Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 84
1.
Diagn Pathol ; 19(1): 1, 2024 Jan 03.
Article En | MEDLINE | ID: mdl-38173003

BACKGROUND: NRG1 fusion is a promising therapeutic target for various tumors but its prevalence is extremely low, and there are no standardized testing algorithms for genetic assessment. MOTHODS: In this study, we analyzed 3008 tumors using Fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) to screen for NRG1 translocation and p-HER3 expression. RESULTS: Our results demonstrated no cases with p-HER3 positivity through IHC. Nonetheless, 29 cases (0.96%) were identified positive for NRG1 translocation through FISH, with three different signal types. FISH-positive cases were subsequently subjected to next-generation sequencing (NGS) testing. However, only eight of these cases were confirmed with NRG1 fusion through NGS. Notably, we divided FISH into three types and FISH type C group was consistent with NGS results. All NGS NRG1 fusion tumors were adenocarcinomas, with a higher prevalence in females. Our findings indicate that although FISH has limitations in screening NRG1 gene rearrangements, NRG1 fusions can be reliably detected with signals exhibiting low copy numbers of the 5'-end of the gene and no fusion signals. CONCLUSION: Considering the high cost of NGS, FISH remains a useful method for screening NRG1 fusions in various types of tumors. This study provides valuable insights into the molecular mechanisms of NRG1 fusion and identifies potential treatment targets for patients suffering from this disease.


Adenocarcinoma , Lung Neoplasms , Female , Humans , Lung Neoplasms/pathology , In Situ Hybridization, Fluorescence/methods , Adenocarcinoma/pathology , Translocation, Genetic , Gene Rearrangement , Oncogene Proteins, Fusion/genetics , Neuregulin-1/genetics , Neuregulin-1/therapeutic use
2.
J Mol Cell Cardiol ; 187: 26-37, 2024 02.
Article En | MEDLINE | ID: mdl-38150867

Stimulating cardiomyocyte proliferation in the adult heart has emerged as a promising strategy for cardiac regeneration following myocardial infarction (MI). The NRG1-ERBB4 signaling pathway has been implicated in the regulation of cardiomyocyte proliferation. However, the therapeutic potential of recombinant human NRG1 (rhNRG1) has been limited due to the low expression of ERBB4 in adult cardiomyocytes. Here, we investigated whether a fusion protein of rhNRG1 and an ERBB3 inhibitor (rhNRG1-HER3i) could enhance the affinity of NRG1 for ERBB4 and promote adult cardiomyocyte proliferation. In vitro and in vivo experiments were conducted using postnatal day 1 (P1), P7, and adult cardiomyocytes. Western blot analysis was performed to assess the expression and activity of ERBB4. Cardiomyocyte proliferation was evaluated using Ki67 and pH 3 immunostaining, while fibrosis was assessed using Masson staining. Our results indicate that rhNRG1-HER3i, but not rhNRG1, promoted P7 and adult cardiomyocyte proliferation. Furthermore, rhNRG1-HER3i improved cardiac function and reduced cardiac fibrosis in post-MI hearts. Administration of rhNRG1-HER3i inhibited ERBB3 phosphorylation while increasing ERBB4 phosphorylation in adult mouse hearts. Additionally, rhNRG1-HER3i enhanced angiogenesis following MI compared to rhNRG1. In conclusion, our findings suggest that rhNRG1-HER3i is a viable therapeutic approach for promoting adult cardiomyocyte proliferation and treating MI by enhancing NRG1-ERBB4 signaling pathway.


Cardiomyopathies , Myocardial Infarction , Mice , Animals , Humans , Signal Transduction , Myocytes, Cardiac/metabolism , Neuregulin-1/therapeutic use , Cardiomyopathies/metabolism , Receptor, ErbB-4/metabolism
3.
Cytokine ; 174: 156439, 2024 02.
Article En | MEDLINE | ID: mdl-38134557

Neuregulin (NRG)-1 plays fundamental roles in several organ systems after binding to its receptors, ErbB2 and ErbB4. This study examines the role of NRG-1 in atopic dermatitis (AD), a chronic skin disease that causes dryness, pruritus, and inflammation. In mice administered Der p 38, the skin presents AD-like symptoms including filaggrin downregulation and infiltration of neutrophils and eosinophils. Noticeably, there is an increased expression of NRG-1, ErbB2, and ErbB4 in the skin. Upregulation of these proteins is significantly correlated to the clinical skin severity score. In human keratinocyte HaCaT cells, exposure to Der p 38 decreased filaggrin expression, and NRG-1 alone had no effect on the expression. However, co-treatment of Der p 38 with NRG-1 enhanced the filaggrin expression decreased by Der p 38. Pre-treatment with AG879 (an ErbB2 inhibitor) or ErbB4 siRNA blocked the recovery of filaggrin expression in the cells after co-treatment with Der p 38 and NRG-1. Der p 38 treatment enhanced the secretion of interleukin-6 (IL-6), IL-8, and monocyte chemoattractant protein-1 (MCP-1). Co-treatment of Der p 38 with NRG-1 lowered the cytokine secretion increased by Der p 38, although NRG-1 alone was not effective on cytokine alteration. Neutrophil apoptosis was not altered by NRG-1 or supernatants of cells treated with NRG-1, but the cell supernatants co-treated with Der p 38 and NRG-1 blocked the anti-apoptotic effects of Der p 38-treated supernatants on neutrophils, which was involved in the activation of caspase 9 and caspase 3. Taken together, we determined that NRG-1 has anti-inflammatory effects in AD triggered by Der p 38. These results will pave the way to understanding the functions of NRG-1 and in the future development of AD treatment.


Dermatitis, Atopic , Mice , Animals , Humans , Dermatitis, Atopic/genetics , Filaggrin Proteins , Neuregulin-1/pharmacology , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , Keratinocytes/metabolism , Skin/metabolism , Cytokines/metabolism , Receptor, ErbB-4/metabolism , Receptor, ErbB-4/pharmacology , Anti-Inflammatory Agents/pharmacology
4.
Colloids Surf B Biointerfaces ; 220: 112936, 2022 Dec.
Article En | MEDLINE | ID: mdl-36265312

Cardiac patch, a scaffold layered on the surface of the heart that can provide mechanical and regeneration support for damaged myocardium, has provided a promising solution to treat severe myocardial infarction (MI). In this work, a fibrin based cardiac patch loaded with neuregulin-1 (NRG-1) is developed to attach locally to the infract area of heart. The composite patch exhibited good biocompatibility and promoted cardiomyocyte proliferation in vitro via NRG-1/ErbB signaling. Moreover, implantation of this patch to the infracted border zone reduced cell apoptosis, promoted angiogenesis and inhibited fibrosis, which reduced infraction size and improved cardiac function consequently. Thus, the combination of natural biomaterial fibrin and bioactive factor NRG-1 might have a promising potential for clinical application of MI treatment.


Myocardial Infarction , Neuregulin-1 , Tissue Scaffolds , Humans , Fibrin , Heart , Myocardial Infarction/drug therapy , Myocardium , Myocytes, Cardiac , Neuregulin-1/pharmacology , Neuregulin-1/therapeutic use
5.
Thorac Cancer ; 13(21): 3063-3067, 2022 11.
Article En | MEDLINE | ID: mdl-36096509

Neuregulin 1 (NRG1) gene fusion is a rare oncogenic driver gene in multiple tumor types, leading to the activation of the epidermal growth factor receptor (ErbB)-mediated pathway. Therefore, afatinib, a pan-ErbB family inhibitor, may be a therapeutic candidate for NRG1 fusion-driven tumors. In this case, we report a multiple primary lung adenocarcinoma patient harboring the CD74-NRG1 fusion, epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (ERBB2) mutation simultaneously. The patient received afatinib and pyrotinib combination therapy and showed a significant treatment response with a progression-free survival of 5 months. Our case further supports the use of targeted therapy for NRG1 fusion-positive non-small-cell lung cancer.


Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Neoplasms, Multiple Primary , Humans , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Afatinib/therapeutic use , Lung Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Mutation
6.
Future Oncol ; 18(26): 2865-2870, 2022 Aug.
Article En | MEDLINE | ID: mdl-35876504

WHAT IS THIS STUDY ABOUT?: This plain language summary reports the findings of a recent review of NRG1 fusion-positive tumors. WHAT ARE FUSIONS?: A gene fusion occurs when two genes join to create a new gene. This rearrangement of DNA can change the processes within normal cells and lead to cancer. One of these gene fusions involves the NRG1 gene. NRG1 fusions have been reported in several types of cancers. These are known as NRG1 fusion-positive cancers. WHAT TREATMENTS ARE AVAILABLE FOR PEOPLE WITH FUSION-POSITIVE CANCER?: One drug that has been studied in people with NRG1 fusion-positive cancer is called afatinib. People with several cancer types have received afatinib in clinical trials, and some people have responded to afatinib. Further studies are required to understand how effective afatinib and other treatments are for NRG1 fusion-positive cancer.


Lung Neoplasms , Afatinib/therapeutic use , Biology , Humans , Language , Lung Neoplasms/pathology , Neuregulin-1/genetics , Neuregulin-1/therapeutic use , Oncogene Proteins, Fusion/genetics
7.
Biol Psychiatry ; 92(5): 419-433, 2022 09 01.
Article En | MEDLINE | ID: mdl-35662508

BACKGROUND: Olanzapine is an effective antipsychotic medication for treatment-resistant schizophrenia (TRS); however, the therapeutic effectiveness of olanzapine has been found to vary in individual patients. It is imperative to unravel its resistance mechanisms and find reliable targets to develop novel precise therapeutic strategies. METHODS: Unbiased RNA sequencing analysis was performed using homogeneous populations of neural stem cells derived from induced pluripotent stem cells in 3 olanzapine responder (reduction of Positive and Negative Syndrome Scale score ≥25%) and 4 nonresponder (reduction of Positive and Negative Syndrome Scale score <25%) inpatients with TRS. We also used a genotyping study from patients with TRS to assess the candidate genes associated with the olanzapine response. CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9-mediated genome editing, neurologic behavioral tests, RNA silencing, and microRNA sequencing were used to investigate the phenotypic mechanisms of an olanzapine resistance gene in patients with TRS. RESULTS: Neuregulin-1 (NRG-1) deficiency-induced mitochondrial dysfunction is associated with olanzapine treatment outcomes in TRS. NRG-1 knockout mice showed schizophrenia-relevant behavioral deficits and yielded olanzapine resistance. Notably, miR143-3p is a critical NRG-1 target related to mitochondrial dysfunction, and miR143-3p levels in neural stem cells associate with severity to olanzapine resistance in TRS. Meanwhile, olanzapine resistance in NRG-1 knockout mice could be rescued by treatment with miR143-3p agomir via intracerebral injection. CONCLUSIONS: Our findings provide direct evidence of olanzapine resistance resulting from NRG-1 deficiency-induced mitochondrial dysfunction, and they link olanzapine resistance and NRG-1 deficiency-induced mitochondrial dysfunction to an NRG-1/miR143-3p axis, which constitutes a novel biomarker and target for TRS.


Antipsychotic Agents , Schizophrenia , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Humans , Mice , Mice, Knockout , Mitochondria , Neuregulin-1/genetics , Neuregulin-1/therapeutic use , Olanzapine/therapeutic use , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenia, Treatment-Resistant
8.
Future Oncol ; 18(18): 2193-2200, 2022 Jun.
Article En | MEDLINE | ID: mdl-35400204

WHAT IS THIS SUMMARY ABOUT?: This plain language summary reports the findings of a case series, a study which evaluated a small number of people who had a certain type of cancer. This case series looked at how well a drug called afatinib worked in people who have a rare type of cancer called neuregulin-1 (also called NRG1) gene fusion-positive cancer. Six people with NRG1 gene fusion-positive cancer were treated with afatinib, including five with lung cancer and one with gastrointestinal cancer. After treatment, people were monitored regularly to see if their tumors had grown (known as tumor progression). WHAT WERE THE RESULTS?: After being treated with afatinib for up to 16 months, two people had stable disease (meaning their cancer did not get worse or improve). Three people had a partial response (meaning they showed a decrease in the size of their tumor) for over 18 months. One person had a partial response after being treated with afatinib for 11 months. WHAT DO THE RESULTS MEAN?: Afatinib showed encouraging results which suggest it may be a potential treatment for NRG1 fusion-driven cancers, as it delayed tumor progression and decreased tumor size for some people with NRG1 gene fusion-positive tumors. These case reports warrant the conduct of a clinical trial including a larger number of people to definitively confirm how well afatinib works in treating people with NRG1 gene fusion-positive cancers.


Language , Lung Neoplasms , Afatinib/therapeutic use , Gene Fusion , Humans , Lung Neoplasms/drug therapy , Neuregulin-1/genetics , Neuregulin-1/therapeutic use , Oncogene Proteins, Fusion/genetics
9.
Biochim Biophys Acta Rev Cancer ; 1877(3): 188707, 2022 05.
Article En | MEDLINE | ID: mdl-35247506

It is widely established that chromosomal rearrangements induce oncogenesis in solid tumors. However, discovering chromosomal rearrangements that are targetable and actionable remains a difficulty. Targeting gene fusion or chromosomal rearrangement seems to be a powerful strategy to address malignancies characterized by gene rearrangement. Oncogenic NRG1 fusions are relatively rare drivers that infrequently occur across most tumor types. NRG1 fusions exhibit unique biological properties and are difficult to identify owing to their large intronic regions. NRG1 fusions can be detected using a variety of techniques, including fluorescence in situ hybridization, immunohistochemistry, or next-generation sequencing (NGS), with NGS-based RNA sequencing being the most sensitive. Previous studies have shown that NRG1 fusion protein induces tumorigenesis, and numerous therapies targeting the ErbB signaling pathway, such as ErbB kinase inhibitors and monoclonal antibodies, have initially demonstrated encouraging anticancer efficacy in malignant tumors carrying NRG1 fusions. In this review, we present the characteristics and prevalence of NRG1 fusions in solid tumors. Additionally, we discuss the laboratory approaches for diagnosing NRG1 gene fusions. More importantly, we outline promising strategies for treating malignancies with NRG1 fusion.


Lung Neoplasms , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/drug therapy , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , Oncogene Proteins, Fusion/genetics
10.
Expert Opin Ther Targets ; 25(10): 865-875, 2021 10.
Article En | MEDLINE | ID: mdl-34706602

INTRODUCTION: Fusions in neuregulin 1 (NRG1) and neuregulin 2 (NRG2) genes are molecular features of non-small cell lung cancer (NSCLC). These rearrangements enhance ectopic expression of the NRG/ErbB receptor-ligand and induce the triggering of downstream pathways. Evidence suggests the involvement of the NRG1/ErbB3 axis deregulation in the progression and treatment resistance of NSCLC cancer (NSCLC) and that NRG1 fusions are prognostic/predictive markers for targeted therapy. AREAS COVERED: Biological and prognostic/predictive value of NRG1 and NRG2 fusions in NSCLC and their related cellular pathways are described and discussed. Publications in English language, peer-reviewed, high-quality international journals were identified on PubMed, as well as scientific official sites were used to update the international clinical trials progress. EXPERT OPINION: NRG1 and NRG2 fusions should be considered as novel markers for biological therapy targeting ErbB2/ErbB3. There is evidence for the involvement of the NRG1/ErbB3 axis deregulation in cancer stem cell phenotype, tumor progression, and resistance to NSCLC therapy. Neuregulin fusions are very complex, hence many question marks must be tackled before translating these molecular lesions into clinical practice. Biology, and aggressiveness of the NRG1 and NRG2 fusions warrant further investigations.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Lung Neoplasms/drug therapy , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use
11.
Oxid Med Cell Longev ; 2020: 1720961, 2020.
Article En | MEDLINE | ID: mdl-32765805

Sepsis-induced diaphragm dysfunction (SIDD) which is mainly characterized by decrease in diaphragmatic contractility has been identified to cause great harms to patients. Therefore, there is an important and pressing need to find effective treatments for improving SIDD. In addition, acetylcholinesterase (AChE) activity is a vital property of the diaphragm, so we evaluated both diaphragmatic contractility and AChE activity. Though neuregulin-1ß (NRG-1ß) is known to exert organ-protective effects in some inflammatory diseases, little is known about the potential of NRG-1ß therapy in the diaphragm during sepsis. Our study was aimed at exploring the effects of NRG-1ß application on diaphragmatic contractility and AChE activity during sepsis. Proinflammatory cytokines, muscle injury biomarkers in serum, contractile force, AChE activity, proinflammatory cytokines, oxidative parameters, histological condition, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining, and expression of phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB/Akt) signaling proteins in the diaphragm were measured and compared between nonseptic and septic groups with or without NRG-1ß treatment. In vitro, the effects of NRG-1ß on reactive oxygen species (ROS) production in the lipopolysaccharide- (LPS-) stimulated L6 rat muscle skeletal cells with or without the Akt inhibitor MK-2206 were detected. NRG-1ß inhibited proinflammatory cytokine release and muscle injury biomarkers soaring in serum and improved the sepsis-induced diaphragm dysfunction and AChE activity decrease significantly during sepsis. Meanwhile, the inflammatory response, oxidative stress, pathological impairment, and cell apoptosis in the diaphragm were mitigated by NRG-1ß. And NRG-1ß activated the PI3K/Akt signaling in the diaphragm of septic rats. Elevated ROS production in the LPS-stimulated L6 rat skeletal muscle cells was reduced after treatment with NRG-1ß, while MK-2206 blocked these effects of NRG-1ß. In conclusion, our findings underlined that NRG-1ß could reduce circulating levels of proinflammatory cytokines in rats with sepsis, adjust diaphragmatic proinflammatory cytokine level, mitigate diaphragmatic oxidative injury, and lessen diaphragm cell apoptosis, thereby improving diaphragmatic function, and play a role in diaphragmatic protection by activating PI3K/Akt signaling.


Diaphragm/drug effects , Neuregulin-1/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/drug therapy , Animals , Inflammation , Male , Neuregulin-1/pharmacology , Oxidative Stress , Rats , Rats, Sprague-Dawley
12.
Int J Mol Med ; 46(3): 925-935, 2020 Sep.
Article En | MEDLINE | ID: mdl-32705151

As regards acute myocardial infarction, great success has been achieved in therapies that reduce the effects of myocardial ischemic injury, while few interventions have achieved satisfactory outcomes for myocardial ischemia­reperfusion (IR) injury. Thus, new research is urgently required to achieve breakthroughs in promising treatments. Neuregulin­1 (NRG­1), which is an endothelium­derived protein and the ligand of ErbB receptors, exerts cardioprotective effects and is rapidly upregulated during IR. NRG­1/ErbB activates several downstream signaling pathways in response to myocardial IR injury. Previous studies have revealed the protective effects of NRG­1 during heart failure, and numerous experiments have explored the mechanisms underlying the NRG­1­induced cardioprotective effects against myocardial IR injury. In the present review, the progress made in the research of NRG­1 as a cardioprotective agent during IR and related conditionings is summarized. Furthermore, the potential benefits of NRG­1 against myocardial IR injury are listed with the prospective use of NRG­1 in clinical applications.


Myocardial Reperfusion Injury/metabolism , Neuregulin-1/metabolism , Animals , Cardiotonic Agents/metabolism , Cardiotonic Agents/therapeutic use , ErbB Receptors/metabolism , Humans , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Neuregulin-1/therapeutic use , Protective Factors , Signal Transduction
13.
Mol Med Rep ; 21(6): 2311-2320, 2020 06.
Article En | MEDLINE | ID: mdl-32236630

Neuregulin-1 (NRG-1) is a type of epidermal growth factor­like protein primarily distributed in the nervous and cardiovascular systems. When sepsis occurs, the incidence of cardiac dysfunction in myocardial injury is high and the mechanism is complicated. It directly causes myocardial cell damage, whilst also causing damage to the structure and function of myocardial cells, weakening of endothelial function and coronary microcirculation, autonomic dysfunction, and activation of myocardial inhibitory factors. Studies investigating NRG­1 have been performed using a variety of methods, including in vitro models, and animal and human clinical trials; however, the results are not consistent. NRG­1/ErbBs signaling is involved in a variety of cardiac processes, from the development of the myocardium and cardiac conduction systems to the promotion of angiogenesis in cardiomyocytes, and in cardio­protective effects during injury. NRG­1 may exert a multifaceted cardiovascular protective effect by activating NRG­1/ErbBs signaling and regulating multiple downstream signaling pathways, thereby improving myocardial cell dysfunction in sepsis, and protecting cardiomyocytes and endothelial cells. It may alleviate myocardial microvascular endothelial injury in sepsis; its anti­inflammatory effects inhibit the production of myocardial inhibitory factors in sepsis, improve myocardial ischemia, decrease oxidative stress, regulate the disruption to the homeostasis of the autonomic nervous system, improve diastolic function, and offer protective effects at multiple target sites. As the mechanism of action of NRG­1 intersects with the pathways involved in the pathogenesis of sepsis, it may be applicable as a treatment strategy to numerous pathological processes in sepsis.


Heart Diseases/drug therapy , Heart Diseases/metabolism , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Sepsis/metabolism , Animals , Cardiomyopathies/metabolism , Endothelial Cells/metabolism , Humans , Myocardial Ischemia/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Neuregulin-1/genetics , Neuregulin-1/therapeutic use , Oxidative Stress/drug effects , Sepsis/complications , Signal Transduction/drug effects
14.
Arch Physiol Biochem ; 126(4): 320-325, 2020 Oct.
Article En | MEDLINE | ID: mdl-30449185

Context: Neuregulin 1 (NRG1) and ErbB receptors are involved in glucose homeostasis. However, the effects of the neuregulin 1-ErbB pathway activation on glucose metabolism in liver are controversial.Objective: Assess NRG1 and ErbB signalling in liver and the effects of 8-week treatment with NRG1 on glucose homeostasis in diabetic db/db mice and in control healthy mice.Results: NRG1 improved glucose, insulin and insulin sensitivity index during OGTT in db/db mice, but not in control mice. Compared with healthy mice, phosphorylation of p38, ErbB-1 and ErbB-3 was increased in diabetic mice, and neuregulin 1 treatment increased phosphorylation of p38 and ErbB-4. Conversely, the AKT/FOXO1 pathway was not affected by the 8-week treatment with NRG1.Conclusion: Diabetic mice showed altered NRG1-ErbB pathway in the liver compared with healthy mice. Moreover, chronic NRG1 treatment increased p38 phosphorylation in liver and improved glucose tolerance in diabetic mice, but not in control mice.


Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Neuregulin-1/pharmacology , Animals , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Neuregulin-1/therapeutic use , Time Factors , Treatment Outcome
15.
Circ Heart Fail ; 12(10): e006288, 2019 10.
Article En | MEDLINE | ID: mdl-31607147

Heart failure is a complex syndrome whose phenotypic presentation and disease progression depends on a complex network of adaptive and maladaptive responses. One of these responses is the endothelial release of NRG (neuregulin)-1-a paracrine growth factor activating ErbB2 (erythroblastic leukemia viral oncogene homolog B2), ErbB3, and ErbB4 receptor tyrosine kinases on various targets cells. NRG-1 features a multitasking profile tuning regenerative, inflammatory, fibrotic, and metabolic processes. Here, we review the activities of NRG-1 on different cell types and organs and their implication for heart failure progression and its comorbidities. Although, in general, effects of NRG-1 in heart failure are compensatory and beneficial, translation into therapies remains unaccomplished both because of the complexity of the underlying pathways and because of the challenges in the development of therapeutics (proteins, peptides, small molecules, and RNA-based therapies) for tyrosine kinase receptors. Here, we give an overview of the complexity to be faced and how it may be tackled.


Endothelial Cells/metabolism , Heart Failure/metabolism , Neuregulin-1/metabolism , Animals , Cardiovascular Agents/therapeutic use , Chronic Disease , Endothelial Cells/drug effects , ErbB Receptors/metabolism , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Ligands , Molecular Targeted Therapy , Neuregulin-1/therapeutic use , Signal Transduction
16.
PLoS One ; 14(9): e0222587, 2019.
Article En | MEDLINE | ID: mdl-31560696

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.


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
17.
J Mol Neurosci ; 69(2): 333-342, 2019 Oct.
Article En | MEDLINE | ID: mdl-31290093

Identifying novel neuroprotectants that can halt or reverse the neurological effects of stroke is of interest to both clinicians and scientists. We and others previously showed the pre-clinical neuroprotective efficacy of neuregulin-1 (NRG-1) in rats following focal brain ischemia. In this study, we examined neuroprotection by exogenous and endogenous NRG-1 using a mouse model of ischemic stroke. C57BL6 mice were subjected to middle cerebral artery occlusion (MCAO) followed by reperfusion. NRG-1 or vehicle was infused intra-arterially (i.a.) or intravenously (i.v.) after MCAO and before the onset of reperfusion. NRG-1 treatment (16 µg/kg; i.a.) reduced cerebral cortical infarct volume by 72% in mice when delivered post-ischemia. NRG-1 also inhibited neuronal injury as measured by Fluoro Jade B labeling and rescued NeuN immunoreactivity in neurons. Neuroprotection by NRG-1 was also observed in mice when administered i.v. (100 µg/kg) in both male and female mice. We investigated whether endogenous NRG-1 was neuroprotective using male and female heterozygous NRG-1 knockout mice (NRG-1+/-) compared with wild-type mice (WT) littermates. NRG-1+/- and WT mice were subjected to MCAO for 45 min, and infarct size was measured 24 h following MCAO. NRG-1+/- mice displayed a sixfold increase in cortical infarct size compared with WT mice. These results demonstrate that NRG-1 treatment mitigates neuronal damage following cerebral ischemia. We further showed that reduced endogenous NRG-1 results in exacerbated neuronal injury in vivo. These findings suggest that NRG-1 represents a promising therapy to treat stroke in human patients.


Infarction, Middle Cerebral Artery/drug therapy , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Female , Heterozygote , Infarction, Middle Cerebral Artery/genetics , Male , Mice , Mice, Inbred C57BL , Neuregulin-1/genetics
18.
Neuroscience ; 404: 510-518, 2019 04 15.
Article En | MEDLINE | ID: mdl-30731156

Brachial plexus root avulsion (BPRA) results in the complete loss of motor function in the upper limb, mainly due to the death of spinal motoneurons (MNs). The survival of spinal MNs is the key to the recovery of motor function. Neuregulin-1 (Nrg1) plays fundamental roles in nervous system development and nerve repair. However, its functional role in BPRA remains unclear. On the basis of our findings that Nrg1 is down-regulated in the ventral horn in a mouse model of BPRA, Nrg1 may be associated with BPRA. Here, we investigated whether recombinant Nrg1ß (rNrg1ß) can enhance the survival of spinal MNs and improve functional recovery in mice following BPRA. In vitro studies on primary cultured mouse MNs showed that rNrg1ß increased the survival rate in a dose-dependent manner, reaching a peak at 5 nM, which increased the survival rate and enhanced the pERK levels in MNs under H2O2-induced oxidative stress. In vivo studies revealed that rNrg1ß improved the functional recovery of elbow flexion, promoted the survival of MNs, enhanced the re-innervation of biceps brachii, and decreased the muscle atrophy. These results suggest that Nrg1 may provide a potential therapeutic strategy for root avulsion.


Brachial Plexus/drug effects , Motor Neurons/drug effects , Neuregulin-1/therapeutic use , Radiculopathy/drug therapy , Recovery of Function/drug effects , Animals , Brachial Plexus/physiopathology , Cell Survival/drug effects , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Neuregulin-1/pharmacology , Radiculopathy/physiopathology , Recovery of Function/physiology
19.
J Drug Target ; 27(5-6): 573-581, 2019.
Article En | MEDLINE | ID: mdl-30289743

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.


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
20.
Clin Exp Pharmacol Physiol ; 46(3): 255-265, 2019 03.
Article En | MEDLINE | ID: mdl-30339273

We have previously shown that treatment with recombinant human neuregulin-1 (rhNRG-1) improves pulmonary arterial hypertension (PAH) in a monocrotaline (MCT)-induced animal model, by decreasing pulmonary arterial remodelling and endothelial dysfunction, as well as by restoring right ventricular (RV) function. Additionally, rhNRG-1 treatment showed direct myocardial anti-remodelling effects in a model of pressure loading of the RV without PAH. This work aimed to study the intrinsic cardiac effects of rhNRG-1 on experimental PAH and RV pressure overload, and more specifically on diastolic stiffness, at both the ventricular and cardiomyocyte level. We studied the effects of chronic rhNRG-1 treatment on ventricular passive stiffness in RV and LV samples from MCT-induced PAH animals and in the RV from animals with compensated and decompensated RV hypertrophy, through a mild and severe pulmonary artery banding (PAB). We also measured passive tension in isolated cardiomyocytes and quantified the expression of myocardial remodelling-associated genes and calcium handling proteins. Chronic rhNRG-1 treatment decreased passive tension development in RV and LV isolated from animals with MCT-induced PAH. This decrease was associated with increased phospholamban phosphorylation, and with attenuation of the expression of cardiac maladaptive remodelling markers. Finally, we showed that rhNRG-1 therapy decreased RV remodelling and cardiomyocyte passive tension development in PAB-induced RV hypertrophy animals, without compromising cardiac function, pointing to cardiac-specific effects in both hypertrophy stages. In conclusion, we demonstrated that rhNRG-1 treatment decreased RV intrinsic diastolic stiffness, through the improvement of calcium handling and cardiac remodelling signalling.


Diastole/physiology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Neuregulin-1/pharmacology , Vascular Stiffness/drug effects , Ventricular Dysfunction, Right/drug therapy , Animals , Calcium Signaling/drug effects , Gene Expression Regulation/drug effects , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neuregulin-1/therapeutic use , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Ventricular Remodeling/drug effects
...