Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
J Mol Cell Cardiol ; 187: 26-37, 2024 02.
Article in English | MEDLINE | ID: mdl-38150867

ABSTRACT

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.


Subject(s)
Cardiomyopathies , Myocardial Infarction , Mice , Animals , Humans , Signal Transduction , Myocytes, Cardiac/metabolism , Neuregulin-1/therapeutic use , Cardiomyopathies/metabolism , Receptor, ErbB-4/metabolism
2.
Cytokine ; 174: 156439, 2024 02.
Article in English | MEDLINE | ID: mdl-38134557

ABSTRACT

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.


Subject(s)
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
3.
Future Oncol ; 18(18): 2193-2200, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35400204

ABSTRACT

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.


Subject(s)
Language , Lung Neoplasms , Afatinib/therapeutic use , Gene Fusion , Humans , Lung Neoplasms/drug therapy , Neuregulin-1/genetics , Neuregulin-1/therapeutic use , Oncogene Proteins, Fusion/genetics
4.
Future Oncol ; 18(26): 2865-2870, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35876504

ABSTRACT

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.


Subject(s)
Lung Neoplasms , Afatinib/therapeutic use , Biology , Humans , Language , Lung Neoplasms/pathology , Neuregulin-1/genetics , Neuregulin-1/therapeutic use , Oncogene Proteins, Fusion/genetics
5.
Clin Exp Pharmacol Physiol ; 46(3): 255-265, 2019 03.
Article in English | MEDLINE | ID: mdl-30339273

ABSTRACT

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.


Subject(s)
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
6.
J Neuroinflammation ; 15(1): 104, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29636063

ABSTRACT

BACKGROUND: Human cerebral malaria (HCM) is a severe form of malaria characterized by sequestration of infected erythrocytes (IRBCs) in brain microvessels, increased levels of circulating free heme and pro-inflammatory cytokines and chemokines, brain swelling, vascular dysfunction, coma, and increased mortality. Neuregulin-1ß (NRG-1) encoded by the gene NRG1, is a member of a family of polypeptide growth factors required for normal development of the nervous system and the heart. Utilizing an experimental cerebral malaria (ECM) model (Plasmodium berghei ANKA in C57BL/6), we reported that NRG-1 played a cytoprotective role in ECM and that circulating levels were inversely correlated with ECM severity. Intravenous infusion of NRG-1 reduced ECM mortality in mice by promoting a robust anti-inflammatory response coupled with reduction in accumulation of IRBCs in microvessels and reduced tissue damage. METHODS: In the current study, we examined how NRG-1 treatment attenuates pathogenesis and mortality associated with ECM. We examined whether NRG-1 protects against CXCL10- and heme-induced apoptosis using human brain microvascular endothelial (hCMEC/D3) cells and M059K neuroglial cells. hCMEC/D3 cells grown in a monolayer and a co-culture system with 30 µM heme and NRG-1 (100 ng/ml) were used to examine the role of NRG-1 on blood brain barrier (BBB) integrity. Using the in vivo ECM model, we examined whether the reduction of mortality was associated with the activation of ErbB4 and AKT and inactivation of STAT3 signaling pathways. For data analysis, unpaired t test or one-way ANOVA with Dunnett's or Bonferroni's post test was applied. RESULTS: We determined that NRG-1 protects against cell death/apoptosis of human brain microvascular endothelial cells and neroglial cells, the two major components of BBB. NRG-1 treatment improved heme-induced disruption of the in vitro BBB model consisting of hCMEC/D3 and human M059K cells. In the ECM murine model, NRG-1 treatment stimulated ErbB4 phosphorylation (pErbB4) followed by activation of AKT and inactivation of STAT3, which attenuated ECM mortality. CONCLUSIONS: Our results indicate a potential pathway by which NRG-1 treatment maintains BBB integrity in vitro, attenuates ECM-induced tissue injury, and reduces mortality. Furthermore, we postulate that augmenting NRG-1 during ECM therapy may be an effective adjunctive therapy to reduce CNS tissue injury and potentially increase the effectiveness of current anti-malaria therapy against human cerebral malaria (HCM).


Subject(s)
Malaria, Cerebral/drug therapy , Neuregulin-1/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Claudin-5/metabolism , Coculture Techniques , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/physiology , Hemangioendothelioma , Humans , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
7.
Glia ; 65(7): 1152-1175, 2017 07.
Article in English | MEDLINE | ID: mdl-28456012

ABSTRACT

Spinal cord injury (SCI) results in glial activation and neuroinflammation, which play pivotal roles in the secondary injury mechanisms with both pro- and antiregeneration effects. Presently, little is known about the endogenous molecular mechanisms that regulate glial functions in the injured spinal cord. We previously reported that the expression of neuregulin-1 (Nrg-1) is acutely and chronically declined following traumatic SCI. Here, we investigated the potential ramifications of Nrg-1 dysregulation on glial and immune cell reactivity following SCI. Using complementary in vitro approaches and a clinically-relevant model of severe compressive SCI in rats, we demonstrate that immediate delivery of Nrg-1 (500 ng/day) after injury enhances a neuroprotective phenotype in inflammatory cells associated with increased interleukin-10 and arginase-1 expression. We also found a decrease in proinflammatory factors including IL-1ß, TNF-α, matrix metalloproteinases (MMP-2 and 9) and nitric oxide after injury. In addition, Nrg-1 modulates astrogliosis and scar formation by reducing inhibitory chondroitin sulfate proteoglycans after SCI. Mechanistically, Nrg-1 effects on activated glia are mediated through ErbB2 tyrosine phosphorylation in an ErbB2/3 heterodimer complex. Furthermore, Nrg-1 exerts its effects through downregulation of MyD88, a downstream adaptor of Toll-like receptors, and increased phosphorylation of Erk1/2 and STAT3. Nrg-1 treatment with the therapeutic dosage of 1.5 µg/day significantly improves tissue preservation and functional recovery following SCI. Our findings for the first time provide novel insights into the role and mechanisms of Nrg-1 in acute SCI and suggest a positive immunomodulatory role for Nrg-1 that can harness the beneficial properties of activated glia and inflammatory cells in recovery following SCI.


Subject(s)
Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Neuregulin-1/therapeutic use , Neuroglia/physiology , Recovery of Function/physiology , Spinal Cord Injuries/complications , Animals , Animals, Newborn , Arginase/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Interleukin-10/metabolism , Lipopolysaccharides/toxicity , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Neuroglia/drug effects , Rats , Recovery of Function/drug effects , Signal Transduction/drug effects , Spinal Cord Injuries/pathology , Time Factors
8.
Growth Factors ; 35(6): 225-233, 2017 12.
Article in English | MEDLINE | ID: mdl-29458290

ABSTRACT

Corneal epithelial cells (CECs) play an important role in the function of the cornea, and are maintained by corneal epithelial stem cells (CESCs). Recent studies have shown that neuronal growth factors affect the proliferation and migration of CESCs. Neuregulin-1 (NR-1) is a neuronal growth factor that is expressed in the early stages of brain development. The aim of this study was to determine whether NR-1 activates corneal wound healing. We observed that NR-1 activated both proliferation and migration of CECs. In addition, the colony-forming efficacy of CESCs was enhanced. In mice, NR-1 treatment improved corneal wound healing. Furthermore, the expression of markers of corneal epithelium maintenance (ΔNp63) and CESC proliferation (Bmi-1 and Abcg2) was increased. These effects were mediated by intracellular signalling pathways (Stat3, Erk1/2 and p38). Taken together, our results suggest that NR-1 accelerates the recovery of corneal wounds, and may represent a novel treatment for corneal damage.


Subject(s)
Corneal Injuries/drug therapy , Epithelial Cells/drug effects , Neuregulin-1/pharmacology , Wound Healing/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/physiology , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuregulin-1/therapeutic use , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , STAT3 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Am J Physiol Heart Circ Physiol ; 312(5): H907-H918, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28235789

ABSTRACT

Immune activation in chronic systolic heart failure (HF) correlates with disease severity and prognosis. Recombinant neuregulin-1 (rNRG-1) is being developed as a possible therapy for HF, based on the activation of ERBB receptors in cardiac cells. Work in animal models of HF led us to hypothesize that there may be direct effects of NRG-1 on immune system activation and inflammation. We investigated the expression of ERBB receptors and the effect of rNRG-1 isoform glial growth factor 2 (GGF2) in subpopulations of peripheral blood mononuclear cells (PB MNCs) in subjects with HF. We found that human monocytes express both ERBB2 and ERBB3 receptors, with high interindividual variability among subjects. Monocyte surface ERBB3 and TNF-α mRNA expression were inversely correlated in subjects with HF but not in human subjects without HF. GGF2 activation of ERBB signaling ex vivo inhibited LPS-induced TNF-α production, specifically in the CD14lowCD16+ population of monocytes in a phosphoinositide 3-kinase-dependent manner. GGF2 suppression of TNF-α correlated directly with the expression of ERBB3. In vivo, a single dose of intravenous GGF2 reduced TNF-α expression in PB MNCs of HF subjects participating in a phase I safety study of GGF2. These results support a role for ERBB3 signaling in the regulation of TNF-α production from CD14lowCD16+ monocytes and a need for further investigation into the clinical significance of NRG-1/ERBB signaling as a modulator of immune system function.NEW & NOTEWORTHY This study identified a novel role of neuregulin-1 (NRG-1)/ERBB signaling in the control of proinflammatory activation of monocytes. These results further improve our fundamental understanding of cardioprotective effects of NRG-1 in patients with heart failure.


Subject(s)
ErbB Receptors/biosynthesis , Inflammation/physiopathology , Monocytes , Signal Transduction , ErbB Receptors/drug effects , ErbB Receptors/genetics , Female , Humans , In Vitro Techniques , Macrophage Activation , Male , Middle Aged , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Receptor, ErbB-3/biosynthesis , Receptor, ErbB-3/genetics , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
10.
Curr Cardiol Rep ; 19(10): 94, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28840572

ABSTRACT

PURPOSE OF REVIEW: This review aims to summarize the growing body of literature of HF with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF) with a focus on recent pharmacologics. RECENT FINDINGS: HFrEF continues to be more widely investigated than HFpEF. Ivabradine and combinatorial treatment with sacubitril and valsartan are promising newly approved therapies. Other experimental therapies have emerged, which include Serelaxin, Empagliflozin, Neuregulin, and Omecamtive mecarbil, among others. These drugs need to continue to be investigated for safety and efficacy. We predict ivabradine and combinatorial treatment with sacubitril-valsartan to develop as a widespread therapy. New therapies should be aimed at treating HFpEF or target the cardiomyocyte itself.


Subject(s)
Cardiovascular Agents/therapeutic use , Heart Failure/drug therapy , Aminobutyrates/therapeutic use , Benzazepines/therapeutic use , Benzhydryl Compounds/therapeutic use , Biphenyl Compounds , Drug Combinations , Glucosides/therapeutic use , Heart Failure/physiopathology , Humans , Ivabradine , Neuregulin-1/therapeutic use , Recombinant Proteins/therapeutic use , Relaxin/therapeutic use , Stroke Volume/physiology , Tetrazoles/therapeutic use , Urea/analogs & derivatives , Urea/therapeutic use , Valsartan
11.
J Neuroinflammation ; 13(1): 237, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27596278

ABSTRACT

BACKGROUND: We previously demonstrated that neuregulin-1 (NRG-1) was neuroprotective in rats following ischemic stroke. Neuroprotection by NRG-1 was associated with the suppression of pro-inflammatory gene expression in brain tissues. Over-activation of brain microglia can induce pro-inflammatory gene expression by activation of transcriptional regulators following stroke. Here, we examined how NRG-1 transcriptionally regulates inflammatory gene expression by computational bioinformatics and in vitro using microglial cells. METHODS: To identify transcriptional regulators involved in ischemia-induced inflammatory gene expression, rats were sacrificed 24 h after middle cerebral artery occlusion (MCAO) and NRG-1 treatment. Gene expression profiles of brain tissues following ischemia and NRG-1 treatment were examined by microarray technology. The Conserved Transcription Factor-Binding Site Finder (CONFAC) bioinformatics software package was used to predict transcription factors associated with inflammatory genes induced following stroke and suppressed by NRG-1 treatment. NF-kappa B (NF-kB) was identified as a potential transcriptional regulator of NRG-1-suppressed genes following ischemia. The involvement of specific NF-kB subunits in NRG-1-mediated inflammatory responses was examined using N9 microglial cells pre-treated with NRG-1 (100 ng/ml) followed by lipopolysaccharide (LPS; 10 µg/ml) stimulation. The effects of NRG-1 on cytokine production were investigated using Luminex technology. The levels of the p65, p52, and RelB subunits of NF-kB and IkB-α were determined by western blot analysis and ELISA. Phosphorylation of IkB-α was investigated by ELISA. RESULTS: CONFAC identified 12 statistically over-represented transcription factor-binding sites (TFBS) in our dataset, including NF-kBP65. Using N9 microglial cells, we observed that NRG-1 significantly inhibited LPS-induced TNFα and IL-6 release. LPS increased the phosphorylation and degradation of IkB-α which was blocked by NRG-1. NRG-1 also prevented the nuclear translocation of the NF-kB p65 subunit following LPS administration. However, NRG-1 increased production of the neuroprotective cytokine granulocyte colony-stimulating factor (G-CSF) and the nuclear translocation of the NF-kB p52 subunit, which is associated with the induction of anti-apoptotic and suppression of pro-inflammatory gene expression. CONCLUSIONS: Neuroprotective and anti-inflammatory effects of NRG-1 are associated with the differential regulation of NF-kB signaling pathways in microglia. Taken together, these findings suggest that NRG-1 may be a potential therapeutic treatment for treating stroke and other neuroinflammatory disorders.


Subject(s)
Encephalitis/drug therapy , Encephalitis/etiology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Microglia/drug effects , Neuregulin-1/therapeutic use , Animals , Cell Line, Transformed , Computational Biology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Granulocyte Colony-Stimulating Factor/metabolism , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Male , Microarray Analysis , NF-kappa B/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
12.
J Neurosci Res ; 94(3): 253-65, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26660233

ABSTRACT

Cimaglermin (neuregulin 1ß3, 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.


Subject(s)
Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Nerve Tissue Proteins/metabolism , Neuregulin-1/therapeutic use , Recovery of Function/drug effects , Analysis of Variance , Animals , Brain Infarction/drug therapy , Brain Infarction/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Rats , Rats, Sprague-Dawley , Time Factors
13.
J Neurochem ; 133(4): 590-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25581060

ABSTRACT

Previously, we demonstrated that systemically injected extracellular domain of neuregulin-1ß1 (Nrg1ß1), a nerve growth and differentiation factor, passes the blood-brain barrier and rescues dopaminergic neurons of substantia nigra in the 6-hydroxydopamine-mouse model of Parkinson's disease (PD). Here, we studied the effects of peripherally administered Nrg1ß1 in another toxin-based mouse model of PD. For this purpose, (i) nigrostriatal pathway injury was induced by treatment of adult wild-type mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in acute and subchronic paradigms; and (ii) Nrg1ß1 or saline (control) were administered 1 h before each MPTP injection. We found that Nrg1ß1 significantly reduced the loss of nigral dopaminergic neurons in both intoxication paradigms (7 days post-injection). However, Nrg1ß1 did not reverse MPTP-induced decrease in dopamine levels and dopaminergic fibers in the striatum. We also show that MPTP conversion to its toxic metabolite 1-methyl-4-phenylpyridinium as well as levels of dopamine transporter, mediating intracellular uptake of 1-methyl-4-phenylpyridinium, are unaffected by Nrg1ß1. Finally, neuroprotective properties of Nrg1ß1 on nigral dopaminergic neurons are specifically mediated by ErbB4 as revealed through the study of ErbB4 knockout mice. In conclusion, systemically administered Nrg1ß1 protects midbrain dopaminergic neurons against this PD-related toxic insult. Thus, Nrg1ß1 may have a benefit in the treatment of PD patients. Previously, we demonstrated that systemically administered neuregulin-1ß1 (Nrg1ß1) passes the blood-brain barrier, phosphorylates ErbB4 receptors and elevates dopamine (DA) levels in the nigrostriatal system of healthy mice. Nrg1ß1 protects nigral DAergic neurons in the 6-hydroxydopamine (6-OHDA) mouse model of Parkinson's disease (PD). Here, we show that Nrg1ß1 rescues nigral DAergic neurons also against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced cell death. ErbB4 expression is essential for the neuroprotective effect of Nrg1ß1 on midbrain DAergic neurons. Nrg1ß1 might be beneficial in PD treatment.


Subject(s)
Dopaminergic Neurons/drug effects , MPTP Poisoning/pathology , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Substantia Nigra/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Animals, Genetically Modified , Disease Models, Animal , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , MPTP Poisoning/chemically induced , Male , Mice , Mice, Inbred C57BL , Neuregulin-1/pharmacology , Neuroprotective Agents/pharmacology , Receptor, ErbB-4/deficiency , Receptor, ErbB-4/genetics , Time Factors
14.
J Neuroinflammation ; 12: 64, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25880399

ABSTRACT

BACKGROUND: Neuregulin-1 (NRG-1) has been shown to act as a neuroprotectant in animal models of nerve agent intoxication and other acute brain injuries. We recently demonstrated that NRG-1 blocked delayed neuronal death in rats intoxicated with the organophosphate (OP) neurotoxin diisopropylflurophosphate (DFP). It has been proposed that inflammatory mediators are involved in the pathogenesis of OP neurotoxin-mediated brain damage. METHODS: We examined the influence of NRG-1 on inflammatory responses in the rat brain following DFP intoxication. Microglial activation was determined by immunohistchemistry using anti-CD11b and anti-ED1 antibodies. Gene expression profiling was performed with brain tissues using Affymetrix gene arrays and analyzed using the Ingenuity Pathway Analysis software. Cytokine mRNA levels following DFP and NRG-1 treatment was validated by real-time reverse transcription polymerase chain reaction (RT-PCR). RESULTS: DFP administration resulted in microglial activation in multiple brain regions, and this response was suppressed by treatment with NRG-1. Using microarray gene expression profiling, we observed that DFP increased mRNA levels of approximately 1,300 genes in the hippocampus 24 h after administration. NRG-1 treatment suppressed by 50% or more a small fraction of DFP-induced genes, which were primarily associated with inflammatory responses. Real-time RT-PCR confirmed that the mRNAs for pro-inflammatory cytokines interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) were significantly increased following DFP exposure and that NRG-1 significantly attenuated this transcriptional response. In contrast, tumor necrosis factor α (TNFα) transcript levels were unchanged in both DFP and DFP + NRG-1 treated brains relative to controls. CONCLUSION: Neuroprotection by NRG-1 against OP neurotoxicity is associated with the suppression of pro-inflammatory responses in brain microglia. These findings provide new insight regarding the molecular mechanisms involved in the neuroprotective role of NRG-1 in acute brain injuries.


Subject(s)
Cholinesterase Inhibitors/toxicity , Cholinesterase Inhibitors/therapeutic use , Encephalitis/chemically induced , Isoflurophate/toxicity , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/drug effects , Injections, Intra-Arterial , Male , Microglia/drug effects , Microglia/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger , Rats , Rats, Sprague-Dawley
15.
Behav Brain Funct ; 11: 5, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-25886297

ABSTRACT

OBJECTIVE: To observe the effects of neuregulin1ß (NRG1ß) on the level of phosphorylated ERK1/2 (p-ERK1/2), and explore the therapeutic mechanism of NRG1ß on the cognitive dysfunction in rats with chronic omethoate poisoning. METHODS: Rats with strong learning and memory ability, 50 in total, were selected by Y-electric maze test. Among which, 15 rats were randomly selected into control group, and the rest 35 rats were used to establish experimental cognitive impairment models by being injected with omethoate subcutaneously. The 30 cases of successful cognitive impairment models were randomly divided into model group and treated group consisting of 15 rats, respectively. Then rats in treated group were injected with NRG1ß into their lateral ventricles, while rats in control and model groups were given equal volume of PBS simultaneously. The cognitive capacity of rats was evaluated with Y-electric maze. The morphology and ultrastructure of hippocampus were observed by hematoxylin eosin (HE) staining and transmission electron microscopy (TEM) respectively. The expression of p-ERK1/2 was determined by immunohistochemical (IHC) staining and Western blotting. RESULTS: Compared with rats in model group, the cognitive ability of rats with omethoate exposed (model and treated groups) reduced significantly, along with the obvious damage of hippocampal neurons and the expression of p-ERK1/2 decreased significantly (P < 0.05). And after treatment with NRG1ß, the cognitive activity of treated rats was improved obviously, and the injury of hippocampal neurons was milder and the expression of p-ERK1/2 increased significantly more than those in model rats (P < 0.05). CONCLUSION: In chronic omethoate poisoning rats, NRG1ß can promote the phosphorylation level of ERK1/2 in hippocampal neurons, and play an important role in the improvement of cognitive function.


Subject(s)
Cognition Disorders/drug therapy , Dimethoate/analogs & derivatives , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Insecticides/toxicity , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/prevention & control , Animals , Cognition Disorders/chemically induced , Cognition Disorders/psychology , Dimethoate/toxicity , Hippocampus/pathology , Injections, Intraventricular , Learning Disabilities/chemically induced , Learning Disabilities/psychology , Male , Neuregulin-1/administration & dosage , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Neurotoxicity Syndromes/psychology , Phosphorylation/drug effects , Rats , Rats, Wistar , Up-Regulation/drug effects
16.
J Neuroinflammation ; 11: 9, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24433482

ABSTRACT

BACKGROUND: Cerebral Malaria (CM) is a diffuse encephalopathy caused by Plasmodium falciparum infection. Despite availability of antimalarial drugs, CM-associated mortality remains high at approximately 30% and a subset of survivors develop neurological and cognitive disabilities. While antimalarials are effective at clearing Plasmodium parasites they do little to protect against CM pathophysiology and parasite-induced brain inflammation that leads to seizures, coma and long-term neurological sequelae in CM patients. Thus, there is urgent need to explore therapeutics that can reduce or prevent CM pathogenesis and associated brain inflammation to improve survival. Neuregulin-1 (NRG-1) is a neurotrophic growth factor shown to protect against brain injury associated with acute ischemic stroke (AIS) and neurotoxin exposure. However, this drug has not been tested against CM-associated brain injury. Since CM-associated brain injuries and AIS share similar pathophysiological features, we hypothesized that NRG-1 will reduce or prevent neuroinflammation and brain damage as well as improve survival in mice with late-stage experimental cerebral malaria (ECM). METHODS: We tested the effects of NRG-1 on ECM-associated brain inflammation and mortality in P. berghei ANKA (PbA)-infected mice and compared to artemether (ARM) treatment; an antimalarial currently used in various combination therapies against malaria. RESULTS: Treatment with ARM (25 mg/kg/day) effectively cleared parasites and reduced mortality in PbA-infected mice by 82%. Remarkably, NRG-1 therapy (1.25 ng/kg/day) significantly improved survival against ECM by 73% despite increase in parasite burden within NRG-1-treated mice. Additionally, NRG-1 therapy reduced systemic and brain pro-inflammatory factors TNFalpha, IL-6, IL-1alpha and CXCL10 and enhanced anti-inflammatory factors, IL-5 and IL-13 while decreasing leukocyte accumulation in brain microvessels. CONCLUSIONS: This study suggests that NRG-1 attenuates ECM-associated brain inflammation and injuries and may represent a novel supportive therapy for the management of CM.


Subject(s)
Antimalarials/therapeutic use , Encephalitis/drug therapy , Malaria, Cerebral/drug therapy , Malaria, Cerebral/mortality , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Artemether , Artemisinins/therapeutic use , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/parasitology , Brain/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalitis/etiology , Encephalitis/pathology , Endothelium/drug effects , Endothelium/pathology , Leukocytes/drug effects , Leukocytes/pathology , Malaria, Cerebral/complications , Mice , Mice, Inbred C57BL , Neuregulin-1/metabolism , Plasmodium berghei/physiology
17.
Curr Heart Fail Rep ; 11(2): 134-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24682830

ABSTRACT

The National Cancer Institute estimates that approximately 13.7 million Americans with a history of cancer were alive on January 1, 2012. With the rising number of cancer survivors, there is an increased focus on how chemotherapy agents modulate the cardiovascular biology and cause chemotherapy-related heart failure in certain patients. Neuregulin-1 (NRG-1) is an important cardiac growth factor that is essential for normal myocardial development and maintenance. Certain chemotherapy agents perturb the normal NRG-1 signaling in the cardiovascular system and cause cardiac dysfunction and, in some cases, symptomatic heart failure. As researchers have learned the critical importance of NRG-1 within the cardiovascular system, more attention has been focused on the potential use of NRG-1 as biomarker and therapy for the treatment of heart failure. This review will highlight the biology of NRG-1 within the cardiovascular system, its role in chemotherapy-induced heart failure, and the translational potential of NRG-1 as treatment for heart failure.


Subject(s)
Antineoplastic Agents/adverse effects , Heart Failure/chemically induced , Neuregulin-1/physiology , Biomarkers/metabolism , Cardiotoxicity/drug therapy , Cardiotoxicity/physiopathology , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Neuregulin-1/metabolism , Neuregulin-1/therapeutic use , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Curr Heart Fail Rep ; 11(1): 40-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24234399

ABSTRACT

The beta isoform of Neuregulin-1 (NRG-1ß), along with its receptors (ErbB2-4), is required for cardiac development. NRG-1ß, 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-1ß on cardiac function. In addition to the possible role for recombinant NRG-1ßs as heart failure therapies, endogenous NRG-1ß/ErbB signaling appears to play a role in restoring cardiac function after injury. The potential mechanisms by which NRG-1ß may act as both a therapy and a mediator of reverse remodeling remain incompletely understood. In addition to direct effects on cardiac myocytes NRG-1ß acts on the vasculature, interstitium, cardiac fibroblasts, and hematopoietic and immune cells, which, collectively, may contribute to NRG-1ß's role in maintaining cardiac structure and function, as well as mediating reverse remodeling.


Subject(s)
Cardiovascular Agents/therapeutic use , Heart Failure/drug therapy , Neuregulin-1/therapeutic use , Biomarkers/blood , Cardiovascular Diseases/diagnosis , ErbB Receptors/physiology , Heart Failure/physiopathology , Humans , Neuregulin-1/blood , Neuregulin-1/physiology , Recombinant Proteins/therapeutic use , Signal Transduction/physiology , Ventricular Remodeling/physiology
19.
Diagn Pathol ; 19(1): 1, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38173003

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL