Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Comp Neurol ; 529(15): 3497-3512, 2021 10.
Article in English | MEDLINE | ID: mdl-34212389

ABSTRACT

Accumulation of amyloid ß (Aß) in the brain is a hallmark of Alzheimer's disease (AD). We previously showed that ErbB4 in parvalbumin (PV)-positive interneurons was associated with Aß-induced cognitive deficits; however, the underlying mechanism remains undetermined. Here we found that specific deletion of ErbB4 in PV neurons significantly attenuated oligomeric Aß-induced neuronal toxicity and inhibited Aß-induced decreases of PSD95 and synaptophysin. Moreover, specific ablation of ErbB4 in PV neurons altered activity-related protein c-Fos and decreased hippocampal PV neurons, especially in the dentate gyrus (DG) of hAPP-J20 mice. Furthermore, c-Jun N-terminal kinase (JNK), a protein downstream of ErbB4, was activated by Aß but not ErbB4's ligand neuregulin 1 (NRG1) ß1, suggesting different downstream pathways for Aß and NRG1ß1. JNK phosphorylation was inhibited by the ErbB4 inhibitor AG1478 and by pretreatment with NRG1ß1. More importantly, siRNA knockdown of ErbB4 decreased JNK phosphorylation and expression, tau phosphorylation at Ser396 and Thr 205, and Bax expression. Therefore, ErbB4 might mediate Aß-induced neuropathology through the JNK/tau pathway and represent a potential therapeutic target in patients with AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , MAP Kinase Signaling System/physiology , Peptide Fragments/toxicity , Receptor, ErbB-4/deficiency , tau Proteins/metabolism , Alzheimer Disease/chemically induced , Alzheimer Disease/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Transgenic , Phosphorylation/drug effects , Phosphorylation/physiology , RNA, Small Interfering/administration & dosage , Receptor, ErbB-4/antagonists & inhibitors , Receptor, ErbB-4/genetics
2.
Am J Physiol Heart Circ Physiol ; 319(2): H443-H455, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32618511

ABSTRACT

Neuregulin-1 (NRG1) is a paracrine growth factor, secreted by cardiac endothelial cells (ECs) in conditions of cardiac overload/injury. The current concept is that the cardiac effects of NRG1 are mediated by activation of erythroblastic leukemia viral oncogene homolog (ERBB)4/ERBB2 receptors on cardiomyocytes. However, recent studies have shown that paracrine effects of NRG1 on fibroblasts and macrophages are equally important. Here, we hypothesize that NRG1 autocrine signaling plays a role in cardiac remodeling. We generated EC-specific Erbb4 knockout mice to eliminate endothelial autocrine ERBB4 signaling without affecting paracrine NRG1/ERBB4 signaling in the heart. We first observed no basal cardiac phenotype in these mice up to 32 wk. We next studied these mice following transverse aortic constriction (TAC), exposure to angiotensin II (ANG II), or myocardial infarction in terms of cardiac performance, myocardial hypertrophy, myocardial fibrosis, and capillary density. In general, no major differences between EC-specific Erbb4 knockout mice and control littermates were observed. However, 8 wk following TAC both myocardial hypertrophy and fibrosis were attenuated by EC-specific Erbb4 deletion, albeit these responses were normalized after 20 wk. Similarly, 4 wk after ANG II treatment, myocardial fibrosis was less pronounced compared with control littermates. These observations were supported by RNA-sequencing experiments on cultured endothelial cells showing that NRG1 controls the expression of various hypertrophic and fibrotic pathways. Overall, this study shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling. This study contributes to understanding the spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury.NEW & NOTEWORTHY The role of NRG1/ERBB signaling in endothelial cells is not completely understood. Our study contributes to the understanding of spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury and shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling.


Subject(s)
Autocrine Communication , Cardiomyopathies/metabolism , Endothelial Cells/metabolism , Hypertrophy, Left Ventricular/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-4/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Paracrine Communication , Receptor, ErbB-4/deficiency , Receptor, ErbB-4/genetics , Signal Transduction
3.
Neurosci Bull ; 36(2): 97-109, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31388929

ABSTRACT

The current study was designed to explore how disruption of specific molecular circuits in the cerebral cortex may cause sensorimotor cortico-striatal community structure deficits in both a mouse model and patients with schizophrenia. We used prepulse inhibition (PPI) and brain structural and diffusion MRI scans in 23 mice with conditional ErbB4 knockout in parvalbumin interneurons and 27 matched controls. Quantitative real-time PCR was used to assess the differential levels of GABA-related transcripts in brain regions. Concurrently, we measured structural and diffusion MRI and the cumulative contribution of risk alleles in the GABA pathway genes in first-episode treatment-naïve schizophrenic patients (n = 117) and in age- and sex-matched healthy controls (n = 86). We present the first evidence of gray and white matter impairment of right sensorimotor cortico-striatal networks and reproduced the sensorimotor gating deficit in a mouse model of schizophrenia. Significant correlations between gray matter volumes (GMVs) in the somatosensory cortex and PPI as well as glutamate decarboxylase 1 mRNA expression were found in controls but not in knockout mice. Furthermore, these findings were confirmed in a human sample in which we found significantly decreased gray and white matter in sensorimotor cortico-striatal networks in schizophrenic patients. The psychiatric risk alleles of the GABA pathway also displayed a significant negative correlation with the GMVs of the somatosensory cortex in patients. Our study identified that ErbB4 ablation in parvalbumin interneurons induced GABAergic dysregulation, providing valuable mechanistic insights into the sensorimotor cortico-striatal community structure deficits associated with schizophrenia.


Subject(s)
Cerebral Cortex/pathology , Corpus Striatum/pathology , Prepulse Inhibition/genetics , Receptor, ErbB-4/deficiency , Schizophrenia/genetics , Schizophrenia/pathology , Animals , Brain/metabolism , Brain/pathology , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , White Matter/pathology
4.
Exp Brain Res ; 237(12): 3351-3362, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31720762

ABSTRACT

Traumatic brain injury (TBI) is a serious health problem in the world. However, little is known about the pathogenesis and molecular mechanisms of TBI. Here, we show that TBI activates neuregulin 1 (NRG1)-ErbB4 signaling, with an increased expression of NRG1 and ErbB4 in the traumatic region. Specifically knocking out ErbB4 in parvalbumin-positive (PV+) interneurons exacerbates motor function deficits in mice after TBI. Consistently, PV-ErbB4-/- mice showed larger necrotic area and more edema when compared with PV-ErbB4+/+ mice. Replenishment of NRG1 through intranasal application of the recombinant protein in PV-ErbB4+/+ mice enhanced neurological function. Moreover, using an in vitro neuronal culture system, we found that NRG1-ErbB4 signaling protects neurons from glutamate-induced death, and such protective effects could be diminished by GABA receptor antagonist. These results indicate that NRG-ErbB4 signaling protects cortical neurons from TBI-induced damage, and such effect is probably mediated by promoting GABA activity. Taken together, these findings unveil a previously unappreciated role for NRG1-ErB4 signaling in preventing neuronal cell death during functional recovery after TBI.


Subject(s)
Brain Injuries, Traumatic , Glutamic Acid/metabolism , Neuregulin-1/metabolism , Neurons/metabolism , Neuroprotection/physiology , Parietal Lobe , Receptor, ErbB-4/metabolism , Recovery of Function/physiology , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Behavior, Animal/physiology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Cell Death/physiology , Cells, Cultured , Disease Models, Animal , GABA Antagonists/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuregulin-1/pharmacology , Parietal Lobe/injuries , Parietal Lobe/metabolism , Parietal Lobe/pathology , Parietal Lobe/physiopathology , Receptor, ErbB-4/deficiency
5.
J Neurosci ; 38(24): 5567-5583, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29844022

ABSTRACT

Both the amygdala and the bed nucleus of the stria terminalis (BNST) have been implicated in maladaptive anxiety characteristics of anxiety disorders. However, the underlying circuit and cellular mechanisms have remained elusive. Here we show that mice with Erbb4 gene deficiency in somatostatin-expressing (SOM+) neurons exhibit heightened anxiety as measured in the elevated plus maze test and the open field test, two assays commonly used to assess anxiety-related behaviors in rodents. Using a combination of electrophysiological, molecular, genetic, and pharmacological techniques, we demonstrate that the abnormal anxiety in the mutant mice is caused by enhanced excitatory synaptic inputs onto SOM+ neurons in the central amygdala (CeA), and the resulting reduction in inhibition onto downstream SOM+ neurons in the BNST. Notably, our results indicate that an increase in dynorphin signaling in SOM+ CeA neurons mediates the paradoxical reduction in inhibition onto SOM+ BNST neurons, and that the consequent enhanced activity of SOM+ BNST neurons is both necessary for and sufficient to drive the elevated anxiety. Finally, we show that the elevated anxiety and the associated synaptic dysfunctions and increased dynorphin signaling in the CeA-BNST circuit of the Erbb4 mutant mice can be recapitulated by stress in wild-type mice. Together, our results unravel previously unknown circuit and cellular processes in the central extended amygdala that can cause maladaptive anxiety.SIGNIFICANCE STATEMENT The central extended amygdala has been implicated in anxiety-related behaviors, but the underlying mechanisms are unclear. Here we found that somatostatin-expressing neurons in the central amygdala (CeA) controls anxiety through modulation of the stria terminalis, a process that is mediated by an increase in dynorphin signaling in the CeA. Our results reveal circuit and cellular dysfunctions that may account for maladaptive anxiety.


Subject(s)
Anxiety/physiopathology , Central Amygdaloid Nucleus/physiopathology , Neural Pathways/physiology , Septal Nuclei/physiopathology , Animals , Central Amygdaloid Nucleus/metabolism , Dynorphins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Receptor, ErbB-4/deficiency , Septal Nuclei/metabolism , Somatostatin/metabolism
6.
Genes Brain Behav ; 17(1): 56-69, 2018 01.
Article in English | MEDLINE | ID: mdl-28792672

ABSTRACT

The receptor tyrosine kinase ErbB4 and its ligand trophic factors of the neuregulin (NRG) family have been associated with schizophrenia and other mental disorders in human genetic studies. In vivo studies in mice have shown how abnormal Nrg-ErbB4 signaling leads to deviant behaviors relevant to distinct aspects of schizophrenia, including hyperactivity, sensory gating deficits, working and spatial memory deficits and impaired social behavior. However, so far little is known on the role of ErbB4 in attention and inhibitory control, two aspects of executive functions that are impaired in schizophrenia. Here we investigated the effects of constitutive loss of ErbB4 in the central nervous system of mice on performance in a 5-choice serial reaction time task (5CSRTT) assessing attention and inhibitory control. In this task, ErbB4-/- mice did not show deficits in various parameters of attention, and premature responses as measure of inhibitory control. Nonetheless, ErbB4-/- mice recapitulated a specific set of behavioral phenotypes associated with schizophrenia, including a deficit in spatial learning and memory in the Barnes Maze and in contextual fear learning, and a trend for a deficit in sensorimotor gating. Furthermore, we investigated the effect of acute pharmacological inhibition of ErbB tyrosine kinase receptor using the pan-ErbB kinase inhibitor JNJ-28871063 (JNJ), in an automated version of the 5CSRTT. JNJ did not affect attention and inhibitory control. In conclusion, our data suggest no direct involvement of a classical Nrg-ErbB4 pathway in attention and inhibitory control in mice, while it confirms the involvement of this pathway in other domains relevant to schizophrenia.


Subject(s)
Attention/physiology , Receptor, ErbB-4/antagonists & inhibitors , Receptor, ErbB-4/deficiency , Animals , Attention/drug effects , Fear/physiology , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuregulin-1/metabolism , Reaction Time , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , Signal Transduction , Synaptic Transmission
7.
Am J Physiol Renal Physiol ; 314(5): F773-F787, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28724608

ABSTRACT

Tubulointerstitial fibrosis (TIF) is a prominent factor in the progression of chronic kidney disease regardless of etiology. Avian erythroblastic leukemia viral oncogene homolog 4 (ErbB4) expression levels were inversely correlated to renal fibrosis in human fibrotic kidneys. In both unilateral ureteral obstruction (UUO) and ischemia-reperfusion injury followed by uninephrectomy (IRI/UNx) mouse models, expression levels of ErbB4 were elevated in the early stage of renal injury. Using mice with global ErbB4 deletion except for transgenic rescue in cardiac tissue ( ErbB4-/-ht+), we determined that UUO induced similar injury in proximal tubules compared with wild-type mice but more severe injury in distal nephrons. TIF was apparent earlier and was more pronounced following UUO in ErbB4-/-ht+ mice. With ErbB4 deletion, UUO injury inhibited protein kinase B phosphorylation and increased the percentage of cells in G2/M arrest. There was also increased nuclear immunostaining of yes-associated protein and increased expression of phospho-Mothers against decapentaplegic homolog 3, snail1, and vimentin. These results indicate that ErbB4 deletion accelerates the development and progression of renal fibrosis in obstructive nephropathy. Similar results were found in a mouse IRI/UNx model. In conclusion, increased expression of ErbB4 in the early stages of renal injury may reflect a compensatory effect to lessen tubulointerstitial injury.


Subject(s)
Acute Kidney Injury/etiology , Gene Deletion , Kidney/metabolism , Receptor, ErbB-4/deficiency , Renal Insufficiency, Chronic/etiology , Reperfusion Injury/etiology , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Case-Control Studies , Cell Cycle Proteins , Cell Dedifferentiation , Disease Models, Animal , Disease Progression , Fibrosis , G2 Phase Cell Cycle Checkpoints , Genetic Predisposition to Disease , Kidney/pathology , Mice, Knockout , Nephrectomy , Phenotype , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Severity of Illness Index , Signal Transduction , Smad3 Protein/metabolism , Snail Family Transcription Factors/metabolism , Time Factors , Ureteral Obstruction/complications , Vimentin/metabolism , YAP-Signaling Proteins
8.
CNS Neurosci Ther ; 23(6): 510-517, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28421673

ABSTRACT

AIMS: The receptor tyrosine kinase ErbB4 is present throughout the primate brain and has a distinct functional profile. In this study, we investigate the potential role of endothelial ErbB4 receptor signaling in the brain. RESULTS: Here, we show that the endothelial cell-specific deletion of ErbB4 induces decreased exploratory behavior in adult mice. However, the water maze task for spatial memory and the memory reconsolidation test reveal no changes; additionally, we observe no impairment in CaMKII phosphorylation in Cdh5Cre;ErbB4f/f mice, which indicates that the endothelial ErbB4 deficit leads to decreased exploratory activity rather than direct memory deficits. Furthermore, decreased brain metabolism, which was measured using micro-positron emission tomography, is observed in the Cdh5Cre;ErbB4f/f mice. Consistently, the immunoblot data demonstrate the downregulation of brain Glut1, phospho-ULK1 (Ser555), and TIGAR in the endothelial ErbB4 conditional knockout mice. Collectively, our findings suggest that endothelial ErbB4 plays a critical role in regulating brain function, at least in part, through maintaining normal brain energy homeostasis. CONCLUSIONS: Targeting ErbB4 or the modulation of endothelial ErbB4 signaling may represent a rational pharmacological approach to treat neurological disorders.


Subject(s)
Brain/physiology , Energy Metabolism/genetics , Exploratory Behavior/physiology , Memory Disorders/genetics , Receptor, ErbB-4/deficiency , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis Regulatory Proteins , Autophagy-Related Protein-1 Homolog/metabolism , Avoidance Learning/physiology , Brain/diagnostic imaging , Cadherins/genetics , Cadherins/metabolism , Endothelial Cells/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Glucose Transporter Type 1/metabolism , Interleukin-1beta/metabolism , Maze Learning/physiology , Memory/physiology , Mice , Mice, Transgenic , Neuregulin-1/metabolism , Phosphoric Monoester Hydrolases , Proteins/metabolism , Receptor, ErbB-4/genetics , Recognition, Psychology/physiology
9.
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
10.
J Comp Neurol ; 522(14): 3351-62, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24752666

ABSTRACT

Dendritic spine loss is observed in many psychiatric disorders, including schizophrenia, and likely contributes to the altered sense of reality, disruption of working memory, and attention deficits that characterize these disorders. ErbB4, a member of the EGF family of receptor tyrosine kinases, is genetically associated with schizophrenia, suggesting that alterations in ErbB4 function contribute to the disease pathology. Additionally, ErbB4 functions in synaptic plasticity, leading us to hypothesize that disruption of ErbB4 signaling may affect dendritic spine development. We show that dendritic spine density is reduced in the dorsomedial prefrontal cortex of ErbB4 conditional whole-brain knockout mice. We find that ErbB4 localizes to dendritic spines of excitatory neurons in cortical neuronal cultures and is present in synaptic plasma membrane preparations. Finally, we demonstrate that selective ablation of ErbB4 from excitatory neurons leads to a decrease in the proportion of mature spines and an overall reduction in dendritic spine density in the prefrontal cortex of weanling (P21) mice that persists at 2 months of age. These results suggest that ErbB4 signaling in excitatory pyramidal cells is critical for the proper formation and maintenance of dendritic spines in excitatory pyramidal cells.


Subject(s)
Dendritic Spines/physiology , Gene Expression Regulation/genetics , Neurons/ultrastructure , Prefrontal Cortex/cytology , Receptor, ErbB-4/deficiency , Age Factors , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Fractionation , Cells, Cultured , Dendritic Spines/metabolism , Disks Large Homolog 4 Protein , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 3/metabolism , Nestin/genetics , Nestin/metabolism , Neurons/metabolism , Prefrontal Cortex/embryology , Prefrontal Cortex/growth & development , Receptor, ErbB-4/genetics , Synapses/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...