Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 464
Filter
1.
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
2.
J Integr Neurosci ; 22(4): 96, 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37519170

ABSTRACT

OBJECTIVE: Few studies have reported the direct effect of C-X-C motif chemokine ligand 10 (CXCL10) and Neuregulin 1 (Nrg1) on neurons after spinal cord injury (SCI). This study reports the role of CXCL10 in the regulation of neuronal damage after SCI and the potential therapeutic effect of Nrg1. METHODS: The expression level of CXCL10 and Nrg1 in SCI mice was analyzed in the Gene Expression Omnibus DataSets, followed by immunohistochemical confirmation using a mouse SCI model. HT22 cells and NSC34 cells were treated with CXCL10 and Nrg1, individually or in combination, and then assayed for cell viability. The percentage of wound closure was determined through the cell scratch injury model using HT22 and NSC34 cells. Potential molecular mechanisms were also tested in response to either the individual administration of CXCL10 and Nrg1 or a mixture of both molecules. RESULTS: CXCL10 expression was significantly increased in both young and old mice subjected to SCI, while Nrg1 expression was significantly decreased. CXCL10 induced a decrease in cell viability, which was partially reversed by Nrg1. CXCL10 failed to inhibit scratch healing in HT22 and NSC34 cells, while Nrg1 promoted scratch healing. At the molecular level, CXCL10-activated cleaved caspase 9 and cleaved caspase 3 were both inhibited by Nrg1 through pERK1/2 signaling in HT22 and NSC34 cells. CONCLUSIONS: CXCL10 is upregulated in SCI. Despite the negative effect on cell viability, CXCL10 failed to inhibit the scratch healing of HT22 and NSC34 cells. Nrg1 may protect neurons by partially antagonizing the effect of CXCL10.


Subject(s)
Neuregulin-1 , Spinal Cord Injuries , Animals , Disease Models, Animal , Neuregulin-1/pharmacology , Neurons/metabolism , Signal Transduction , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Mice
3.
J Cell Biochem ; 124(9): 1273-1288, 2023 09.
Article in English | MEDLINE | ID: mdl-37450666

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive primary brain malignancy. Studies have shown that autophagy-related (ATG) genes play important roles in regulating GBM malignancy. However, the mechanism still needs to be fully elucidated. Based on clinical and gene expression information of GBM patients downloaded from The The Cancer Genome Atlas database, Kaplan-Meier, univariate Cox regression, least absolute shrinkage and selection operator regression and multivariate Cox regression were applied to construct a risk signature for GBM prognosis, followed by validation using receiver operating characteristic analysis. Next, Cell Counting Kit-8, wound healing assay, flow cytometry, monodansyl cadaverine autophagy staining assay, immunofluorescence staining and western blot, either in the absence or presence of ERBB2/AKT/mTOR inhibitors, were carried out in GBM U87 cell line to explore molecular pathway underlying GBM malignancy. A three-ATG-gene signature (HIF1A, ITGA3, and NGR1) was constructed for GBM prognosis with the greatest contribution from NRG1. In vitro experiments showed that NRG1 promoted U87 cell migration and proliferation by inhibiting autophagy, and ERBB2/AKT/mTOR is a downstream pathway that mediates the autophagy-inhibitory effects of NRG1. We constructed an ATG gene prognostic model for GBM and demonstrated that NRG1 inhibited autophagy by activating ERBB2/AKT/mTOR, promoting GBM malignancy, thus providing new insights into the molecular contribution of autophagy in GBM malignancy.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Prognosis , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Brain Neoplasms/pathology , Autophagy , Biomarkers , Cell Line, Tumor , Neuregulin-1/pharmacology , Receptor, ErbB-2/genetics
4.
Arch Biochem Biophys ; 743: 109631, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37276924

ABSTRACT

Immobilization-induced Neuromuscular Dysfunction (NMD) increases morbidity and mortality of patients in Intensive Care Units. However, the underlying mechanism of NMD remain poorly elucidated which limited the development of therapeutic method for NMD. Here we developed an immobilization rat model and tested the hypothesis that decreased expression of NRG-1, abnormal expression and distribution of nicotinic acetylcholine receptors (nAChRs) in skeletal muscle caused by immobilization can lead to NMD. To investigate the role of NRG-1/ErbB pathway on immobilization-induced NMD, exogenous recombinant human neuregulin-1 (rhNRG-1) was used to increase the expression of NRG-1 in skeletal muscle during immobilization. It was observed rhNRG-1 significantly alleviated the muscle loss and enhanced the expression of ε-nAChR, while diminished the expression of γ- and α7-nAChR and NMD. Interestingly, ErbB inhibitor PD158780 blocked the protective effects of rhNRG-1. Collectively, the results of present study suggested that rhNRG-1 attenuated immobilization-induced muscle loss and NMD, suppressed γ- and α7-nAChR production, enhanced ε-nAChR synthesis via activating NRG-1/ErbB pathway. Taken together, our findings provide novel insights into NMD contribution, suggesting that the rhNRG-1 is a promising therapy to protect against immobilization-induced myopathy.


Subject(s)
Muscular Diseases , Neuregulin-1 , Rats , Humans , Animals , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Signal Transduction
5.
Interdiscip Sci ; 15(3): 452-464, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37389721

ABSTRACT

Diabetes mellitus significantly contributes to breast cancer progression, where hyperglycemia upregulates specific genes, leading to more aggressive tumor growth. In patients with BC that develop diabetes, neuregulin 1 (NRG1) and epidermal growth factor receptor 3 (ERBB3) overexpression exacerbate tumor growth and progression. Since the interaction between NRG1 and ERBB3 is critical for tumor growth, understanding the molecular mechanisms underlying NRG1-ERBB3 complex formation is essential for elucidating diabetes-assisted breast cancer progression. However, the key residues forming the NRG1-ERBB3 complex remain unknown. Here, we substituted specific residues in NRG1 with alanine and studied its interactions with ERBB3 using computational structural biology tools. We further screened the South African natural compounds database to target the complex's interface residues to discover potential inhibitors. The conformational stability and dynamic features of NRG1-WT, -H2A, -L3A, and -K35A complexed with ERBB3 were subjected to 400 ns molecular dynamics simulations. The free binding energies of all NRG1-ERBB3 complexes were calculated using the molecular mechanics-generalized Born surface area (MM/GBSA). The H2 and L3 alanine substitutions caused a loss of interaction with ERBB3 residue D73, weakening the interaction with ERBB3. Screening 1300 natural compounds identified four (SANC00643, SANC00824, SANC00975, and SANC00335) with the best potential to inhibit ERRB3-NRG1 coupling. The binding free energies for each complex were - 48.55 kcal/mol for SANC00643, - 47.68 kcal/mol for SANC00824, - 46.04 kcal/mol for SANC00975, and - 45.29 kcal/mol for SANC00335, showing their overall stronger binding with ERBB3 than NRG1 and their potential to act as ERBB3-NRG1 complex inhibitors. In conclusion, this complex may represent a residue-specific drug target to inhibit BC progression.


Subject(s)
Breast Neoplasms , Diabetes Mellitus , Humans , Female , Breast Neoplasms/drug therapy , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
6.
Int J Biol Macromol ; 242(Pt 2): 124765, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37156315

ABSTRACT

Among the EGFR family of receptors, HER3 is considered as a pseudo-kinase which primarily interacts with HER2 in presence of heregulin-1ß. We identified two hotspot mutations i.e. G284R and D297Y and one double mutant HER2-S310F/HER3-G284R in breast cancer patients. Long term MDS (7.5 µs) revealed that HER3-D297Y and HER2-S310F:HER3-G284R do not allow the interaction with HER2 as these mutations cause dramatic conformational changes in its flanking regions. This results in formation of an unstable HER2-WT:HER3-D297Y heterodimer, thereby abrogating the downstream signalling by AKT. We found that His228 and Ser300 of HER3-D297Y form stable interactions with Glu245 and Tyr270 of EGFR-WT, in the presence of either EGF or heregulin-1ß. Applying TRIM-ing mediated direct knockdown of endogenous EGFR protein, specificity of the unconventional EGFR:HER3-D297Y interaction was validated. Due to this unusual ligand mediated interaction, cancer cells were found susceptible to EGFR targeted therapeutics i.e. Gefitinib and Erlotinib. Further, in TCGA analysis, BC patients harbouring HER3-D297Y mutation showed increased p-EGFR levels as compared to the patients harbouring HER3-WT and HER3-G284R mutations. For the first time, this comprehensive study showed the importance of specific hotspot mutations in HER3 dimerization domain can defy the Trastuzumab therapy, rather cells become susceptible to the EGFR inhibitors.


Subject(s)
Breast Neoplasms , Receptor, ErbB-3 , Female , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Dimerization , ErbB Receptors/genetics , ErbB Receptors/metabolism , Mutation , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
7.
EMBO Rep ; 24(5): e56689, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37009825

ABSTRACT

The growth factor Neuregulin-1 (NRG-1) regulates myocardial growth and is currently under clinical investigation as a treatment for heart failure. Here, we demonstrate in several in vitro and in vivo models that STAT5b mediates NRG-1/EBBB4-stimulated cardiomyocyte growth. Genetic and chemical disruption of the NRG-1/ERBB4 pathway reduces STAT5b activation and transcription of STAT5b target genes Igf1, Myc, and Cdkn1a in murine cardiomyocytes. Loss of Stat5b also ablates NRG-1-induced cardiomyocyte hypertrophy. Dynamin-2 is shown to control the cell surface localization of ERBB4 and chemical inhibition of Dynamin-2 downregulates STAT5b activation and cardiomyocyte hypertrophy. In zebrafish embryos, Stat5 is activated during NRG-1-induced hyperplastic myocardial growth, and chemical inhibition of the Nrg-1/Erbb4 pathway or Dynamin-2 leads to loss of myocardial growth and Stat5 activation. Moreover, CRISPR/Cas9-mediated knockdown of stat5b results in reduced myocardial growth and cardiac function. Finally, the NRG-1/ERBB4/STAT5b signaling pathway is differentially regulated at mRNA and protein levels in the myocardium of patients with pathological cardiac hypertrophy as compared to control human subjects, consistent with a role of the NRG-1/ERBB4/STAT5b pathway in myocardial growth.


Subject(s)
Dynamin II , Neuregulin-1 , Mice , Humans , Animals , Dynamin II/metabolism , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Zebrafish/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Hypertrophy
8.
Theriogenology ; 195: 209-216, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36368115

ABSTRACT

Oocyte in vitro maturation (IVM) is still a major challenge in human and animal assisted reproduction. Gradual instead of abrupt activation of the ovulatory cascade during IVM has been proposed to enhance nuclear-cytoplasmic synchrony and cumulus-oocyte communication, thus favoring oocyte developmental competence. Herein, we assessed the effects of neuregulin 1 (NRG1), an EGF-like factor that modulates EGFR signaling, on oocyte nuclear maturation dynamics, cumulus expansion and expression of mRNAs regulating these processes during IVM, as well as on post-IVF embryo development following AREG-stimulated IVM in cattle. In experiment 1, cumulus-oocyte complexes (COCs) were subjected to IVM with graded doses of NRG1 (1, 10 or 100 ng/mL) for 6, 9, 12, 20, and 24 h, after which oocyte nuclear status and cumulus mRNA expression were assessed. At 6 h of IVM, NRG1 at 1 ng/mL significantly decreased the percentage of GVBD (germinal vesicle breakdown) oocytes without altering later meiotic dynamics or the percentage of oocytes achieving meiosis II. In experiment 2, adding NRG1 (1 ng/mL) to the IVM medium did not affect cumulus expansion but increased the percentage of expanded and hatched blastocysts, and blastocyst total cell number following IVF/IVC. NRG1 decreased EGFR mRNA abundance while increasing NPR2 and PTX3 mRNA levels at 9 h, and TNFAIP6 mRNA abundance at 20 h of IVM. This is the first study that reports the modulatory effect of NGR1 during oocyte maturation in a mono-ovulatory species and demonstrates that this action may be applied during IVM to improve post-IVF embryo development.


Subject(s)
Neuregulin-1 , Oocytes , Humans , Animals , Cattle , Neuregulin-1/pharmacology , RNA, Messenger , Embryonic Development , ErbB Receptors , Fertilization in Vitro/veterinary
9.
Apoptosis ; 28(1-2): 124-135, 2023 02.
Article in English | MEDLINE | ID: mdl-36241947

ABSTRACT

The Notch signaling pathway is related to endothelial dysfunction in coronary atherosclerosis. Our objective was to explore the role of Notch signaling in coronary microvascular dysfunction (CMD). CMD models were constructed by sodium laurate injection in vivo and homocysteine (Hcy) stimulation in vitro. The binding ability of Notch Intracellular Domain (NICD)/H3K9Ac/GCN5 (General Control Non-derepressible 5) to Neuregulin-1 (Nrg-1) promoter was examined by chromatin immunoprecipitation. Immunofluorescence staining was conducted to detect CD31 positive cells, NICD localization, and co-localization of NICD and GCN5. Flow cytometry and Tunel staining were conducted to identify the apoptosis. Hematoxylin and eosin staining, quantitative real-time PCR, western blot, immunohistochemical staining, co-immunoprecipitation, and double luciferase report analysis were also conducted. Notch signaling pathway-related protein levels were decreased, levels of Nrg-1 and the phosphorylation of ErbB2 and ErbB4 were enhanced in CMD models. Interference with Nrg-1 further increased the apoptosis in Hcy-induced cardiac microvascular endothelial cells (CMECs). Meanwhile, the activation of the Notch signaling pathway increased the levels of Nrg-1 and the phosphorylation of ErbB2 and ErbB4, as well as inhibited the apoptosis induced by Hcy. Furthermore, NICD and histone acetyltransferase enzyme GCN5 could regulate Nrg-1 promoter activity by affecting the expression of acetylation-modified protein H3K9Ac. In addition, NICD also interacted with GCN5. In vivo results also confirmed that the activation of the Notch signal alleviated CMD. Notch signaling pathway regulates Nrg-1 level through synergistic interaction with GCN5, thereby mitigating CMD.


Subject(s)
Endothelial Cells , Myocardial Ischemia , Humans , Endothelial Cells/metabolism , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Histone Code , Apoptosis , Signal Transduction , Receptor, ErbB-4/metabolism , Myocardial Ischemia/metabolism , Receptor, Notch1
10.
Oncogene ; 42(7): 491-500, 2023 02.
Article in English | MEDLINE | ID: mdl-36357571

ABSTRACT

Pancreatic stellate cells (PSCs) are key to the treatment-refractory desmoplastic phenotype of pancreatic ductal adenocarcinoma (PDAC) and have received considerable attention as a stromal target for cancer therapy. This approach demands detailed understanding of their pro- and anti-tumourigenic effects. Interrogating PSC-cancer cell interactions in 3D models, we identified nuclear FGFR1 as critical for PSC-led invasion of cancer cells. ChIP-seq analysis of FGFR1 in PSCs revealed a number of FGFR1 interaction sites within the genome, notably NRG1, which encodes the ERBB ligand Neuregulin. We show that nuclear FGFR1 regulates transcription of NRG1, which in turn acts in autocrine fashion through an ERBB2/4 heterodimer to promote invasion. In support of this, recombinant NRG1 in 3D model systems rescued the loss of invasion incurred by FGFR inhibition. In vivo we demonstrate that, while FGFR inhibition does not affect the growth of pancreatic tumours in mice, local invasion into the pancreas is reduced. Thus, FGFR and NRG1 may present new stromal targets for PDAC therapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , Up-Regulation , Neuregulin-1/genetics , Neuregulin-1/pharmacology , Pancreatic Stellate Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/genetics
11.
Tissue Cell ; 80: 101985, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36459840

ABSTRACT

Schwann cells (SCs) are the major component of myelin sheath in the peripheral nervous system, which are necessary in the development, function maintenance, and repair of peripheral nerves. This study aimed to investigate the potential mechanism of low-intensity pulsed ultrasound (LIPUS) affecting the proliferation and myelinating activity of SCs. Rat Schwann cell line RSC96 were cultured and exposed to LIPUS of different duty ratios (control, 20 %, 50 %, 80 %). Results demonstrated that LIPUS with a duty ratio of 50 % showing the maximal effect in facilitating proliferation of SCs. The expressions of Krox20 and myelin basic protein (MBP), the key molecules of SC myelination, and the potent inducer of myelination neuregulin 1 (NRG1) and its receptors ErbB2 and ErbB3 increased significantly by LIPUS. The reaction of these factors to LIPUS were both time- and duty ratio-dependent: namely LIPUS with higher duty ratios took effects when applied repeatedly over more consecutive days. These observations indicated that NRG1/ErbB signaling pathway might contribute to the effects of LIPUS on the proliferation and myelinating status of SCs, which could be one of the mechanisms in the protective role of LIPUS in nerve repair and regeneration. Our work provided novel insights for promising strategies of nerve repair therapy.


Subject(s)
Neuregulin-1 , Schwann Cells , Animals , Rats , Cell Proliferation/genetics , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Schwann Cells/metabolism , Signal Transduction , Ultrasonic Waves , ErbB Receptors
12.
Colloids Surf B Biointerfaces ; 220: 112936, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36265312

ABSTRACT

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


Subject(s)
Myocardial Infarction , Neuregulin-1 , Tissue Scaffolds , Humans , Fibrin , Heart , Myocardial Infarction/drug therapy , Myocardium , Myocytes, Cardiac , Neuregulin-1/pharmacology , Neuregulin-1/therapeutic use
13.
Inflammopharmacology ; 30(5): 1891-1907, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35876932

ABSTRACT

Schizophrenia is a common mental disorder affecting patients' thoughts, behavior, and cognition. Recently, the NRG1/ErbB4 signaling pathway emerged as a candidate therapeutic target for schizophrenia. This study investigates the effects of aripiprazole and sertindole on the NRG1/ErbB4 and PI3K/AKT/mTOR signaling pathways in ketamine-induced schizophrenia in rats. Young male Wistar rats received ketamine (30 mg/kg, intraperitoneally) for 5 consecutive days and aripiprazole (3 mg/kg, orally) or sertindole (2.5 mg/kg, orally) for 14 days. The proposed pathway was investigated by injecting LY294002 (a selective PI3K inhibitor) (25 µg/kg, intrahippocampal injection) 30 min before the drugs. Twenty-four hours after the last injection, animals were subjected to behavioral tests: the open field test, sucrose preference test, novel object recognition task, and social interaction test. Both aripiprazole and sertindole significantly ameliorated ketamine-induced schizophrenic-like behavior, as expected, because of their previously demonstrated antipsychotic activity. Besides, both drugs alleviated ketamine-induced oxidative stress and neurotransmitter level changes in the hippocampus. They also increased the gamma-aminobutyric acid and glutamate levels and glutamate decarboxylase 67 and parvalbumin mRNA expression in the hippocampus. Moreover, aripiprazole and sertindole increased the NRG1 and ErbB4 mRNA expression levels and PI3K, p-Akt, and mTOR protein expression levels. Interestingly, pre-injecting LY294002 abolished all the effects of the drugs. This study reveals that the antipsychotic effects of aripiprazole and sertindole are partly due to oxidative stress reduction as well as NRG1/ErbB4 and PI3K/AKT/mTOR signaling pathways activation. The NRG1/ErbB4 and PI3K signaling pathways may offer a new therapeutic approach for treating schizophrenia in humans.


Subject(s)
Antipsychotic Agents , Ketamine , Schizophrenia , Animals , Antipsychotic Agents/adverse effects , Aripiprazole/adverse effects , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/pharmacology , Glutamate Decarboxylase/therapeutic use , Glutamates/adverse effects , Humans , Imidazoles , Indoles , Ketamine/pharmacology , Male , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Parvalbumins/adverse effects , Parvalbumins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Receptor, ErbB-4/therapeutic use , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Signal Transduction , Sucrose/adverse effects , TOR Serine-Threonine Kinases/metabolism , gamma-Aminobutyric Acid
14.
J Ovarian Res ; 15(1): 86, 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35883098

ABSTRACT

BACKGROUND: Granulosa cells (GCs) are multilayered somatic cells within the follicle that provide physical support and microenvironment for the developing oocyte. In recent years, the role of Neuregulin-1 (NRG1), a member of the EGF-like factor family, has received considerable attention due to its neurodevelopmental and cardiac function. However, the exact physiological role of NRG1 in GC is mainly unknown. In order to confirm that NRG1 plays a regulatory role in rat GC functions, endogenous NRG1-knockdown studies were carried out in GCs using RNA interference methodology. RESULTS: Knockdown of NRG1 in GCs resulted in the enhanced expression and secretion of the cytokines and chemokines. In addition, the phosphorylation of PI3K/Akt/ERK1/2 was significantly low in GCs under these experimental conditions. Moreover, in vitro experimental studies suggest that tumor necrosis factor-α (TNFα) treatment causes the physical destruction of GCs by activating caspase-3/7 activity. In contrast, exogenous NRG1 co-treatment of GCs delayed the onset of TNFα-induced apoptosis and inhibited the activation of caspase-3/7 activity. Furthermore, current experimental studies suggest that gonadotropins promote differential expression of NRG1 and ErbB3 receptors in GCs of the antral follicle. Interestingly, NRG1 and ErbB3 were intensely co-localized in the mural and cumulus GCs and cumulus-oocyte complex of pre-ovulatory follicles in the estrus stage. CONCLUSIONS: The present studies suggest that gonadotropins-dependent NRG1-signaling in GCs may require the balance of the cytokines and chemokines expression and secretion, ultimately which may be supporting the follicular maturation and oocyte competence for ovulation and preventing follicular atresia.


Subject(s)
Cytokines , Neuregulin-1 , Animals , Caspase 3 , Chemokines/genetics , Chemokines/metabolism , Cytokines/metabolism , Female , Follicular Atresia , Gonadotropins/pharmacology , Granulosa Cells/metabolism , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism
15.
Ecotoxicol Environ Saf ; 241: 113771, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35716406

ABSTRACT

Environmental sanitation of maternal contact during pregnancy is extremely important for the development of different fetal tissues and organs. In particular, during early pregnancy, any adverse exposure may cause abnormal fetal growth or inhibit the development of embryogenic organs. The potential risks of phthalate exposure, which affects the development of humans and animals, are becoming a serious concern worldwide. However, the specific molecular mechanism of di-(2-ethylhexyl) phthalate (DEHP)-induced cardiotoxicity in fetal mice remains unclear. In this study, animal models of DEHP gavage at concentrations of 250, 500, and 1000 mg/kg/day within 8.5-18.5 days of pregnancy were established. The cell proliferation, survival, and apoptosis rates were evaluated using CCK8, EdU, TUNEL and flow cytometry. The molecular mechanism was assessed via transcriptome sequencing, immunohistochemistry, immunofluorescence, reverse transcription-quantitative polymerase chain reaction, and Western blot analysis. In vivo, DEHP increased apoptosis, decreased Ki67 and CD31 expression, reduced heart weight and area, slowed down myocardial sarcomere development, and caused cardiac septal defect in fetal mice heart. Transcriptome sequencing showed that DEHP decreased NRG1 expression and downregulated the ErbB2/ErbB4-PI3K/AKT signaling pathway-related target genes. In vitro, primary cardiomyocytes were cultured with DEHP at a concentration of 150 µg/mL combined with ErbB inhibitor (AG1478, 10 µmol/L) and/or NRG1 protein (100 ng/mL) for 72 h. After DEHP intervention, the expression of NRG1 and the phosphorylation level of ErbB2, ErbB4, PI3K, and AKT decreased, and the apoptosis-related protein levels increased. Moreover, the apoptosis rate increased. After adding exogenous NRG1, the phosphorylation level of the NRG1/ERbB2/ERbB4-PI3K/AKT pathway increased, and the apoptosis-related protein levels decreased. Further, the apoptosis rate reduced. Interestingly, after exposure to DEHP and AG1478 + NRG1, the anti-apoptotic effect of NRG1 and cardiomyocyte proliferation decreased by inhibiting the NRG1/ERbB2/ERbB4-PI3K/AKT pathway. Hence, the NRG1-dependent regulation of the ERbB2/ERbB4-PI3K/AKT signaling pathway may be a key mechanism of DEHP-induced myocardial cytotoxicity.


Subject(s)
Diethylhexyl Phthalate , Proto-Oncogene Proteins c-akt , Animals , Diethylhexyl Phthalate/toxicity , Female , Fetus , Humans , Mice , Neuregulin-1/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phthalic Acids , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2 , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Receptor, ErbB-4/pharmacology , Signal Transduction
16.
Neuron ; 110(14): 2315-2333.e6, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35561677

ABSTRACT

Heat perception enables acute avoidance responses to prevent tissue damage and maintain body thermal homeostasis. Unlike other modalities, how heat signals are processed in the spinal cord remains unclear. By single-cell gene profiling, we identified ErbB4, a transmembrane tyrosine kinase, as a novel marker of heat-sensitive spinal neurons in mice. Ablating spinal ErbB4+ neurons attenuates heat sensation. These neurons receive monosynaptic inputs from TRPV1+ nociceptors and form excitatory synapses onto target neurons. Activation of ErbB4+ neurons enhances the heat response, while inhibition reduces the heat response. We showed that heat sensation is regulated by NRG1, an activator of ErbB4, and it involves dynamic activity of the tyrosine kinase that promotes glutamatergic transmission. Evidence indicates that the NRG1-ErbB4 signaling is also engaged in hypersensitivity of pathological pain. Together, these results identify a spinal neuron connection consisting of ErbB4+ neurons for heat sensation and reveal a regulatory mechanism by the NRG1-ErbB4 signaling.


Subject(s)
Hot Temperature , Neuregulin-1 , Neurons , Thermosensing , Animals , Mice , Neuregulin-1/pharmacology , Neurons/physiology , Receptor, ErbB-4/genetics
17.
Mol Biol Rep ; 49(5): 3975-3986, 2022 May.
Article in English | MEDLINE | ID: mdl-35166983

ABSTRACT

BACKGROUND: The inflammatory response caused by microglia in the central nervous system plays an important role in Alzheimer's disease. Neuregulin-1 (NRG1) is a member of the neuregulin family and has been demonstrated to have anti-inflammatory properties. The relationship between NRG1, microglia phenotype and neuroinflammation remains unclear. MATERIALS AND METHODS: BV2 cells were used to examine the mechanism of NRG1 in regulating microglia polarization. Neuronal apoptosis, inflammatory factors TNF-α and iNOS, microglia polarization, ErbB4 and NF-κB p65 expression were assessed. RESULTS: We found that exogenous NRG1 treatment or overexpression improved microglial activity and reduced the secretion of the inflammatory factors TNF-α and iNOS in vitro. The expression of Bax in SH-SY5Y neuron cells incubated with medium collected from the NRG1 treatment group decreased. Additionally, our study showed that NRG1 treatment reduced the levels of the M1 microglia markers CD120 and iNOS and increased the levels of the M2 microglia markers CD206 and Arg-1. Furthermore, we observed that NRG1 treatment attenuated Aß-induced NF-κB activation and promoted the expression of p-ErbB4 and that knockdown of ErbB4 abrogated the effects of NRG1 on NF-κB, Bax levels and M2 microglial polarization. CONCLUSION: NRG1 inhibits the release of inflammatory factors in microglia and regulates the switching of the M1/M2 microglia phenotype, most likely via ErbB4-dependent inhibition of the NF-κB pathway.


Subject(s)
Microglia , NF-kappa B , Neuregulin-1 , Neuroblastoma , Receptor, ErbB-4 , Cell Polarity/drug effects , Humans , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , NF-kappa B/metabolism , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Neuroblastoma/metabolism , Phenotype , Receptor, ErbB-4/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
Transl Psychiatry ; 12(1): 83, 2022 02 26.
Article in English | MEDLINE | ID: mdl-35220393

ABSTRACT

Childhood obesity leads to hippocampal atrophy and altered cognition. However, the molecular mechanisms underlying these impairments are poorly understood. The neurotrophic factor neuregulin-1 (NRG1) and its cognate ErbB4 receptor play critical roles in hippocampal maturation and function. This study aimed to determine whether exogenous NRG1 administration reduces hippocampal abnormalities and neuroinflammation in rats exposed to an obesogenic Western-like diet (WD). Lewis rats were randomly divided into four groups (12 rats/group): (1) control diet+vehicle (CDV); (2) CD + NRG1 (CDN) (daily intraperitoneal injections: 5 µg/kg/day; between postnatal day, PND 21-PND 41); (3) WD + VEH (WDV); (4) WD + NRG1 (WDN). Neurobehavioral assessments were performed at PND 43-49. Brains were harvested for MRI and molecular analyses at PND 49. We found that NRG1 administration reduced hippocampal volume (7%) and attenuated hippocampal-dependent cued fear conditioning in CD rats (56%). NRG1 administration reduced PSD-95 protein expression (30%) and selectively reduced hippocampal cytokine levels (IL-33, GM-CSF, CCL-2, IFN-γ) while significantly impacting microglia morphology (increased span ratio and reduced circularity). WD rats exhibited reduced right hippocampal volume (7%), altered microglia morphology (reduced density and increased lacunarity), and increased levels of cytokines implicated in neuroinflammation (IL-1α, TNF-α, IL-6). Notably, NRG1 synergized with the WD to increase hippocampal ErbB4 phosphorylation and the tumor necrosis alpha converting enzyme (TACE/ADAM17) protein levels. Although the results did not provide sufficient evidence to conclude that exogenous NRG1 administration is beneficial to alleviate obesity-related outcomes in adolescent rats, we identified a potential novel interaction between obesogenic diet exposure and TACE/ADAM17-NRG1-ErbB4 signaling during hippocampal maturation. Our results indicate that supraoptimal ErbB4 activities may contribute to the abnormal hippocampal structure and cognitive vulnerabilities observed in obese individuals.


Subject(s)
Neuregulin-1 , Pediatric Obesity , Animals , Anxiety , Diet , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Neuroinflammatory Diseases , Rats , Rats, Inbred Lew
19.
Cell Biol Int ; 46(5): 792-805, 2022 May.
Article in English | MEDLINE | ID: mdl-35077607

ABSTRACT

Neuregulin-1 (NRG-1) can promote the proliferation, migration, and angiogenesis of multiple stem cells, as well as prohibit cell apoptosis. In the present study, we aimed to explore the effects of NRG-1 on the proliferation, migration, apoptosis, angiogenic, and osteogenic differentiation of periodontal ligament stem cells (PDLSCs) in vitro. The expression of erythroblastic leukemia viral oncogene homolog 2 (ERBB2), ERBB3, and ERBB4 on PDLSCs were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and immunofluorescence. The effects of NRG-1 on the proliferation, migration, apoptosis, angiogenic and osteogenic differentiation of PDLSCs were assessed by cell proliferation and viability assays, transwell migration assay, flow cytometry assay, tubule formation assay, alkaline phosphatase (ALP) activity, and Alizarin Red S staining, respectively. Gene expression of angiogenesis and osteogenesis-related markers were detected by qRT-PCR. Among the ERBB family members, ERBB2 had the highest expression level in PDLSCs. Further, 10 ng/ml NRG-1 exhibited the maximal effect on proliferation, migration and remarkably inhibited the apoptosis of PDLSCs (p < .05). Moreover, NRG-1 upregulated the expression of vascular endothelial growth factor (VEGF), platelet/endothelial cell adhesion molecule-1 (CD31), hypoxia-inducible factor (HIF), kinase insert domain-containing receptor (KDR) in a dose-dependent manner as well as induced more tube formation. However, NRG-1 did not affect osteogenesis (p > .05). In summary, our study demonstrated that NRG-1 promotes the proliferation, migration, and angiogenesis and inhibits the apoptosis of PDLSCs in vitro and can potentially be used in tissue engineering for periodontal regeneration.


Subject(s)
Osteogenesis , Periodontal Ligament , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Periodontal Ligament/metabolism , Stem Cells , Vascular Endothelial Growth Factor A/metabolism
20.
J Thorac Cardiovasc Surg ; 164(6): e493-e510, 2022 12.
Article in English | MEDLINE | ID: mdl-34922752

ABSTRACT

OBJECTIVES: Right ventricular (RV) failure is a leading cause of death in patients with congenital heart disease. RV failure is kept at bay during childhood. Limited proliferation of cardiomyocytes is present in the postnatal heart. We propose that cardiomyocyte proliferation improves RV adaptation to pressure load (PL). We studied adaptation in response to increased RV PL and the role of increased cardiomyocyte cell cycle activity (CCA) in rat pups growing into adulthood. METHODS: We induced RV PL at day of weaning in rats (3 weeks; 30-40 g) by pulmonary artery banding and followed rats into adulthood (300 g). We performed histological analyses and RNA sequencing analysis. To study the effects of increased cardiomyocyte cell cycle activity, we administered neuregulin-1 (NRG1), a growth factor involved in cardiac development. RESULTS: PL induced an increase in CCA, with subsequent decline of CCA (sham/PL at 4 weeks: 0.14%/0.83%; P = .04 and 8 weeks: 0.00%/0.00%; P = .484) and cardiac function (cardiac index: control/PL 4 weeks: 4.41/3.29; P = .468 and 8 weeks: 3.57/1.44; P = .024). RNA sequencing analysis revealed delayed maturation and increased CCA pathways. NRG1 stimulated CCA (PL vehicle/NRG1 at 2 weeks: 0.62%/2.28%; P = .003), improved cardiac function (cardiac index control vs vehicle/NRG1 at 2 weeks: 4.21 vs 3.07/4.17; P = .009/.705) and postponed fibrosis (control vs vehicle/NRG1 at 4 weeks: 1.66 vs 4.82%/2.97%; P = .009/.078) in RV PL rats during childhood. CONCLUSIONS: RV PL during growth induces a transient CCA increase. Further CCA stimulation improves cardiac function and delays fibrosis. This proof-of-concept study shows that stimulation of CCA can improve RV adaptation to PL in the postnatal developing heart and might provide a new approach to preserve RV function in patients with congenital heart disease.


Subject(s)
Heart Failure , Ventricular Dysfunction, Right , Rats , Animals , Hypertrophy, Right Ventricular/metabolism , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/prevention & control , Ventricular Dysfunction, Right/metabolism , Ventricular Pressure/physiology , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Ventricular Function, Right , Myocytes, Cardiac/metabolism , Fibrosis , Heart Failure/metabolism , Cell Cycle , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...