proliferation of heart muscle cells, cardiomyocytes, and was most effective when administration began during the neonatal period. Our results suggested the use of rNRG1 to treat pediatric patients with heart failure. However, administration in this age group may stimulate growth outside of the heart. METHODS: NRG1 and ErbB receptor expression was determined by RT-PCR. rNRG1 concentrations in serum were quantified by ELISA. Mice that received protocols of recombinant neuregulin1-ß1 administration (rNRG1, 100 ng/g body weight, daily subcutaneous injection for the first month of life), previously shown to induce cardiac regeneration, were examined at pre-determined intervals. Somatic growth was quantified by weighing. Organ growth was quantified by MRI and by weighing. Neoplastic growth was examined by MRI, visual inspection, and histopathological analyses. Phospho-ERK1/2 and S6 kinase were analyzed with Western blot and ELISA, respectively. RESULTS: Lung, spleen, liver, kidney, brain, and breast gland exhibited variable expression of the NRG1 receptors ErbB2, ErbB3, ErbB4, and NRG1. Body weight and tibia length were not altered in mice receiving rNRG1. MRI showed that administration of rNRG1 did not alter the volume of the lungs, liver, kidneys, brain, or spinal cord. Administration of rNRG1 did not alter the weight of the lungs, spleen, liver, kidneys, or brain. MRI, visual inspection, and histopathological analyses showed no neoplastic growth. Follow-up for 6 months showed no alteration of somatic or organ growth. rNRG1 treatment increased the levels of phospho-ERK1/2, but not phospho-S6 kinase. CONCLUSIONS: Administration protocols of rNRG1 for stimulating cardiac regeneration in mice during the first month of life did not induce unwanted growth effects. Further studies may be required to determine whether this is the case in a corresponding human population.


Subject(s)
Aging/physiology , Heart/physiology , Morphogenesis/drug effects , Neoplasms/pathology , Neuregulin-1/administration & dosage , Neuregulin-1/pharmacology , Regeneration/drug effects , Animals , Animals, Newborn , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Heart/drug effects , Humans , Kidney/drug effects , Kidney/enzymology , Mice , Neuregulin-1/blood , Neuregulin-1/genetics , Organ Size/drug effects , Phosphorylation/drug effects , Recombinant Proteins/pharmacology , Ribosomal Protein S6 Kinases/metabolism
20.
Sci Rep ; 6: 25932, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27184924

ABSTRACT

Cardiovascular protein therapeutics such as neuregulin (NRG1) and acidic-fibroblast growth factor (FGF1) requires new formulation strategies that allow for sustained bioavailability of the drug in the infarcted myocardium. However, there is no FDA-approved injectable protein delivery platform due to translational concerns about biomaterial administration through cardiac catheters. We therefore sought to evaluate the efficacy of percutaneous intramyocardial injection of poly(lactic-co-glycolic acid) microparticles (MPs) loaded with NRG1 and FGF1 using the NOGA MYOSTAR injection catheter in a porcine model of ischemia-reperfusion. NRG1- and FGF1-loaded MPs were prepared using a multiple emulsion solvent-evaporation technique. Infarcted pigs were treated one week after ischemia-reperfusion with MPs containing NRG1, FGF1 or non-loaded MPs delivered via clinically-translatable percutaneous transendocardial-injection. Three months post-treatment, echocardiography indicated a significant improvement in systolic and diastolic cardiac function. Moreover, improvement in bipolar voltage and decrease in transmural infarct progression was demonstrated by electromechanical NOGA-mapping. Functional benefit was associated with an increase in myocardial vascularization and remodeling. These findings in a large animal model of ischemia-reperfusion demonstrate the feasibility and efficacy of using MPs as a delivery system for growth factors and provide strong evidence to move forward with clinical studies using therapeutic proteins combined with catheter-compatible biomaterials.


Subject(s)
Fibroblast Growth Factor 1/administration & dosage , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Neuregulin-1/administration & dosage , Administration, Cutaneous , Animals , Cell Line , Disease Models, Animal , Female , Heart Function Tests/drug effects , Injections , Lactic Acid , Male , Mice , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Swine , Swine, Miniature , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL