Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 64
1.
J Physiol Pharmacol ; 75(1)2024 02.
Article En | MEDLINE | ID: mdl-38583443

Neuregulin receptor degradation protein-1 (Nrdp1) is a newly discovered E3 ligase that plays a role in the apoptosis process of multiple diseases. Previous studies has shown that Nrdp1 exerted a proapoptotic effect in cardiac diseases. The purpose of this study is to investigate the potential involvement of Nrdp1 in the pathological processes of inflammatory bowel disease (IBD). To create a mouse model of experimental colitis, trinitrobenzenesulfonic acid (TNBS) was administered and the severity of colitis was assessed based on changes in weight and histological scores. Using Western blot and immunohistochemistry, significant increase in Nrdp1 expression was observed in intestinal epithelial cells (IECs). This was accompanied with the up-regulation of cleaved PARP and active caspase-3 in IECs, indicating a potential function in IECs. To study this further, we built an in vitro model of tumor necrosis factor-alpha (TNF-α)-induced apoptosis using human IEC line HT-29 cells. When Nrdp1 was knocked down, a decrease in apoptosis was observed, suggesting that Nrdp1 may play a proapoptotic role in IEC apoptosis. The mechanism behind this phenomenon is associated with the suppression of downstream targets of Nrdp1, such as protein kinase B (AKT). Furthermore, immunohistochemistry analysis in patients with Crohn's disease (CD) and normal controls supported the same results as observed in experimental colitis. We conclude that Nrdp1 may be a promising new therapeutic target for ameliorating IBD in humans.


Colitis , Crohn Disease , Animals , Humans , Mice , Apoptosis , Colitis/metabolism , Crohn Disease/drug therapy , Intestinal Mucosa , Intestines/pathology , Neuregulins/metabolism , Neuregulins/pharmacology , Neuregulins/therapeutic use
2.
Eur J Pharmacol ; 884: 173350, 2020 Oct 05.
Article En | MEDLINE | ID: mdl-32726654

Neuregulin 4 (Nrg4) is a brown fat-enriched endocrine factor that exerts beneficial metabolic effects on insulin resistance and hepatic steatosis. Autophagy is a mechanism that is essential for preventing hepatic steatosis. The aim of this study was to explore whether Nrg4 ameliorates hepatic steatosis by inducing autophagy. Aged C57BL/6 mice were maintained on a high fat diet with or without Nrg4 intervention for 3 months. Lipid accumulation in the liver was investigated. Autophagy related protein levels along with related signaling pathways that regulate autophagy were evaluated. In addition, the effects of Nrg4 on autophagy were also determined in cultured L-02 cells. Nrg4 decreased high-fat induced intrahepatic lipid content both in vivo and in vitro. Autophagy level in the liver also decreased in obese mice and Nrg4 intervention reactivated autophagy. Further, Nrg4 intervention was found to have activated autophagy via the adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling pathway. Moreover, when the AMPK/mTOR pathway was suppressed or autophagy was inhibited, the beneficial effects of Nrg4 intervention on hepatic steatosis were diminished. These results indicated that Nrg4 intervention attenuated hepatic steatosis by promoting autophagy in the liver of aged obese mice. Additionally, Nrg4 induced autophagy via the AMPK/mTOR signaling pathway.


AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Liver/drug effects , Neuregulins/pharmacology , Non-alcoholic Fatty Liver Disease/prevention & control , TOR Serine-Threonine Kinases/metabolism , Age Factors , Animals , Cell Line , Diet, High-Fat , Disease Models, Animal , Enzyme Activation , Lipid Metabolism/drug effects , Liver/enzymology , Liver/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction
3.
Glia ; 67(2): 309-320, 2019 02.
Article En | MEDLINE | ID: mdl-30485552

Gonadotropin releasing hormone (GnRH)-secretion is not only regulated by neuronal factors but also by astroglia cells via growth factors and ErbB receptors of the epidermal growth factor family. Studies in transgenic mice carrying mutations in the ErbB receptor system experience impaired reproductive capacity. In addition, some of these animals show a typical skin phenotype with wavy hair and curly whiskers. The rat strain SPRD-CU3 (CU3), examined in this study, displays a similar skin phenotype and a significant impairment of the timing of puberty onset and reproductive performance, suggesting a disruption in the astrocytic to GnRH neuronal communication. To address this issue, we analyzed astrocytic prostaglandin E2 (PGE2 ) release from primary hypothalamic astrocytic cell cultures after stimulation with transforming growth factor α (TGFα), ligand for ErbB1/ErbB2, or Neuregulin 1 beta 2 (NRG1ß2 ), ligand for ErbB4/ErbB2 signaling pathway. Compared to cultures from wild type animals, astrocytic cultures from CU3 rats were unable to respond to NRG stimulation, suggesting a disruption of the ErbB4/ErbB2 signaling pathway. This is confirmed by mutational analysis of ErbB4 that revealed a single point mutation at 3125 bp resulting in an amino acid change from proline to glutamine located at the carboxy-terminal region. As a consequence, substantial conformational changes occur in the transmembrane and intracellular domain of the protein, affecting the ability to form a receptor dimer with a partner and the ability to function as a transcriptional regulator. Thus, astroglia to GnRH neuronal signaling via ErbB4 is essential of timely onset of puberty and reproductive function.


Astrocytes/drug effects , Dinoprostone/metabolism , Disorders of Sex Development/pathology , Gonadotropin-Releasing Hormone/metabolism , Neuregulins/pharmacology , Neurons/metabolism , Receptor, ErbB-4/genetics , Animals , Astrocytes/metabolism , Cells, Cultured , Disease Models, Animal , Disorders of Sex Development/drug therapy , Disorders of Sex Development/genetics , Disorders of Sex Development/metabolism , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Hypothalamus/cytology , Models, Molecular , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Point Mutation/genetics , Rats , Rats, Transgenic , Receptor, ErbB-4/metabolism , Transforming Growth Factor alpha/metabolism
4.
Nat Neurosci ; 21(10): 1412-1420, 2018 10.
Article En | MEDLINE | ID: mdl-30224804

Schizophrenia is a severely debilitating neurodevelopmental disorder. Establishing a causal link between circuit dysfunction and particular behavioral traits that are relevant to schizophrenia is crucial to shed new light on the mechanisms underlying the pathology. We studied an animal model of the human 22q11 deletion syndrome, the mutation that represents the highest genetic risk of developing schizophrenia. We observed a desynchronization of hippocampal neuronal assemblies that resulted from parvalbumin interneuron hypoexcitability. Rescuing parvalbumin interneuron excitability with pharmacological or chemogenetic approaches was sufficient to restore wild-type-like CA1 network dynamics and hippocampal-dependent behavior during adulthood. In conclusion, our data provide insights into the network dysfunction underlying schizophrenia and highlight the use of reverse engineering to restore physiological and behavioral phenotypes in an animal model of neurodevelopmental disorder.


CA1 Region, Hippocampal/pathology , Mental Disorders/etiology , Nerve Net/pathology , Nonlinear Dynamics , Schizophrenia/pathology , Schizophrenia/physiopathology , 22q11 Deletion Syndrome/complications , 22q11 Deletion Syndrome/genetics , Action Potentials/drug effects , Action Potentials/physiology , Animals , Animals, Newborn , Clozapine/analogs & derivatives , Clozapine/pharmacology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/physiopathology , Neuregulins/pharmacology , Neurons/drug effects , Neurons/physiology , Parvalbumins/genetics , Parvalbumins/metabolism , Prepulse Inhibition/physiology , Reflex, Startle/physiology , Schizophrenia/etiology , Schizophrenia/genetics
5.
Am J Physiol Endocrinol Metab ; 315(4): E583-E593, 2018 10 01.
Article En | MEDLINE | ID: mdl-29944391

ErbB4, a member of the EGF receptor family, plays a variety of roles in physiological and pathological states. Genetic studies have indicated a link between ErbB4 and type 2 diabetes and obesity, but its role in metabolic syndrome (MetS) has not been reported. In the current study we found that mice with ErbB4 deletion developed MetS after 24 wk on a medium-fat diet (MFD), as indicated by development of obesity, dyslipidemia, hepatic steatosis, hyperglycemia, hyperinsulinemia, and insulin resistance, compared with wild-type mice. ErbB4 deletion mice also exhibited increased amounts of subcutaneous and visceral fat, with increased serum leptin levels, compared with wild-type mice, whereas levels of adiponectin were not significantly different. Histologically, severe inflammation, indicated by F4/80 immunostaining and M1 macrophage polarization, was detected in inguinal and epididymal white adipose tissue in ErbB4 deletion mice. ErbB4 expression decreased during 3T3-L1 preadipocyte differentiation. Administration of neuroregulin 4, a specific ligand for ErbB4, to 3T3-L1 adipocytes had no effect on adipogenesis and lipolysis but significantly inhibited lipogenesis, promoted browning, induced GLUT4 redistribution to the cell membrane, and increased glucose uptake. Neuroregulin 4 also significantly increased glucose uptake in adipocytes isolated from wild-type mice, while these effects were significantly decreased in adipocytes isolated from ErbB4 deletion mice. In conclusion, our results indicate that ErbB4 may play an important role in glucose homeostasis and lipogenesis. ErbB4 deficiency-related obesity and adipose tissue inflammation may contribute to the development of MetS.


Dietary Fats , Dyslipidemias/genetics , Fatty Liver/genetics , Hyperglycemia/genetics , Insulin Resistance/genetics , Metabolic Syndrome/genetics , Obesity/genetics , Receptor, ErbB-4/genetics , 3T3-L1 Cells , Adipogenesis/drug effects , Adiponectin/metabolism , Adipose Tissue, White/immunology , Animals , Gene Deletion , Genetic Predisposition to Disease , Glucose Transporter Type 4/drug effects , Glucose Transporter Type 4/metabolism , Hyperinsulinism/genetics , Inflammation , Intra-Abdominal Fat , Leptin/metabolism , Lipogenesis/drug effects , Macrophages , Male , Mice , Neuregulins/pharmacology , Subcutaneous Fat
6.
Mol Neurobiol ; 55(8): 6637-6660, 2018 Aug.
Article En | MEDLINE | ID: mdl-29327207

This study comprehensively addresses the phenotype, function, and whole transcriptome of primary human and rodent Schwann cells (SCs) and highlights key species-specific features beyond the expected donor variability that account for the differential ability of human SCs to proliferate, differentiate, and interact with axons in vitro. Contrary to rat SCs, human SCs were insensitive to mitogenic factors other than neuregulin and presented phenotypic variants at various stages of differentiation, along with a mixture of proliferating and senescent cells, under optimal growth-promoting conditions. The responses of human SCs to cAMP-induced differentiation featured morphological changes and cell cycle exit without a concomitant increase in myelin-related proteins and lipids. Human SCs efficiently extended processes along those of other SCs (human or rat) but failed to do so when placed in co-culture with sensory neurons under conditions supportive of myelination. Indeed, axon contact-dependent human SC alignment, proliferation, and differentiation were not observed and could not be overcome by growth factor supplementation. Strikingly, RNA-seq data revealed that ~ 44 of the transcriptome contained differentially expressed genes in human and rat SCs. A bioinformatics approach further highlighted that representative SC-specific transcripts encoding myelin-related and axon growth-promoting proteins were significantly affected and that a deficient expression of key transducers of cAMP and adhesion signaling explained the fairly limited potential of human SCs to differentiate and respond to axonal cues. These results confirmed the significance of combining traditional bioassays and high-resolution genomics methods to characterize human SCs and identify genes predictive of cell function and therapeutic value.


Biological Assay/methods , Schwann Cells/cytology , Sequence Analysis, RNA/methods , Adolescent , Adult , Aged , Animals , Axons/drug effects , Axons/metabolism , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Child , Colforsin/pharmacology , Cyclic AMP/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Mitogens/pharmacology , Myelin Sheath/genetics , Neuregulins/pharmacology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred F344 , Schwann Cells/drug effects , Schwann Cells/ultrastructure , Transcriptome/genetics , Young Adult
7.
Am J Physiol Endocrinol Metab ; 310(6): E440-51, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26714846

Neuregulin (NRG) is an EGF-related growth factor that binds to the tyrosine kinase receptors ErbB3 and ErbB4, thus inducing tissue development and muscle glucose utilization during contraction. Here, we analyzed whether NRG has systemic effects regulating glycemia in control and type 2 diabetic rats. To this end, recombinant NRG (rNRG) was injected into Zucker diabetic fatty (ZDF) rats and their respective lean littermates 15 min before a glucose tolerance test (GTT) was performed. rNRG enhanced glucose tolerance without promoting the activation of the insulin receptor (IR) or insulin receptor substrates (IRS) in muscle and liver. However, in control rats, rNRG induced the phosphorylation of protein kinase B (PKB) and glycogen synthase kinase-3 (GSK-3) in liver but not in muscle. In liver, rNRG increased ErbB3 tyrosine phosphorylation and its binding to phosphatidylinositol 3-kinase (PI3K), thus indicating that rNRG activates the ErbB3/PI3K/PKB signaling pathway. rNRG increased glycogen content in liver but not in muscle. rNRG also increased the content of fructose-2,6-bisphosphate (Fru-2,6-P2), an activator of hepatic glycolysis, and lactate in liver but not in muscle. Increases in lactate were abrogated by wortmannin, a PI3K inhibitor, in incubated hepatocytes. The liver of ZDF rats showed a reduced content of ErbB3 receptors, entailing a minor stimulation of the rNRG-induced PKB/GSK-3 cascade and resulting in unaltered hepatic glycogen content. Nonetheless, rNRG increased hepatic Fru-2,6-P2 and augmented lactate both in liver and in plasma of diabetic rats. As a whole, rNRG improved response to the GTT in both control and diabetic rats by enhancing hepatic glucose utilization.


Blood Glucose/drug effects , Diabetes Mellitus, Type 2/metabolism , Liver/drug effects , Muscle, Skeletal/drug effects , Neuregulins/pharmacology , Animals , Blood Glucose/metabolism , Case-Control Studies , Fructosediphosphates/metabolism , Glucose/metabolism , Glucose Tolerance Test , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Insulin , Insulin Receptor Substrate Proteins/drug effects , Insulin Receptor Substrate Proteins/metabolism , Lactic Acid/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinase/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Zucker , Receptor, ErbB-3/drug effects , Receptor, ErbB-3/metabolism , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism
8.
Biophys J ; 108(9): 2148-57, 2015 May 05.
Article En | MEDLINE | ID: mdl-25954873

Cell fates change dynamically in response to various extracellular signals, including growth factors that stimulate differentiation and proliferation. The processes underlying cell-fate decisions are complex and often include large cell-to-cell variations, even within a clonal population in the same environment. To understand the origins of these cell-to-cell variations, we must detect the internal dynamics of single cells that reflect their changing chemical milieu. In this study, we used the Raman spectra of single cells to trace their internal dynamics during the early stages of growth factor stimulation. This method allows nondestructive and inclusive time-series analyses of chemical compositions of the same single cells. Applying a Gaussian mixture model to the major principal components of the single-cell Raman spectra, we detected the dynamics of the chemical states in MCF-7 cancer-derived cells in the absence and presence of differentiation and proliferation factors. The dynamics displayed characteristic variations according to the functions of the growth factors. In the differentiation pathway, the chemical composition changed directionally between multiple states, including both reversible and irreversible state transitions. In contrast, in the proliferation pathway, the chemical composition was homogenized into a single state. The differentiation factor also stimulated fluctuations in the chemical composition, whereas the proliferation factor did not.


Insulin-Like Growth Factor I/pharmacology , Neuregulins/pharmacology , Single-Cell Analysis/methods , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Humans , MCF-7 Cells , Mice , Models, Biological , Spectrum Analysis, Raman/methods
9.
Sci Transl Med ; 7(281): 281ra45, 2015 Apr 01.
Article En | MEDLINE | ID: mdl-25834111

Therapies developed for adult patients with heart failure have been shown to be ineffective in pediatric clinical trials, leading to the recognition that new pediatric-specific therapies for heart failure must be developed. Administration of the recombinant growth factor neuregulin-1 (rNRG1) stimulates regeneration of heart muscle cells (cardiomyocytes) in adult mice. Because proliferation-competent cardiomyocytes are more abundant in growing mammals, we hypothesized that administration of rNRG1 during the neonatal period might be more effective than in adulthood. If so, neonatal rNRG1 delivery could be a new therapeutic strategy for treating heart failure in pediatric patients. To evaluate the effectiveness of rNRG1 administration in cardiac regeneration, newborn mice were subjected to cryoinjury, which induced myocardial dysfunction and scar formation and decreased cardiomyocyte cell cycle activity. Early administration of rNRG1 to mice from birth to 34 days of age improved myocardial function and reduced the prevalence of transmural scars. In contrast, administration of rNRG1 from 4 to 34 days of age only transiently improved myocardial function. The mechanisms of early administration involved cardiomyocyte protection (38%) and proliferation (62%). We also assessed the ability of rNRG1 to stimulate cardiomyocyte proliferation in intact cultured myocardium from pediatric patients. rNRG1 induced cardiomyocyte proliferation in myocardium from infants with heart disease who were less than 6 months of age. Our results identify an effective time period within which to execute rNRG1 clinical trials in pediatric patients for the stimulation of cardiomyocyte regeneration.


Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/therapy , Myocardium/pathology , Myocytes, Cardiac/pathology , Neuregulins/pharmacology , Regeneration/drug effects , Animals , Animals, Newborn , Cardiovascular Diseases/pathology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Child , Cicatrix/pathology , Cold Temperature , Gene Expression Regulation/drug effects , Humans , Infant , Mice , Myocytes, Cardiac/drug effects , Neuregulins/administration & dosage , Receptor, ErbB-4/metabolism
10.
Breast Cancer Res ; 17: 20, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-25849870

INTRODUCTION: Human epidermal growth factor receptor HER3 has been implicated in promoting the aggressiveness and metastatic potential of breast cancer. Upregulation of HER3 has been found to be a major mechanism underlying drug resistance to EGFR and HER2 tyrosine kinase inhibitors and to endocrine therapy in the treatment of breast cancer. Thus, agents that reduce HER3 expression at the plasma membrane may synergize with current therapies and offer a novel therapeutic strategy to improve treatment. METHODS: We devised an image-based screening platform using membrane localized HER3-YFP to identify small molecules that promote HER3 internalization and degradation. In vitro and in vivo tumor models were used to characterize the signaling effects of perhexiline, an anti-anginal drug, identified by the screening platform. RESULTS: We found perhexiline, an anti-anginal drug, selectively internalized HER3, decreased HER3 expression, and subsequently inhibited signaling downstream of HER3. Consistent with these results, perhexiline inhibited breast cancer cell proliferation in vitro and tumor growth in vivo. CONCLUSIONS: This is the first demonstration that HER3 can be targeted with small molecules by eliminating it from the cell membrane. The novel approach used here led to the discovery that perhexiline ablates HER3 expression, and offers an opportunity to identify HER3 ablation modulators as innovative therapeutics to improve survival in breast cancer patients.


Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Perhexiline/pharmacology , Receptor, ErbB-3/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Mice , Neuregulins/metabolism , Neuregulins/pharmacology , Protein Transport/drug effects , Proteolysis/drug effects , Receptor, ErbB-3/genetics , Signal Transduction/drug effects , Tumor Burden/drug effects , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
11.
PLoS One ; 8(7): e70338, 2013.
Article En | MEDLINE | ID: mdl-23936190

Neuregulin, previously known as ARIA, is a signaling protein involved in cell survival, synaptic plasticity, cell communication and differentiation. Neuregulin has also been described as a potent inducer of acetylcholine receptor transcription in muscle and although both neuregulin and acetylcholine have been individually described to have neuroprotective roles, their relationship in the cholinergic anti-inflammatory pathway of the brain has not been examined. Using three cell lines, BV-2, EOC-20 and RAW 264.7, we investigated the role that neuregulin signaling through the Erb family of tyrosine kinases may play in the anti-inflammatory process mediated by the α7 nicotinic acetylcholine receptors. Here we show that ErbB4 is expressed in all of our cell lines and is phosphorylated upon treatment with neuregulin. Neuregulin treatment further increases the expression of α7 nicotinic acetylcholine receptors in the microglial lines tested. Given the central role of α7 nicotinic acetylcholine receptors in regulating system inflammation we analyzed the expression of several pro-inflammatory cytokines in our system. Using ELISAs for TNF-α and IL-6 we show that treatment with NRG can produce a nearly a 33% decrease in the levels of tumor necrosis factor-α secreted by activated microglia and a nearly 88% decrease in IL-6. Given these results we propose a neuroprotective role for neuregulin wherein it modulates the expression of TNF-α and thus inflammation in the CNS via the upregulation of α7 nicotinic acetylcholine receptor expression in microglia in vitro. We suggest that the disregulation of neuregulin expression may be pivotal in neurological disorders characterized by inflammation.


Gene Expression Regulation/drug effects , Microglia/drug effects , Microglia/metabolism , Neuregulins/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/genetics , Animals , Cell Line , Cytokines/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/immunology , Phosphorylation , Receptor, ErbB-4 , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism
12.
Biochim Biophys Acta ; 1833(12): 2690-2702, 2013 Dec.
Article En | MEDLINE | ID: mdl-23845988

TTF-1 is an important transcription factor in lung development and lung disease and is essential for lung cell differentiation, specifically surfactant protein (Sftp) expression. The molecular mechanisms that drive the expression and transcriptional control of TTF-1 are not fully understood. In the fetal lung, ErbB4 functions as a transcriptional co-factor and regulates the timely onset of fetal Sftp expression. We speculate that ErbB4 is an upstream regulator of TTF-1 and regulates Sftpb expression via this pathway in alveolar type II cells. Neuregulin-induced ErbB4 and TTF-1 signaling interactions were studied by co-immunoprecipitation and confocal microscopy. Overexpression of ErbB4 and TTF-1 was analyzed in its effect on cell viability, Sftpb expression, TTF-1 expression, and Sftpb and TTF-1 promoter activity. The effect of ErbB4 deletion and ErbB4 nuclear translocation on TTF-1 expression was studied in primary fetal type II epithelial cells, isolated from transgenic HER4(heart(-/-)) mice. ErbB4 ligand neuregulin induces ErbB4 and TTF-1 co-precipitation and nuclear colocalization. Combined ErbB4 and TTF-1 overexpression inhibits cell viability, while promoting Sftpb expression more than single overexpression of each protein. NRG stimulates TTF-1 expression in ErbB4-overexpressing epithelial cells, while this effect is absent in ErbB4-depleted cells. In primary fetal type II cells, ErbB4 nuclear translocation is critical for its regulation of TTF-1-induced Sftpb upregulation. TTF-1 overexpression did not overcome this important requirement. We conclude that ErbB4 is a critical upstream regulator of TTF-1 in type II epithelial cells and that this interaction is important for Sftpb regulation.


Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , DNA-Binding Proteins/metabolism , ErbB Receptors/metabolism , Fetus/cytology , Alveolar Epithelial Cells/drug effects , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/genetics , ErbB Receptors/chemistry , Fetus/drug effects , Fetus/metabolism , Gene Expression Regulation, Developmental/drug effects , Humans , Immunoprecipitation , Mice , Models, Biological , Neuregulins/pharmacology , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary , Protein Transport/drug effects , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-4 , Transcription Factors
13.
PLoS One ; 8(6): e66720, 2013.
Article En | MEDLINE | ID: mdl-23776695

BACKGROUND: Activation of ErbB2/4 receptor tyrosine kinases in cardiomyocytes by neuregulin treatment is associated with improvement in cardiac function, supporting its use in human patients with heart failure despite the lack of a specific mechanism. Neuregulin infusion in rodents increases cardiac myosin light chain kinase (cMLCK) expression and cardiac myosin regulatory light chain (RLC) phosphorylation which may improve actin-myosin interactions for contraction. We generated a cMLCK knockout mouse to test the hypothesis that cMLCK is necessary for neuregulin-induced improvement in cardiac function by increasing RLC phosphorylation. PRINCIPAL FINDINGS: The cMLCK knockout mice have attenuated RLC phosphorylation and decreased cardiac performance measured as fractional shortening. Neuregulin infusion for seven days in wildtype mice increased cardiac cMLCK protein expression and RLC phosphorylation while increasing Akt phosphorylation and decreasing phospholamban phosphorylation. There was no change in fractional shortening. In contrast, neuregulin infusion in cMLCK knockout animals increased cardiac performance in the absence of cMLCK without increasing RLC phosphorylation. In addition, CaMKII signaling appeared to be enhanced in neuregulin-treated knockout mice. CONCLUSIONS: Thus, Neuregulin may improve cardiac performance in the failing heart without increasing cMLCK and RLC phosphorylation by activating other signaling pathways.


Heart/drug effects , Myosin-Light-Chain Kinase/deficiency , Myosin-Light-Chain Kinase/metabolism , Neuregulins/pharmacology , Animals , Male , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myosin-Light-Chain Kinase/genetics , Phosphorylation/drug effects , RNA, Messenger/genetics , Signal Transduction/drug effects
14.
Endocrinology ; 154(7): 2385-92, 2013 Jul.
Article En | MEDLINE | ID: mdl-23610133

Islet cell growth and function are affected by ligands from the epidermal growth factor (EGF) family. We describe here the expression, regional distribution, and effect on growth and secretion of insulin of a subset of these, the neuregulin (NRG) family. The expression of NRG1α, NRG1ß, NRG2α, NRG2ß, NRG3, and NRG4 in rat islets was determined using immunohistochemical and double immunofluorescent staining. We also report the expression of the 4 receptors and the remaining 7 ligands using immunohistochemistry. The NRG1α splice variant was expressed in ß-cells and the NRG1ß variant mainly in α-cells. NRG3 was also predominantly present in α-cells. Most of the members of the EGF family of ligands were also expressed, with Epigen being present at the highest levels. The rat islet-derived cell line CRI-G1 was used to study the effect of addition of EGF, NRG1ß, NRG3, and NRG4 on cell growth and insulin secretion. Synthetic refolded NRG3 strongly stimulated the growth of the CRI-G1 cells, and NRG4 gave the greatest stimulation of insulin release. Different members of the NRG family are therefore potentially potent stimuli for islet cell growth and insulin release and differ in expression in α- and ß-cells.


Islets of Langerhans/metabolism , Neuregulins/metabolism , Neuregulins/pharmacology , Animals , Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , Female , Insulin/metabolism , Male , Nerve Growth Factors/metabolism , Neuregulin-1/metabolism , Rats , Rats, Sprague-Dawley
15.
PLoS One ; 7(7): e40674, 2012.
Article En | MEDLINE | ID: mdl-22815787

During nervous system development different cell-to-cell communication mechanisms operate in parallel guiding migrating neurons and growing axons to generate complex arrays of neural circuits. How such a system works in coordination is not well understood. Cross-regulatory interactions between different signalling pathways and redundancy between them can increase precision and fidelity of guidance systems. Immunoglobulin superfamily proteins of the NCAM and L1 families couple specific substrate recognition and cell adhesion with the activation of receptor tyrosine kinases. Thus it has been shown that L1CAM-mediated cell adhesion promotes the activation of the EGFR (erbB1) from Drosophila to humans. Here we explore the specificity of the molecular interaction between L1CAM and the erbB receptor family. We show that L1CAM binds physically erbB receptors in both heterologous systems and the mammalian developing brain. Different Ig-like domains located in the extracellular part of L1CAM can support this interaction. Interestingly, binding of L1CAM to erbB enhances its response to neuregulins. During development this may synergize with the activation of erbB receptors through L1CAM homophilic interactions, conferring diffusible neuregulins specificity for cells or axons that interact with the substrate through L1CAM.


Immunoglobulins/chemistry , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/metabolism , Neuregulins/pharmacology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction/drug effects , Animals , Cell Adhesion/drug effects , HEK293 Cells , Humans , MCF-7 Cells , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary , Rats , Repetitive Sequences, Amino Acid , Structure-Activity Relationship
16.
Proc Natl Acad Sci U S A ; 109(32): 13118-23, 2012 Aug 07.
Article En | MEDLINE | ID: mdl-22822214

The neuregulin/ErbB signaling network is genetically associated with schizophrenia and modulates hippocampal γ oscillations--a type of neuronal network activity important for higher brain processes and altered in psychiatric disorders. Because neuregulin-1 (NRG-1) dramatically increases extracellular dopamine levels in the hippocampus, we investigated the relationship between NRG/ErbB and dopamine signaling in hippocampal γ oscillations. Using agonists for different D1- and D2-type dopamine receptors, we found that the D4 receptor (D4R) agonist PD168077, but not D1/D5 and D2/D3 agonists, increases γ oscillation power, and its effect is blocked by the highly specific D4R antagonist L-745,870. Using double in situ hybridization and immunofluorescence histochemistry, we show that hippocampal D4R mRNA and protein are more highly expressed in GAD67-positive GABAergic interneurons, many of which express the NRG-1 receptor ErbB4. Importantly, D4 and ErbB4 receptors are coexpressed in parvalbumin-positive basket cells that are critical for γ oscillations. Last, we report that D4R activation is essential for the effects of NRG-1 on network activity because L-745,870 and the atypical antipsychotic clozapine dramatically reduce the NRG-1-induced increase in γ oscillation power. This unique link between D4R and ErbB4 signaling on γ oscillation power, and their coexpression in parvalbumin-expressing interneurons, suggests a cellular mechanism that may be compromised in different psychiatric disorders affecting cognitive control. These findings are important given the association of a DRD4 polymorphism with alterations in attention, working memory, and γ oscillations, and suggest potential benefits of D4R modulators for targeting cognitive deficits.


Brain Waves/physiology , Dopamine/metabolism , Hippocampus/physiology , Neuregulins/metabolism , Receptors, Dopamine D4/metabolism , Signal Transduction/physiology , Animals , Dopamine/pharmacology , Fluorescent Antibody Technique , Fourier Analysis , Hippocampus/drug effects , Immunohistochemistry , In Situ Hybridization , Interneurons/metabolism , Neuregulins/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D4/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction
17.
PLoS One ; 7(5): e36828, 2012.
Article En | MEDLINE | ID: mdl-22606295

BACKGROUND: Growth factors activating the ErbB receptors have been described in prostate tumors. The androgen dependent prostate cancer cell line, LNCaP, expresses the ErbB-1, ErbB-2 and ErbB-3 receptor tyrosine kinases. Previously, it was demonstrated that NRG activates ErbB-2/ErbB-3 heterodimers to induce LNCaP cell death, whereas, EGF activates ErbB-1/ErbB-1 or ErbB-1/ErbB-2 dimers to induce cell growth and survival. It was also demonstrated that PI3K inhibitors repressed this cell death suggesting that in androgen deprived LNCaP cells, NRG activates a PI3K-dependent pathway associated with cell death. METHODOLOGY/PRINCIPAL FINDINGS: In the present study we demonstrate that NRG induces autophagy in LNCaP cells, using LC3 as a marker. However, the autophagy induced by NRG may be incomplete since p62 levels elevate. We also demonstrated that NRG- induced autophagy is independent of mammalian target of rapamycin (mTOR) inhibition since NRG induces Akt and S6K activation. Interestingly, inhibition of reactive oxygen species (ROS) by N-acetylcysteine (NAC), inhibited NRG-induced autophagy and cell death. Our study also identified JNK and Beclin 1 as important components in NRG-induced autophagy and cell death. NRG induced elevation in JNK phosphorylation that was inhibited by NAC. Moreover, inhibitor of JNK inhibited NRG-induced autophagy and cell death. Also, in cells overexpressing Bcl-2 or cells expressing sh-RNA against Beclin 1, the effects of NRG, namely induction of autophagy and cell death, were inhibited. CONCLUSIONS/SIGNIFICANCE: Thus, in LNCaP cells, NRG-induces incomplete autophagy and cell death that depend on ROS levels. These effects of NRG are mediated by signaling pathway that activates JNK and Beclin 1, but is independent of mTOR inhibition.


Neuregulins/physiology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Anthracenes/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy/physiology , Beclin-1 , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/physiopathology , Neuregulins/pharmacology , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/physiology
18.
Development ; 138(15): 3159-68, 2011 Aug.
Article En | MEDLINE | ID: mdl-21715427

The transition from mitosis to meiosis is unique to germ cells. In murine embryonic ovaries and juvenile testes, retinoic acid (RA) induces meiosis via the stimulated by retinoic acid gene 8 (Stra8), but its molecular pathway requires elucidation. We present genetic evidence in vivo and in vitro that neuregulins (NRGs) are essential for the proliferation of spermatogonia and the initiation of meiosis. Tamoxifen (TAM) was injected into 14-day post-partum (dpp) Sertoli cell-specific conditional Nrg1(Ser-/-) mutant mice. TAM induced testis degeneration, suppressed BrdU incorporation into spermatogonia and pre-leptotene primary spermatocytes, and decreased and increased the number of STRA8-positive and TUNEL-positive cells, respectively. In testicular organ cultures from 5-6 dpp wild-type mice and cultures of their re-aggregated spermatogonia and Sertoli cells, FSH, RA [all-trans-retinoic acid (ATRA), AM580, 9-cis-RA] and NRG1 promoted spermatogonial proliferation and meiotic initiation. However, TAM treatment of testicular organ cultures from the Nrg1(Ser-/-) mutants suppressed spermatogonial proliferation and meiotic initiation that was promoted by FSH or AM580. In re-aggregated cultures of purified spermatogonia, NRG1, NRG3, ATRA and 9-cis-RA promoted their proliferation and meiotic initiation, but neither AM580 nor FSH did. In addition, FSH, RAs and NRG1 promoted Nrg1 and Nrg3 mRNA expression in Sertoli cells. These results indicate that in juvenile testes RA and FSH induced meiosis indirectly through Sertoli cells when NRG1 and NRG3 were upregulated, as NRG1 amplified itself and NRG3. The amplified NRG1 and NRG3 directly induced meiosis in spermatogonia. In addition, ATRA and 9-cis-RA activated spermatogonia directly and promoted their proliferation and eventually meiotic initiation.


Cell Proliferation , Meiosis/physiology , Neuregulins/metabolism , Spermatogonia/physiology , Testis/cytology , Animals , Benzoates/pharmacology , Cell Differentiation/physiology , Cell Proliferation/drug effects , Follicle Stimulating Hormone/pharmacology , Male , Mice , Mice, Inbred C57BL , Mutation , Neuregulins/genetics , Neuregulins/pharmacology , Oncogene Proteins v-erbB/genetics , Oncogene Proteins v-erbB/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatogonia/cytology , Teratogens/pharmacology , Testis/drug effects , Testis/pathology , Testis/physiology , Tetrahydronaphthalenes/pharmacology , Tissue Culture Techniques , Tretinoin/pharmacology
19.
J Cell Physiol ; 226(11): 3014-21, 2011 Nov.
Article En | MEDLINE | ID: mdl-21302299

Neuregulin (NRG), a member of the epidermal growth factor family, plays important roles in the development of the nervous system and heart, and in cancer progression. Recent reports have suggested that NRG is involved in wound healing in keratinocytes, although the cellular mechanisms remain unclear. Here, we showed that NRG treatment increased slingshot-1L (SSH-1L)-mediated cofilin dephosphorylation and activation in HaCaT keratinocytes. Additionally, Rac1 activation and NADPH-oxidase (Nox)-dependent reactive oxygen species (ROS) generation, both known to be upstream regulators of the SSH-cofilin pathway, were increased in NRG-stimulated HaCaT cells. Inhibition of Rac1 or Nox activity blocked NRG-induced cofilin activation and cell migration by HaCaT cells. Moreover, the effects of Rac1 on cofilin activation were dependent on Nox activity. These findings indicate that NRG-induced HaCaT cell migration via the ROS-SSH-1L-cofilin pathway is activated as a consequence of Rac1 and Nox activation.


Cell Movement/drug effects , Keratinocytes/drug effects , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Neuregulins/pharmacology , rac1 GTP-Binding Protein/metabolism , Actin Depolymerizing Factors/metabolism , Cell Line , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Keratinocytes/physiology , Membrane Glycoproteins/antagonists & inhibitors , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors
20.
Breast Cancer Res Treat ; 126(2): 311-8, 2011 Apr.
Article En | MEDLINE | ID: mdl-20419393

The final signal for triggering the formation of lamellipodia that initiate directional migration of mammalian cells is binding of the Wiskott-Aldrich syndrome (WASP)/WASP family verproline-homologous protein 2 (WAVE2) to the actin-related protein 2 and 3 (Arp2/3) complex. This WAVE2-Arp2/3 signal is suggested to be enhanced in some breast cancers, facilitating invasion, and/or metastasis. Here, we demonstrated one cause of the enhanced signal using four breast cancer cell lines (SKBR3, AU565, MCF7, and MDA-MB-231). The WAVE2-Arp2/3 signal was estimated semi-quantitatively by counting the number of lamellipodia expressing both WAVE2 and Arp2 using high-power confocal laser microscopy. Higher expression of the WAVE2-Arp2/3 signal was detected in SKBR3 and AU565, which have HER2 gene amplification, than in the other two cell lines that lack HER2 gene amplification. Trastuzumab suppressed both the formation of lamellipodia and migration in a Boyden chamber experiment in SKBR3 and AU565. When the HER2 gene was transfected into MCF7, the number of both lamellipodia and migrated cells was increased. This enhancement of migration did not occur in the presence of extracellular matrix, and zymographic analysis showed no clear difference between HER2 gene-transfected cells and MCF7 cells. Immunohistochemical analysis of 115 cases of breast cancer revealed that coexpression of WAVE2 and Arp2 was significantly correlated with HER2-overexpression (P < 0.0001). These data indicate that an abnormal signal resulting from HER2 gene amplification activates lamellipodia formation in breast cancer cells, which initiates their metalloproteinase-independent migration.


Actin-Related Protein 2-3 Complex/metabolism , Breast Neoplasms/pathology , Cell Movement , Matrix Metalloproteinase 2/metabolism , Receptor, ErbB-2/biosynthesis , Recombinant Proteins/biosynthesis , Signal Transduction , Wiskott-Aldrich Syndrome Protein Family/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , ErbB Receptors/metabolism , Extracellular Matrix/metabolism , Female , Humans , Neoplasm Invasiveness , Neuregulins/pharmacology , Phosphorylation , Pseudopodia/drug effects , Pseudopodia/metabolism , Trastuzumab
...