1
|
Guan Y, Gao F, Chen B, Yu T, Meng L, Chen Q, Xiao X. Soluble TREM2 ameliorates pathological phenotypes in ischemic stroke models via modulating neuronal and microglial functions. Exp Brain Res 2025; 243:149. [PMID: 40379866 DOI: 10.1007/s00221-025-07094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/25/2025] [Indexed: 05/19/2025]
Abstract
Although the neuroprotective effects of triggering receptor expressed on myeloid cell 2 (TREM2) upregulation after ischemic stroke has been demonstrated, the level change and effect of soluble TREM2 (sTREM2) derived from proteolytic cleavage of the TREM2 extracellular domain in ischemic stroke remain unknown. In our study, the level and function of sTREM2 were detected in neuron-microglia co-cultures subjected to oxygen glucose deprivation (OGD) and in the ischemic striatum of C57BL/6 J mice in a transient middle cerebral artery occlusion (tMCAO) model. sTREM2's effect on neuronal nitric oxide synthase (nNOS)-postsynaptic density protein-95 (PSD-95) interaction was determined by co-immunoprecipitation. The microglial-activated morphology in the striatum was identified by immunohistochemistry. Quantitative real-time polymerase chain reactionwas used to detect the transcriptional levels of TREM2, shorter variant TREM2, insulin-like growth factor 1, interleukin (IL)-4, and IL-13. Levels of sTREM2, generated through the cleavage of full-length TREM2 at the His157-Ser158 peptide bond, declined after OGD and tMCAO. sTREM2 reduced neuronal death after OGD and alleviated brain infarction and neurological deficits after tMCAO by disrupting the nNOS-PSD-95 interaction, promoting microglial activation, and increasing the expression of some cytokines associated with microglial polarization towards an anti-inflammatory phenotype. To the best of our knowledge, this is the first study to suggest that sTREM2 protects against transient cerebral ischemia.
Collapse
Affiliation(s)
- Yanfei Guan
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, 523808, China.
- Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| | - Feng Gao
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, 523808, China
- Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China
| | - Bo Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, 523808, China
- Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China
| | - Tiansheng Yu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, 523808, China
| | - Linxin Meng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, 523808, China
| | - Qingzhuang Chen
- Department of Clinical Pharmacy, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, 510800, China
| | - Xiaodan Xiao
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, 523808, China.
- Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Huang J, Shen Q, Wang Z, Ni S, Sun F, Hua Y, Huang J. The influence of the NRG1/ERBB4 signaling pathway on pulmonary artery endothelial cells. Pulm Circ 2024; 14:e12439. [PMID: 39411231 PMCID: PMC11475022 DOI: 10.1002/pul2.12439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
This study aimed to examine the influence of the Neuregulin-1 (NRG1)/ERBB4 signaling pathway on the function of human pulmonary artery endothelial cells (HPAECs) and investigate the underlying mechanisms. Enzyme-linked immunosorbent assay indicated that ERBB4 levels in the serum of patients with pulmonary embolism (PE) were significantly higher than those of healthy controls (p < 0.05). In cellular studies, thrombin stimulation for 6 h led to a significant decrease in cell viability and overexpression of ERBB4 compared to control (p < 0.05). In the NRG1 group, apoptosis of HPAECs was reduced (p < 0.05), accompanied by a decrease in ERBB4 expression and an increase in p-ERBB4, phosphorylated serine/threonine kinase proteins (Akt) (p-Akt), and p-phosphoinositide 3-kinase (PI3K) expression (p < 0.05). In the AG1478 group, there was a significant increase in HPAEC apoptosis and a significant decrease in p-ERBB4 and ERBB4 expression compared to the Con group (p < 0.05). In the AG1478 + NRG1 group, there was an increase in the apoptosis rate and a significant decrease in the expression of p-ERBB4, ERBB4, p-Akt, and phosphorylated PI3K compared to the NRG1 group (p < 0.05). In animal studies, the PE group showed an increase in the expression of ERBB4 and p-ERBB4 compared to the Con group (p < 0.05). NRG1 treatment led to a significant reduction in embolism severity with decreased ERBB4 expression and increased p-ERBB4 expression (p < 0.05). Gene set enrichment analysis identified five pathways that were significantly associated with high ERBB4 expression, including CHOLESTEROL HOMEOSTASIS, OXIDATIVE PHOSPHORYLATION, and FATTY ACID METABOLISM (p < 0.05). Therefore, NRG1 inhibits apoptosis of HPAECs, accompanied by a decrease in ERBB4 and an increase in p-ERBB4. NRG1 inhibition in HPAECs apoptosis can be partially reversed by inhibiting ERBB4 expression with AG1478. ERBB4 has the potential to be a novel biological marker of PE.
Collapse
Affiliation(s)
- Jin‐Bo Huang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Qin Shen
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Zhi‐Qi Wang
- Jiangnan University Affiliated Wuxi Fifth People's HospitalWuxiJiangsuChina
| | - Song‐Shi Ni
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Fei Sun
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Yun Hua
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Jian‐An Huang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
3
|
Fan J, Zhong L, Yan F, Li X, Li L, Zhao H, Han Z, Wang R, Tao Z, Zheng Y, Ma Q, Luo Y. Alteration of N6-methyladenosine modification profiles in the neutrophilic RNAs following ischemic stroke. Neuroscience 2024; 553:56-73. [PMID: 38945353 DOI: 10.1016/j.neuroscience.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/04/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND N6-methyladenosine (m6A) is one of the most extensive RNA methylation modifications in eukaryotes and participates in the pathogenesis of numerous diseases including ischemic stroke. Peripheral blood neutrophils are forerunners after ischemic brain injury and exert crucial functions. This study aims to explore the transcriptional profiles of m6A modification in neutrophils of patients with ischemic stroke. RESULTS We found that the expression levels of m6A regulators FTO and YTHDC1 were notably decreased in the neutrophils following ischemic stroke, and FTO expression was negatively correlated with neutrophil counts and neutrophil-to-lymphocyte ratio (NLR). The m6A mRNA&lncRNA epigenetic transcriptome microarray identified 416 significantly upregulated and 500 significantly downregulated mRNA peaks in neutrophils of ischemic stroke patients. Moreover, 48 mRNAs and 18 lncRNAs were hypermethylated, and 115 mRNAs and 29 lncRNAs were hypomethylated after cerebral ischemia. Gene ontology (GO) analysis identified that these m6A-modified mRNAs were primarily enriched in calcium ion transport, long-term synaptic potentiation, and base-excision repair. The signaling pathways involved were EGFR tyrosine kinase inhibitor resistance, ErbB, and base excision repair signaling pathway. MeRIP-qPCR validation results showed that NRG1 and GDPD1 were significantly hypermethylated, and LIG1, CHRND, lncRNA RP11-442J17.2, and lncRNA RP11-600P1.2 were significantly hypomethylated after cerebral ischemia. Moreover, the expression levels of major m6A regulators Mettl3, Fto, Ythdf1, and Ythdf3 were obviously declined in the brain and leukocytes of post-stroke mouse models. CONCLUSION This study explored the RNA m6A methylation pattern in the neutrophils of ischemic stroke patients, indicating that it is an intervention target of epigenetic regulation in ischemic stroke.
Collapse
Affiliation(s)
- Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China.
| | - Liyuan Zhong
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Feng Yan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Xue Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Lingzhi Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Haiping Zhao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Ziping Han
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Zhen Tao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Yangmin Zheng
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Qingfeng Ma
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China.
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
4
|
Noll JM, Sherafat AA, Ford GD, Ford BD. The case for neuregulin-1 as a clinical treatment for stroke. Front Cell Neurosci 2024; 18:1325630. [PMID: 38638304 PMCID: PMC11024452 DOI: 10.3389/fncel.2024.1325630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/01/2024] [Indexed: 04/20/2024] Open
Abstract
Ischemic stroke is the leading cause of serious long-term disability and the 5th leading cause of death in the United States. Revascularization of the occluded cerebral artery, either by thrombolysis or endovascular thrombectomy, is the only effective, clinically-approved stroke therapy. Several potentially neuroprotective agents, including glutamate antagonists, anti-inflammatory compounds and free radical scavenging agents were shown to be effective neuroprotectants in preclinical animal models of brain ischemia. However, these compounds did not demonstrate efficacy in clinical trials with human patients following stroke. Proposed reasons for the translational failure include an insufficient understanding on the cellular and molecular pathophysiology of ischemic stroke, lack of alignment between preclinical and clinical studies and inappropriate design of clinical trials based on the preclinical findings. Therefore, novel neuroprotective treatments must be developed based on a clearer understanding of the complex spatiotemporal mechanisms of ischemic stroke and with proper clinical trial design based on the preclinical findings from specific animal models of stroke. We and others have demonstrated the clinical potential for neuregulin-1 (NRG-1) in preclinical stroke studies. NRG-1 significantly reduced ischemia-induced neuronal death, neuroinflammation and oxidative stress in rodent stroke models with a therapeutic window of >13 h. Clinically, NRG-1 was shown to be safe in human patients and improved cardiac function in multisite phase II studies for heart failure. This review summarizes previous stroke clinical candidates and provides evidence that NRG-1 represents a novel, safe, neuroprotective strategy that has potential therapeutic value in treating individuals after acute ischemic stroke.
Collapse
Affiliation(s)
- Jessica M. Noll
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA, United States
- Nanostring Technologies, Seattle, WA, United States
| | - Arya A. Sherafat
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA, United States
| | - Gregory D. Ford
- Southern University-New Orleans, New Orleans, LA, United States
| | - Byron D. Ford
- Department of Anatomy, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
5
|
Li B, Wu J, Cao D, Cao C, Zhang J, Li X, Li H, Shen H, Yu Z. ERBB1 alleviates secondary brain injury induced by experimental intracerebral hemorrhage in rats by modulating neuronal death via PLC-γ/PKC pathway. CNS Neurosci Ther 2024; 30:e14679. [PMID: 38528842 PMCID: PMC10964039 DOI: 10.1111/cns.14679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
AIMS Intracerebral hemorrhage (ICH) is a disease with high rates of disability and mortality. The role of epidermal growth factor receptor 1 (ERBB1) in ICH was elucidated in this study. METHODS ICH model was constructed by injecting autologous arterial blood into the right basal ganglia. The protein level of ERBB1 was detected by western blot analysis. To up- and downregulation of ERBB1 in rats, intraventricular injection of a lentivirus overexpression vector of ERBB1 and AG1478 (a specific inhibitor of ERBB1) was used. The cell apoptosis, neuronal loss, and pro-inflammatory cytokines were assessed by TUNEL, Nissl staining, and ELISA. Meanwhile, behavioral cognitive impairment of ICH rats was evaluated after ERBB1-targeted interventions. RESULTS ERBB1 increased significantly in brain tissue of ICH rats. Overexpression of ERBB1 remarkably reduced cell apoptosis and neuronal loss induced by ICH, as well as pro-inflammatory cytokines and oxidative stress. Meanwhile, the behavioral and cognitive impairment of ICH rats were alleviated after upregulation of ERBB1; however, the secondary brain injury (SBI) was aggravated by AG1478 treatment. Furthermore, the upregulation of PLC-γ and PKC in ICH rats was reversed by AG1478 treatment. CONCLUSIONS ERBB1 can improve SBI and has a neuroprotective effect in experimental ICH rats via PLC-γ/PKC pathway.
Collapse
Affiliation(s)
- Bing Li
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
- Department of Neurosurgery, Yancheng City No. 1 People's Hospital, Yancheng First HospitalAffiliated Hospital of Nanjing University Medical SchoolYanchengJiangsu ProvinceChina
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Demao Cao
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
- Department of NeurosurgeryThe Affiliated Hospital of Yangzhou UniversityYangzhouJiangsu ProvinceChina
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
- Department of Neurocritical Intensive Care UnitJiangyin Clinical College of Xuzhou Medical CollegeJiangyinJiangsu ProvinceChina
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu ProvinceChina
| |
Collapse
|
6
|
Pei X, Zhang L, Liu D, Wu Y, Li X, Cao Y, Du X. Notoginsenoside R1 attenuates brain injury in rats with traumatic brain injury: Possible mediation of apoptosis via ERK1/2 signaling pathway. PLoS One 2023; 18:e0295903. [PMID: 38109303 PMCID: PMC10727368 DOI: 10.1371/journal.pone.0295903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/24/2023] [Indexed: 12/20/2023] Open
Abstract
Traumatic brain injury (TBI) occurs worldwide and is associated with high mortality and disability rate. Apoptosis induced by TBI is one of the important causes of secondary injury after TBI. Notoginsenoside R1 (NGR1) is the main phytoestrogen extracted from Panax notoginseng. Many studies have shown that NGR1 has potent neuroprotective, anti-inflammatory, and anti-apoptotic properties and is effective in ischemia-reperfusion injury. Therefore, we investigated the potential neuroprotective effects of NGR1 after TBI and explored its molecular mechanism of action. A rat model of TBI was established using the controlled cortical impact (CCI) method. The expression levels of Bcl-2, Bax, caspase 3, and ERK1/2-related molecules in the downstream pathway were also detected by western blotting. The expression levels of pro-inflammatory cytokines were detected by real-time quantitative PCR. Nissl staining was used to clarify the morphological changes around the injury foci in rats after TBI. Fluoro-Jade B (FJB) and terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling (TUNEL) fluorescence staining were used to detect the apoptosis of neural cells in each group of rats. The results showed that NGR1 administration reduced neurological deficits after TBI, as well as brain edema and brain tissue apoptosis. It also significantly inhibited the expression of pro-inflammatory cytokines. Furthermore, NGR1 decreased the expression levels of extracellular signal-regulated kinase (ERK) and p-RSK1, which are phosphorylated after trauma. This study suggests that NGR1 can improve neuronal apoptosis in brain injury by inhibiting the ERK signaling pathway. NGR1 is a potential novel neuroprotective agent for the treatment of secondary brain injury after TBI.
Collapse
Affiliation(s)
- Xiaoxian Pei
- Department of Psychiatric, The Fourth People’s Hospital of Zhangjiagang City, Suzhou, China
- Medical College of Soochow University, Suzhou, China
| | - Ling Zhang
- Translational Medicine Center, The First People’s Hospital of Zhangjiagang City, Suzhou, China
| | - Dan Liu
- Department of Psychiatric, The Fourth People’s Hospital of Zhangjiagang City, Suzhou, China
| | - Yajuan Wu
- Department of Psychiatric, The Fourth People’s Hospital of Zhangjiagang City, Suzhou, China
| | - Xiaowei Li
- Department of Psychiatric, The Fourth People’s Hospital of Zhangjiagang City, Suzhou, China
| | - Ying Cao
- Department of Psychiatric, The Fourth People’s Hospital of Zhangjiagang City, Suzhou, China
| | - Xiangdong Du
- Department of Psychiatric, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Sun W, Wang M, Zhao J, Zhao S, Zhu W, Wu X, Li F, Liu W, Wang Z, Gao M, Zhang Y, Xu J, Zhang M, Wang Q, Wen Z, Shen J, Zhang W, Huang Z. Sulindac selectively induces autophagic apoptosis of GABAergic neurons and alters motor behaviour in zebrafish. Nat Commun 2023; 14:5351. [PMID: 37660128 PMCID: PMC10475106 DOI: 10.1038/s41467-023-41114-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 08/22/2023] [Indexed: 09/04/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs compose one of the most widely used classes of medications, but the risks for early development remain controversial, especially in the nervous system. Here, we utilized zebrafish larvae to assess the potentially toxic effects of nonsteroidal anti-inflammatory drugs and found that sulindac can selectively induce apoptosis of GABAergic neurons in the brains of zebrafish larvae brains. Zebrafish larvae exhibit hyperactive behaviour after sulindac exposure. We also found that akt1 is selectively expressed in GABAergic neurons and that SC97 (an Akt1 activator) and exogenous akt1 mRNA can reverse the apoptosis caused by sulindac. Further studies showed that sulindac binds to retinoid X receptor alpha (RXRα) and induces autophagy in GABAergic neurons, leading to activation of the mitochondrial apoptotic pathway. Finally, we verified that sulindac can lead to hyperactivity and selectively induce GABAergic neuron apoptosis in mice. These findings suggest that excessive use of sulindac may lead to early neurodevelopmental toxicity and increase the risk of hyperactivity, which could be associated with damage to GABAergic neurons.
Collapse
Affiliation(s)
- Wenwei Sun
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meimei Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jun Zhao
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Shuang Zhao
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wenchao Zhu
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Xiaoting Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Feifei Li
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wei Liu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zhuo Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meng Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Meijia Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Qiang Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, People's Republic of China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518055, China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Wenqing Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Zhibin Huang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
8
|
Neuregulin-1/PI3K signaling effects on oligodendrocyte proliferation, remyelination and behaviors deficit in a male mouse model of ischemic stroke. Exp Neurol 2023; 362:114323. [PMID: 36690057 DOI: 10.1016/j.expneurol.2023.114323] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023]
Abstract
In this study, we investigated the effect of neuregulin-1 (NRG1) on demyelination and neurological function in an ischemic stroke model, and further explored its neuroprotective mechanisms. Adult male ICR mice underwent photothrombotic ischemia surgery and were injected with NRG1 beginning 30 min after ischemia. Cylinder and grid walking tests were performed to evaluate the forepaw function. In addition, the effect of NRG1 on neuronal damage/death (Cresyl violet, CV), neuronal nuclei (NeuN), nestin, doublecortin (DCX), myelin basic protein (MBP), non-phosphorylated neurofilaments (SMI-32), adenomatous polyposis coli (APC), erythroblastic leukemia viral oncogene homolog (ErbB) 2, 4 and serine-threonine protein kinase (Akt) in cortex were evaluated using immunohistochemistry, immunofluorescence and western blot. The cylinder and grid walking tests exposed that treatment of NRG1 observably regained the forepaw function. NRG1 treatment reduced cerebral infarction, restored forepaw function, promoted proliferation and differentiation of neuron and increased oligodendrogliogenesis. The neuroprotective effect of NRG1 is involved in its activation of PI3K/Akt signaling pathway via ErbB2, as shown by the suppression of the effect of NRG1 by the PI3K inhibitor LY294002. Our results demonstrate that NRG1 is effective in ameliorating the both acute phase neuroprotection and long-term neurological functions via resumption of neuronal proliferation and differentiation and oligodendrogliogenesis in a male mouse model of ischemic stroke.
Collapse
|
9
|
Yoo JY, Kim HB, Lee YJ, Kim YJ, Yoo SY, Choi Y, Lee MJ, Kim IS, Baik TK, Lee JH, Woo RS. Neuregulin-1 reverses anxiety-like behavior and social behavior deficits induced by unilateral micro-injection of CoCl 2 into the ventral hippocampus (vHPC). Neurobiol Dis 2023; 177:105982. [PMID: 36592864 DOI: 10.1016/j.nbd.2022.105982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/28/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Neuregulin-1 (NRG1) is an epidermal growth factor family member with essential roles in the developing and adult nervous systems. In recent years, establishing evidence has collectively suggested that NRG1 is a new modulator of central nervous system (CNS) injury and disease, with multifaceted roles in neuroprotection, remyelination, neuroinflammation, and other repair mechanisms. NRG1 signaling exerts its effects via the tyrosine kinase receptors ErbB2-ErbB4. The NRG1/ErbB network in CNS pathology and repair has evolved, primarily in recent years. In the present study, we demonstrated that a unilateral microinjection of CoCl2 into the ventral hippocampus (vHPC) induced hypoxic insult and led to anxiety-related behaviors and deficit sociability in mice. NRG1 treatment significantly alleviated the CoCl2-induced increase of hypoxic-related molecules and behavioral abnormalities. Furthermore, NRG1 reduced the CoCl2-induced neuroinflammation and neuronal deficits in the vHPC or primary hippocampal neurons in mice. Collectively, these results suggest that NRG1 ameliorates hypoxia by alleviating synaptic deficits and behavioral abnormalities of the CoCl2-induced vHPC hypoxic model.
Collapse
Affiliation(s)
- Ji-Young Yoo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Han-Byeol Kim
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Ye-Ji Lee
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Yu-Jin Kim
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Seung-Yeon Yoo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Yoori Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Mi-Jo Lee
- Department of Radiation Oncology, Eulji University Hospital, Daejeon 35233, Republic of Korea
| | - In-Sik Kim
- Department of Biomedical Laboratory Science, School of Medicine and Department of Senior Healthcare, Graduate School, Eulji University, Uijeongbu 11759, Republic of Korea
| | - Tai-Kyoung Baik
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, Republic of Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon 34520, Republic of Korea.
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, Republic of Korea.
| |
Collapse
|
10
|
Guo YL, Zhai QY, Ye YH, Ren YQ, Song ZH, Ge KL, Cheng BH. Neuroprotective effects of neural stem cells pretreated with neuregulin1β on PC12 cells exposed to oxygen-glucose deprivation/reoxygenation. Neural Regen Res 2023; 18:618-625. [DOI: 10.4103/1673-5374.350207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
11
|
Spahic H, Parmar P, Miller S, Emerson PC, Lechner C, St. Pierre M, Rastogi N, Nugent M, Duck SA, Kirkwood A, Chavez-Valdez R. Dysregulation of ErbB4 Signaling Pathway in the Dorsal Hippocampus after Neonatal Hypoxia-Ischemia and Late Deficits in PV + Interneurons, Synaptic Plasticity and Working Memory. Int J Mol Sci 2022; 24:ijms24010508. [PMID: 36613949 PMCID: PMC9820818 DOI: 10.3390/ijms24010508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal hypoxic-ischemic (HI) injury leads to deficits in hippocampal parvalbumin (PV)+ interneurons (INs) and working memory. Therapeutic hypothermia (TH) does not prevent these deficits. ErbB4 supports maturation and maintenance of PV+ IN. Thus, we hypothesized that neonatal HI leads to persistent deficits in PV+ INs, working memory and synaptic plasticity associated with ErbB4 dysregulation despite TH. P10 HI-injured mice were randomized to normothermia (NT, 36 °C) or TH (31 °C) for 4 h and compared to sham. Hippocampi were studied for α-fodrin, glial fibrillary acidic protein (GFAP), and neuroregulin (Nrg) 1 levels; erb-b2 receptor tyrosine kinase 4 (ErbB4)/ Ak strain transforming (Akt) activation; and PV, synaptotagmin (Syt) 2, vesicular-glutamate transporter (VGlut) 2, Nrg1, and ErbB4 expression in coronal sections. Extracellular field potentials and behavioral testing were performed. At P40, deficits in PV+ INs correlated with impaired memory and coincided with blunted long-term depression (LTD), heightened long-term potentiation (LTP) and increased Vglut2/Syt2 ratio, supporting excitatory-inhibitory (E/I) imbalance. Hippocampal Nrg1 levels were increased in the hippocampus 24 h after neonatal HI, delaying the decline documented in shams. Paradoxically ErbB4 activation decreased 24 h and again 30 days after HI. Neonatal HI leads to persistent deficits in hippocampal PV+ INs, memory, and synaptic plasticity. While acute decreased ErbB4 activation supports impaired maturation and survival after HI, late deficit reemergence may impair PV+ INs maintenance after HI.
Collapse
Affiliation(s)
- Harisa Spahic
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Pritika Parmar
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sarah Miller
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul Casey Emerson
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Charles Lechner
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mark St. Pierre
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Neetika Rastogi
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Nugent
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah Ann Duck
- Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Alfredo Kirkwood
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Raul Chavez-Valdez
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Correspondence:
| |
Collapse
|
12
|
Vega-Torres JD, Ontiveros-Angel P, Terrones E, Stuffle EC, Solak S, Tyner E, Oropeza M, dela Peña I, Obenaus A, Ford BD, Figueroa JD. Short-term exposure to an obesogenic diet during adolescence elicits anxiety-related behavior and neuroinflammation: modulatory effects of exogenous neuregulin-1. Transl Psychiatry 2022; 12:83. [PMID: 35220393 PMCID: PMC8882169 DOI: 10.1038/s41398-022-01788-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 11/21/2022] Open
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.
Collapse
Affiliation(s)
- Julio David Vega-Torres
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Perla Ontiveros-Angel
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Esmeralda Terrones
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Erwin C. Stuffle
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| | - Sara Solak
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Emma Tyner
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Marie Oropeza
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Ike dela Peña
- grid.43582.380000 0000 9852 649XDepartment of Pharmaceutical and Administrative Sciences, Loma Linda University Health School of Pharmacy, Loma Linda, CA USA
| | - Andre Obenaus
- grid.266093.80000 0001 0668 7243Department of Pediatrics, University of California-Irvine, Irvine, CA USA
| | - Byron D. Ford
- grid.266097.c0000 0001 2222 1582Division of Biomedical Sciences, University of California-Riverside School of Medicine, Riverside, CA USA
| | - Johnny D. Figueroa
- grid.43582.380000 0000 9852 649XCenter for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University Health School of Medicine, Loma Linda, CA USA
| |
Collapse
|
13
|
Yoo JY, Kim HB, Baik TK, Lee JH, Woo RS. Neuregulin 1/ErbB4/Akt signaling attenuates cytotoxicity mediated by the APP-CT31 fragment of amyloid precursor protein. Exp Mol Pathol 2021; 120:104622. [PMID: 33684392 DOI: 10.1016/j.yexmp.2021.104622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/15/2021] [Accepted: 03/03/2021] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by neuronal and synaptic loss. The cytoplasmic tail of amyloid precursor protein (APP) undergoes sequential cleavage at a specific intracellular caspase site to generate the cytoplasmic terminal 31 (CT31) fragment. The APP-CT31 fragment is a potent inducer of apoptosis. The cytotoxicity of APP-CT31 in SH-SY5Y cells was evaluated by the lactate dehydrogenase (LDH) assay. TUNEL staining was used to detect apoptotic signals in SH-SY5Y cells and primary cortical neurons. The expression of apoptosis-related proteins, such as p53, PUMA (p53 up-regulated modulator of apoptosis), and cleaved was investigated by immunofluorescence analysis and Western blotting. In this study, we investigated the neuroprotective effect of neuregulin 1 (NRG1) against cytotoxicity induced by APP-CT31. Our data showed that CT31 induced cytotoxicity and apoptosis in SH-SY5Y cells and primary cortical neurons. NRG1 attenuated the neurotoxicity induced by the expression of APP-CT31. We also showed that APP-CT31 altered the expression of p53 and cleaved caspase 3. However, treatment with NRG1 rescued the APP-CT31-induced upregulation of p53 and cleaved caspase 3 expression. The protective effect of NRG1 was abrogated by inhibition of the ErbB4 receptor and Akt. These results indicate an important role of ErbB4/Akt signaling in NRG1-mediated neuroprotection, suggesting that endogenous NRG1/ErbB4 signaling represents a valuable therapeutic target in AD.
Collapse
Affiliation(s)
- Ji-Young Yoo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Han-Byeol Kim
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Tai-Kyoung Baik
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon 34520, Republic of Korea.
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 301-746, Republic of Korea.
| |
Collapse
|
14
|
Yoo JY, Kim HB, Yoo SY, Yoo HI, Song DY, Baik TK, Lee JH, Woo RS. Neuregulin 1/ErbB4 signaling attenuates neuronal cell damage under oxygen-glucose deprivation in primary hippocampal neurons. Anat Cell Biol 2019; 52:462-468. [PMID: 31949986 PMCID: PMC6952697 DOI: 10.5115/acb.19.210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/25/2022] Open
Abstract
The hippocampus is one of the most important brain areas of cognition. This region is particularly sensitive to hypoxia and ischemia. Neuregulin-1 (NRG1) has been shown to be able to protect against focal cerebral ischemia. The aim of the present study was to investigate the neuroprotective effect of NRG1 in primary hippocampal neurons and its underlying mechanism. Our data showed oxygen-glucose deprivation (OGD)-induced cytotoxicity and overexpression of ErbB4 in primary hippocampal neurons. Moreover, pretreatment with NRG1 could inhibit OGD-induced overexpression of ErbB4. In addition, NRG1 significantly attenuated neuronal death induced by OGD. The neuroprotective effect of NRG1 was blocked in ischemic neurons after pretreatment with AG1478, an inhibitor of ErbB4, but not after pretreatment with AG879, an inhibitor of ErbB2. These results indicate an important role of ErbB4 in NRG1-mediated neuroprotection, suggesting that endogenous ErbB4 might serve as a valuable therapeutic target for treating global cerebral ischemia.
Collapse
Affiliation(s)
- Ji-Young Yoo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Han-Byeol Kim
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Seung-Yeon Yoo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Hong-Il Yoo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Dae-Yong Song
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Tai-Kyoung Baik
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, Eulji University College of Medicine, Daejeon, Korea
| |
Collapse
|
15
|
NRG1-ErbB4 signaling promotes functional recovery in a murine model of traumatic brain injury via regulation of GABA release. Exp Brain Res 2019; 237:3351-3362. [PMID: 31720762 DOI: 10.1007/s00221-019-05680-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/09/2019] [Indexed: 12/20/2022]
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.
Collapse
|
16
|
Ryu S, Lee JM, Bae CA, Moon CE, Cho KO. Therapeutic efficacy of neuregulin 1-expressing human adipose-derived mesenchymal stem cells for ischemic stroke. PLoS One 2019; 14:e0222587. [PMID: 31560696 PMCID: PMC6764745 DOI: 10.1371/journal.pone.0222587] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 09/02/2019] [Indexed: 12/22/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (AdMSCs) have been reported to ameliorate neurological deficits after acute ischemic stroke. As neuregulin 1 (NRG1, or heregulin 1), a growth factor with versatile functions in the central nervous system, has demonstrated protective effects against ischemic brain injuries, we have generated NRG1-overexpressing AdMSCs in order to investigate whether NRG1-AdMSCs could enhance therapeutic benefits of AdMSCs in ischemic stroke. After AdMSCs were infected with adenoviral NRG1, increased NRG1 secretion in NRG1-AdMSCs was confirmed with ELISA. At 1 d after ischemic stroke that was induced by the occlusion of middle cerebral artery (MCAo) for 60 min in Sprague Dawley (SD) rats, adenoviral NRG1, AdMSCs, NRG1-AdMSCs, or PBS were injected into the striatum and serial neurologic examinations were performed. Administration of NRG1-AdMSCs resulted in significant improvement of functional outcome following stroke compared to AdMSCs- or adenoviral NRG1-treated group, in addition to the reduction in the infarct size evaluated by hematoxylin and eosin staining. When NRG1 expression in the brain was examined by double immunofluorescence to human nuclei (HuNu)/NRG1 and ELISA, NRG1-AdMSCs demonstrated marked increase in NRG1 expression. Moreover, western blot analysis further showed that transplantation of NRG1-AdMSCs significantly increased both endogenous and adenoviral NRG1 expression compared to AdMSCs-treated group. To elucidate molecular mechanisms, NRG1-associated downstream molecules were evaluated by western blot analysis. Expression of ErbB4, a receptor for NRG1, was markedly increased by NRG1-AdMSCs administration, in addition to pMAPK and pAkt, crucial molecules of NRG1-ErbB4 signaling. Taken together, our data suggest that NRG1-AdMSCs can provide excellent therapeutic potential in ischemic stroke by activating NRG1-ErbB4 signaling network.
Collapse
Affiliation(s)
- Sun Ryu
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jae-Min Lee
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Cheong A. Bae
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Chae-Eun Moon
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Kyung-Ok Cho
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Institute of Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- * E-mail:
| |
Collapse
|
17
|
Noll JM, Li Y, Distel TJ, Ford GD, Ford BD. Neuroprotection by Exogenous and Endogenous Neuregulin-1 in Mouse Models of Focal Ischemic Stroke. J Mol Neurosci 2019; 69:333-342. [PMID: 31290093 DOI: 10.1007/s12031-019-01362-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/25/2019] [Indexed: 11/30/2022]
Abstract
Identifying novel neuroprotectants that can halt or reverse the neurological effects of stroke is of interest to both clinicians and scientists. We and others previously showed the pre-clinical neuroprotective efficacy of neuregulin-1 (NRG-1) in rats following focal brain ischemia. In this study, we examined neuroprotection by exogenous and endogenous NRG-1 using a mouse model of ischemic stroke. C57BL6 mice were subjected to middle cerebral artery occlusion (MCAO) followed by reperfusion. NRG-1 or vehicle was infused intra-arterially (i.a.) or intravenously (i.v.) after MCAO and before the onset of reperfusion. NRG-1 treatment (16 μg/kg; i.a.) reduced cerebral cortical infarct volume by 72% in mice when delivered post-ischemia. NRG-1 also inhibited neuronal injury as measured by Fluoro Jade B labeling and rescued NeuN immunoreactivity in neurons. Neuroprotection by NRG-1 was also observed in mice when administered i.v. (100 μg/kg) in both male and female mice. We investigated whether endogenous NRG-1 was neuroprotective using male and female heterozygous NRG-1 knockout mice (NRG-1+/-) compared with wild-type mice (WT) littermates. NRG-1+/- and WT mice were subjected to MCAO for 45 min, and infarct size was measured 24 h following MCAO. NRG-1+/- mice displayed a sixfold increase in cortical infarct size compared with WT mice. These results demonstrate that NRG-1 treatment mitigates neuronal damage following cerebral ischemia. We further showed that reduced endogenous NRG-1 results in exacerbated neuronal injury in vivo. These findings suggest that NRG-1 represents a promising therapy to treat stroke in human patients.
Collapse
Affiliation(s)
- Jessica M Noll
- Division of Biomedical Sciences, University of California - Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA
| | - Yonggang Li
- Division of Biomedical Sciences, University of California - Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA.,ICF, Atlanta, GA, 30329, USA
| | - Timothy J Distel
- Division of Biomedical Sciences, University of California - Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA
| | - Gregory D Ford
- Fort Valley State University, 1005 State University Dr., Fort Valley, GA, 31030, USA
| | - Byron D Ford
- Division of Biomedical Sciences, University of California - Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA.
| |
Collapse
|
18
|
Vullhorst D, Buonanno A. NMDA Receptors Regulate Neuregulin 2 Binding to ER-PM Junctions and Ectodomain Release by ADAM10 [corrected]. Mol Neurobiol 2019; 56:8345-8363. [PMID: 31240601 DOI: 10.1007/s12035-019-01659-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
Abstract
Unprocessed pro-neuregulin 2 (pro-NRG2) accumulates on neuronal cell bodies at junctions between the endoplasmic reticulum and plasma membrane (ER-PM junctions). NMDA receptors (NMDARs) trigger NRG2 ectodomain shedding from these sites followed by activation of ErbB4 receptor tyrosine kinases, and ErbB4 signaling cell-autonomously downregulates intrinsic excitability of GABAergic interneurons by reducing voltage-gated sodium channel currents. NMDARs also promote dispersal of Kv2.1 clusters from ER-PM junctions and cause a hyperpolarizing shift in its voltage-dependent channel activation, suggesting that NRG2/ErbB4 and Kv2.1 work together to regulate intrinsic interneuron excitability in an activity-dependent manner. Here we explored the cellular processes underlying NMDAR-dependent NRG2 shedding in cultured rat hippocampal neurons. We report that NMDARs control shedding by two separate but converging mechanisms. First, NMDA treatment disrupts binding of pro-NRG2 to ER-PM junctions by post-translationally modifying conserved Ser/Thr residues in its intracellular domain. Second, using a mutant NRG2 protein that cannot be modified at these residues and that fails to accumulate at ER-PM junctions, we demonstrate that NMDARs also directly promote NRG2 shedding by ADAM-type metalloproteinases. Using pharmacological and shRNA-mediated knockdown, and metalloproteinase overexpression, we unexpectedly find that ADAM10, but not ADAM17/TACE, is the major NRG2 sheddase acting downstream of NMDAR activation. Together, these findings reveal how NMDARs exert tight control over the NRG2/ErbB4 signaling pathway, and suggest that NRG2 and Kv2.1 are co-regulated components of a shared pathway that responds to elevated extracellular glutamate levels.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA.
| |
Collapse
|
19
|
Hei Y, Chen R, Mao X, Wang J, Long Q, Liu W. Neuregulin1 attenuates cognitive deficits and hippocampal CA1 neuronal apoptosis partly via ErbB4 receptor in a rat model of chronic cerebral hypoperfusion. Behav Brain Res 2019; 365:141-149. [PMID: 30826297 DOI: 10.1016/j.bbr.2019.02.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/15/2022]
Abstract
Neuregulin1 (NRG1) is an effective neuroprotectant. Previously we demonstrated that the expression of hippocampal NRG1/ErbB4 gradually decreased and correlates with neuronal apoptosis during chronic cerebral hypoperfusion (CCH). Here we aimed to further investigate the protective role of NRG1 in CCH. AG1478, an ErbB4 inhibitor, was used to explore the involvement of ErbB4 receptors in NRG1's action. Permanent bilateral common carotid artery occlusion (2VO) or sham operation was performed in Sprague-Dawley rats. NRG1 (100 μM) and AG1478 (50 mM) was administered intraventricularly. Eight weeks post-surgery, cognitive impairment was analyzed using Morris water maze (MWM) and radial arm water maze (RAWM) tests, followed by histological assessment of the survival and apoptosis of hippocampal CA1 neurons using NeuN and TUNEL immunostaining respectively. Expression of apoptosis-related proteins and ErbB4 activation (pErbB4/ErbB4) was evaluated by Western blotting. The results showed that NRG1 significantly improved the performances in MWM (spatial learning and memory) and RAWM (spatial working and reference memory), attenuated hippocampal CA1 neuronal loss and apoptosis, upregulated the expression of pErbB4/ErbB4 and the anti-apoptotic protein Bcl-2, and downregulated the expression of pro-apoptotic proteins of Cleaved (Cl)-caspase3 and Bax. In addition, the protective effects of NRG1 could be partly abolished by AG1478. Taken together, our study suggested that NRG1 ameliorates cognitive impairment and neuronal apoptosis partly via ErbB4 receptors in rats with CCH.
Collapse
Affiliation(s)
- Yue Hei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Rong Chen
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Xingang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Jiancai Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China
| | - Qianfa Long
- Department of Neurosurgery, Institute of Mini-invasive Neurosurgery and Translational Medicine, Xi'an Central Hospital, No. 185 Houzai Gate of North Street, Xi'an, 710003, PR China
| | - Weiping Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, No.17 Changle West Road, Xi'an, 710032, PR China.
| |
Collapse
|
20
|
Cespedes JC, Liu M, Harbuzariu A, Nti A, Onyekaba J, Cespedes HW, Bharti PK, Solomon W, Anyaoha P, Krishna S, Adjei A, Botchway F, Ford B, Stiles JK. Neuregulin in Health and Disease. INTERNATIONAL JOURNAL OF BRAIN DISORDERS AND TREATMENT 2018; 4:024. [PMID: 31032468 PMCID: PMC6483402 DOI: 10.23937/2469-5866/1410024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Juan Carlos Cespedes
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Mingli Liu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Adriana Harbuzariu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Annette Nti
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - John Onyekaba
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Hanna Watson Cespedes
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | | | - Wesley Solomon
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Precious Anyaoha
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| | - Sri Krishna
- ICMR-National Institute for Research in Tribal Health, India
| | - Andrew Adjei
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Ghana
| | - Felix Botchway
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Ghana
| | - Byron Ford
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, USA
| | - Jonathan K Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, USA
| |
Collapse
|
21
|
Rytel L. The Influence of Bisphenol A (BPA) on Neuregulin 1-Like Immunoreactive Nerve Fibers in the Wall of Porcine Uterus. Int J Mol Sci 2018; 19:ijms19102962. [PMID: 30274171 PMCID: PMC6213500 DOI: 10.3390/ijms19102962] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/22/2018] [Accepted: 09/24/2018] [Indexed: 12/14/2022] Open
Abstract
Bisphenol A (BPA), a substance commonly used in the manufacture of plastics, shows multidirectional negative effects on humans and animals. Due to similarities to estrogens, BPA initially leads to disorders in the reproductive system. On the other hand, it is known that neuregulin 1 (NRG-1) is an active substance which enhances the survivability of cells, inhibits apoptosis, and protects tissues against damaging factors. Because the influence of BPA on the nervous system has also been described, the aim of the present study was to investigate for the first time the influence of various doses of BPA on neuregulin 1-like immunoreactive (NRG-1-LI) nerves located in the porcine uterus using the routine single- and double-immunofluorescence technique. The obtained results have shown that BPA increases the number and affects the neurochemical characterization of NRG-1-LI in the uterus, and changes are visible even under the impact of small doses of this toxin. The character of observed changes depended on the dose of BPA and the part of the uterus studied. These observations suggest that NRG-1 in nerves supplying the uterus may play roles in adaptive and protective mechanisms under the impact of BPA.
Collapse
Affiliation(s)
- Liliana Rytel
- Department of Internal Disease with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury, ul. Oczapowskiego 14, 10-719 Olsztyn, Poland.
| |
Collapse
|
22
|
Chandrasekar A, Olde Heuvel F, Wepler M, Rehman R, Palmer A, Catanese A, Linkus B, Ludolph A, Boeckers T, Huber-Lang M, Radermacher P, Roselli F. The Neuroprotective Effect of Ethanol Intoxication in Traumatic Brain Injury Is Associated with the Suppression of ErbB Signaling in Parvalbumin-Positive Interneurons. J Neurotrauma 2018; 35:2718-2735. [PMID: 29774782 DOI: 10.1089/neu.2017.5270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ethanol intoxication (EI) is a frequent comorbidity of traumatic brain injury (TBI), but the impact of EI on TBI pathogenic cascades and prognosis is unclear. Although clinical evidence suggests that EI may have neuroprotective effects, experimental support is, to date, inconclusive. We aimed at elucidating the impact of EI on TBI-associated neurological deficits, signaling pathways, and pathogenic cascades in order to identify new modifiers of TBI pathophysiology. We have shown that ethanol administration (5 g/kg) before trauma enhances behavioral recovery in a weight-drop TBI model. Neuronal survival in the injured somatosensory cortex was also enhanced by EI. We have used phospho-receptor tyrosine kinase (RTK) arrays to screen the impact of ethanol on TBI-induced activation of RTK in somatosensory cortex, identifying ErbB2/ErbB3 among the RTKs activated by TBI and suppressed by ethanol. Phosphorylation of ErbB2/3/4 RTKs were upregulated in vGlut2+ excitatory synapses in the injured cortex, including excitatory synapses located on parvalbumin (PV)-positive interneurons. Administration of selective ErbB inhibitors was able to recapitulate, to a significant extent, the neuroprotective effects of ethanol both in sensorimotor performance and structural integrity. Further, suppression of PV interneurons in somatosensory cortex before TBI, by engineered receptors with orthogonal pharmacology, could mimic the beneficial effects of ErbB inhibitors. Thus, we have shown that EI interferes with TBI-induced pathogenic cascades at multiple levels, with one prominent pathway, involving ErbB-dependent modulation of PV interneurons.
Collapse
Affiliation(s)
| | | | - Martin Wepler
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Rida Rehman
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Annette Palmer
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Alberto Catanese
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Birgit Linkus
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Albert Ludolph
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Tobias Boeckers
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Markus Huber-Lang
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Peter Radermacher
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Francesco Roselli
- 1 Department of Neurology, Ulm University , Ulm, Germany .,4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| |
Collapse
|
23
|
Pankratova S, Klingelhofer J, Dmytriyeva O, Owczarek S, Renziehausen A, Syed N, Porter AE, Dexter DT, Kiryushko D. The S100A4 Protein Signals through the ErbB4 Receptor to Promote Neuronal Survival. Theranostics 2018; 8:3977-3990. [PMID: 30083275 PMCID: PMC6071530 DOI: 10.7150/thno.22274] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 04/10/2018] [Indexed: 12/21/2022] Open
Abstract
Understanding the mechanisms of neurodegeneration is crucial for development of therapies to treat neurological disorders. S100 proteins are extensively expressed in the injured brain but S100's role and signalling in neural cells remain elusive. We recently demonstrated that the S100A4 protein protects neurons in brain injury and designed S100A4-derived peptides mimicking its beneficial effects. Here we show that neuroprotection by S100A4 involves the growth factor family receptor ErbB4 and its ligand Neuregulin 1 (NRG), key regulators of neuronal plasticity and implicated in multiple brain pathologies. The neuroprotective effect of S100A4 depends on ErbB4 expression and the ErbB4 signalling partners ErbB2/Akt, and is reduced by functional blockade of NRG/ErbB4 in cell models of neurodegeneration. We also detect binding of S100A4 with ErbB1 (EGFR) and ErbB3. S100A4-derived peptides interact with, and signal through ErbB, are neuroprotective in primary and immortalized dopaminergic neurons, and do not affect cell proliferation/motility - features which make them promising as potential neuroprotectants. Our data suggest that the S100-ErbB axis may be an important mechanism regulating neuronal survival and plasticity.
Collapse
|
24
|
Zhou Z, Lu J, Liu WW, Manaenko A, Hou X, Mei Q, Huang JL, Tang J, Zhang JH, Yao H, Hu Q. Advances in stroke pharmacology. Pharmacol Ther 2018; 191:23-42. [PMID: 29807056 DOI: 10.1016/j.pharmthera.2018.05.012] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Stroke occurs when a cerebral blood vessel is blocked or ruptured, and it is the major cause of death and adult disability worldwide. Various pharmacological agents have been developed for the treatment of stroke either through interrupting the molecular pathways leading to neuronal death or enhancing neuronal survival and regeneration. Except for rtPA, few of these agents have succeeded in clinical trials. Recently, with the understanding of the pathophysiological process of stroke, there is a resurrection of research on developing neuroprotective agents for stroke treatment, and novel molecular targets for neuroprotection and neurorestoration have been discovered to predict or offer clinical benefits. Here we review the latest major progress of pharmacological studies in stroke, especially in ischemic stroke; summarize emerging potential therapeutic mechanisms; and highlight recent clinical trials. The aim of this review is to provide a panorama of pharmacological interventions for stroke and bridge basic and translational research to guide the clinical management of stroke therapy.
Collapse
Affiliation(s)
- Zhenhua Zhou
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA; Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Wu Liu
- Department of Diving and Hyperbaric Medicine, the Second Military Medical University, Shanghai 200433, China
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Xianhua Hou
- Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, the Second Military Medical University, Shanghai 200003, China
| | - Jun-Long Huang
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China.
| | - Qin Hu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
25
|
Hei Y, Chen R, Yi X, Wei L, Long Q, Liu W. The Expression of Hippocampal NRG1/ErbB4 Correlates With Neuronal Apoptosis, but Not With Glial Activation During Chronic Cerebral Hypoperfusion. Front Aging Neurosci 2018; 10:149. [PMID: 29875654 PMCID: PMC5974051 DOI: 10.3389/fnagi.2018.00149] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
Permanent bilateral common carotid occlusion (2VO) is well-established to investigate the chronic cerebral hypoperfusion (CCH)-induced cognitive deficits. Besides, previous studies suggested that disturbance of Neuregulin1 (NRG1)/ErbB4 signaling is associated with cognitive impairments, as well as neuronal apoptosis and neuroinflammation in CNS. However, the expression pattern of hippocampal NRG1/ErbB4 has not been systematically investigated during CCH. Here, we aim to investigate the temporal changes of hippocampal NRG1/ErbB4 during CCH and their possible relationship with neuronal apoptosis and glial activation. Morris water maze (MWM) and Radial arm water maze (RAWM) tests were used to analyze cognitive impairment in 2VO rats at 28 days post-surgery, and Enzyme-Linked Immunosorbent Assay (ELISA), western blotting and immunostaining were performed at different time points (24 h, 7 days, 14 days, 28 days) to detect the expression pattern of NRG1/ErbB4 and the distribution of ErbB4. Neuronal nuclei (NeuN), NeuN/TUNEL, Iba1 and GFAP immunostaining and caspase activity in hippocampal CA1 subarea were assessed during CCH as well. We found that the expression of NRG1 and phosphorylated ErbB4 (pErbB4)/ErbB4 changed in a time-dependent manner (up-regulated in the acute phase and then decreased in the chronic phase of CCH). Besides, ErbB4-expressed neurons and selective types of GABAergic cells decreased after CCH, but the distribution pattern of ErbB4 remained unchanged. In addition, the expression of hippocampal NRG1/ErbB4 positively correlated with the level of neuronal apoptosis (both NeuN/TUNEL immunostaining and caspase-3 activity), but not with glial activation according to Pearson’s correlation. These findings indicated that hippocampal NRG1/ErbB4 may be involved in the pathogenesis of CCH, especially neuronal apoptosis during CCH.
Collapse
Affiliation(s)
- Yue Hei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Rong Chen
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xicai Yi
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lizhou Wei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qianfa Long
- Department of Neurosurgery, Institute of Mini-invasive Neurosurgery and Translational Medicine, Xi'an Central Hospital, Xi'an, China
| | - Weiping Liu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
26
|
Zhang P, Xu R, Guo Y, Qin J, Dai Y, Liu N, Wu C. DL-3-n-butylphthalide promotes dendrite development in cortical neurons subjected to oxygen-glucose deprivation/reperfusion. Cell Biol Int 2018; 42:1041-1049. [PMID: 29696738 DOI: 10.1002/cbin.10980] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/21/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Peng Zhang
- Affiliated Bayi Brain Hospital; The PLA Army General Hospital; Beijing 100700 China
| | - Ruxiang Xu
- Affiliated Bayi Brain Hospital; The PLA Army General Hospital; Beijing 100700 China
| | - Yang Guo
- Department of Neurology; Zhujiang Hospital; Guangzhou 510282 China
| | - Jiazhen Qin
- Affiliated Bayi Brain Hospital; The PLA Army General Hospital; Beijing 100700 China
| | - Yiwu Dai
- Affiliated Bayi Brain Hospital; The PLA Army General Hospital; Beijing 100700 China
| | - Ning Liu
- Affiliated Bayi Brain Hospital; The PLA Army General Hospital; Beijing 100700 China
| | - Cuiying Wu
- Affiliated Bayi Brain Hospital; The PLA Army General Hospital; Beijing 100700 China
| |
Collapse
|
27
|
Shim JW, Madsen JR. VEGF Signaling in Neurological Disorders. Int J Mol Sci 2018; 19:ijms19010275. [PMID: 29342116 PMCID: PMC5796221 DOI: 10.3390/ijms19010275] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/06/2018] [Accepted: 01/10/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent growth factor playing diverse roles in vasculogenesis and angiogenesis. In the brain, VEGF mediates angiogenesis, neural migration and neuroprotection. As a permeability factor, excessive VEGF disrupts intracellular barriers, increases leakage of the choroid plexus endothelia, evokes edema, and activates the inflammatory pathway. Recently, we discovered that a heparin binding epidermal growth factor like growth factor (HB-EGF)—a class of EGF receptor (EGFR) family ligands—contributes to the development of hydrocephalus with subarachnoid hemorrhage through activation of VEGF signaling. The objective of this review is to entail a recent update on causes of death due to neurological disorders involving cerebrovascular and age-related neurological conditions and to understand the mechanism by which angiogenesis-dependent pathological events can be treated with VEGF antagonisms. The Global Burden of Disease study indicates that cancer and cardiovascular disease including ischemic and hemorrhagic stroke are two leading causes of death worldwide. The literature suggests that VEGF signaling in ischemic brains highlights the importance of concentration, timing, and alternate route of modulating VEGF signaling pathway. Molecular targets distinguishing two distinct pathways of VEGF signaling may provide novel therapies for the treatment of neurological disorders and for maintaining lower mortality due to these conditions.
Collapse
Affiliation(s)
- Joon W Shim
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
28
|
Yan F, Tan X, Wan W, Dixon BJ, Fan R, Enkhjargal B, Li Q, Zhang J, Chen G, Zhang JH. ErbB4 protects against neuronal apoptosis via activation of YAP/PIK3CB signaling pathway in a rat model of subarachnoid hemorrhage. Exp Neurol 2017; 297:92-100. [PMID: 28756200 DOI: 10.1016/j.expneurol.2017.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/17/2017] [Accepted: 07/25/2017] [Indexed: 12/21/2022]
Abstract
Neuronal apoptosis is a central pathological process in subarachnoid hemorrhage (SAH)-induced early brain injury. Previous studies indicated that ErbB4 (EGFR family member v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4) is essential for normal development and maintenance of the nervous system. In this study, we explored the neuroprotective effects of ErbB4 and its downstream YAP (yes-associated protein)/PIK3CB signaling pathway in early brain injury after SAH in a rat model using the endovascular perforation method. Rats were neurologically evaluated with the Modified Garcia Scale and beam balance test at 24h and 72h after SAH. An ErbB4 activator Neuregulin 1β1 (Nrg 1β1), ErbB4 siRNA and YAP siRNA were used to explore this pathway. The expression of p-ErbB4 and YAP was significantly increased after SAH. Multiple immunofluorescence labeling experiments demonstrated that ErbB4 is mainly expressed in neurons. Activation of ErbB4 and its downstream signals improved the neurological deficits after SAH and significantly reduced neuronal cell death. Inhibition of ErbB4 reduced YAP and PIK3CB expression, and aggravated cell apoptosis. YAP knockdown reduced the PIK3CB level and eliminated the anti-apoptotic effects of ErbB4 activation. These findings indicated that ErbB4 plays a neuroprotective role in early brain injury after SAH, possibly via the YAP/PIK3CB signaling pathway.
Collapse
Affiliation(s)
- Feng Yan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA; Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaoxiao Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Weifeng Wan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Brandon J Dixon
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Ruiming Fan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Qian Li
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, CA, USA.
| |
Collapse
|
29
|
Shi J, Chen X, Li H, Wu Y, Wang S, Shi W, Chen J, Ni Y. Neuron-autonomous transcriptome changes upon ischemia/reperfusion injury. Sci Rep 2017; 7:5800. [PMID: 28724924 PMCID: PMC5517505 DOI: 10.1038/s41598-017-05342-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 05/30/2017] [Indexed: 01/22/2023] Open
Abstract
Ischemic stroke and the following reperfusion, an acute therapeutic intervention, can cause irreversible brain damages. However, the underlying pathological mechanisms are still under investigation. To obtain a comprehensive, real-time view of the cell-autonomous mechanisms involved in ischemic stroke and reperfusion, we applied the next-generation sequencing (NGS) technology to characterize the temporal changes in gene expression profiles using primarily cultured hippocampal neurons under an oxygen-glucose deprivation/reperfusion (OGD/R) condition. We first identified the differentially expressed genes (DEGs) between normal cultured neurons, neurons with OGD, and neurons with OGD followed by reperfusion for 6 h, 12 h, and 18 h, respectively. We then performed bioinformatics analyses, including gene ontological (GO) and pathway analysis and co-expression network analysis to screen for novel key pathways and genes involved in the pathology of OGD/R. After we confirmed the changes of selected key genes in hippocampal cultures with OGD/R, we further validated their expression changes in an in vivo ischemic stroke model (MCAO). Finally, we demonstrated that prevention of the up-regulation of a key gene (Itga5) associated with OGD/R promoted hippocampal neuronal survival. Our research thereby provided novel insights into the molecular mechanisms in ischemic stroke pathophysiology and potential targets for therapeutic intervention after ischemic stroke.
Collapse
Affiliation(s)
- Jinlong Shi
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair and Department of Neurosurgery, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Xia Chen
- Basic Medical Research Center, Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Haiying Li
- Department of Pediatrics, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Youjia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Shouyan Wang
- Basic Medical Research Center, Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Wei Shi
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair and Department of Neurosurgery, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Jian Chen
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair and Department of Neurosurgery, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Yaohui Ni
- Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair and Department of Neurology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
30
|
Repair Injured Heart by Regulating Cardiac Regenerative Signals. Stem Cells Int 2016; 2016:6193419. [PMID: 27799944 PMCID: PMC5075315 DOI: 10.1155/2016/6193419] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 01/10/2023] Open
Abstract
Cardiac regeneration is a homeostatic cardiogenic process by which the sections of malfunctioning adult cardiovascular tissues are repaired and renewed employing a combination of both cardiomyogenesis and angiogenesis. Unfortunately, while high-quality regeneration can be performed in amphibians and zebrafish hearts, mammalian hearts do not respond in kind. Indeed, a long-term loss of proliferative capacity in mammalian adult cardiomyocytes in combination with dysregulated induction of tissue fibrosis impairs mammalian endogenous heart regenerative capacity, leading to deleterious cardiac remodeling at the end stage of heart failure. Interestingly, several studies have demonstrated that cardiomyocyte proliferation capacity is retained in mammals very soon after birth, and cardiac regeneration potential is correspondingly preserved in some preadolescent vertebrates after myocardial infarction. There is therefore great interest in uncovering the molecular mechanisms that may allow heart regeneration during adult stages. This review will summarize recent findings on cardiac regenerative regulatory mechanisms, especially with respect to extracellular signals and intracellular pathways that may provide novel therapeutics for heart diseases. Particularly, both in vitro and in vivo experimental evidences will be presented to highlight the functional role of these signaling cascades in regulating cardiomyocyte proliferation, cardiomyocyte growth, and maturation, with special emphasis on their responses to heart tissue injury.
Collapse
|
31
|
Regulation of inflammatory responses by neuregulin-1 in brain ischemia and microglial cells in vitro involves the NF-kappa B pathway. J Neuroinflammation 2016; 13:237. [PMID: 27596278 PMCID: PMC5011915 DOI: 10.1186/s12974-016-0703-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/22/2016] [Indexed: 12/20/2022] Open
Abstract
Background We previously demonstrated that neuregulin-1 (NRG-1) was neuroprotective in rats following ischemic stroke. Neuroprotection by NRG-1 was associated with the suppression of pro-inflammatory gene expression in brain tissues. Over-activation of brain microglia can induce pro-inflammatory gene expression by activation of transcriptional regulators following stroke. Here, we examined how NRG-1 transcriptionally regulates inflammatory gene expression by computational bioinformatics and in vitro using microglial cells. Methods To identify transcriptional regulators involved in ischemia-induced inflammatory gene expression, rats were sacrificed 24 h after middle cerebral artery occlusion (MCAO) and NRG-1 treatment. Gene expression profiles of brain tissues following ischemia and NRG-1 treatment were examined by microarray technology. The Conserved Transcription Factor-Binding Site Finder (CONFAC) bioinformatics software package was used to predict transcription factors associated with inflammatory genes induced following stroke and suppressed by NRG-1 treatment. NF-kappa B (NF-kB) was identified as a potential transcriptional regulator of NRG-1-suppressed genes following ischemia. The involvement of specific NF-kB subunits in NRG-1-mediated inflammatory responses was examined using N9 microglial cells pre-treated with NRG-1 (100 ng/ml) followed by lipopolysaccharide (LPS; 10 μg/ml) stimulation. The effects of NRG-1 on cytokine production were investigated using Luminex technology. The levels of the p65, p52, and RelB subunits of NF-kB and IkB-α were determined by western blot analysis and ELISA. Phosphorylation of IkB-α was investigated by ELISA. Results CONFAC identified 12 statistically over-represented transcription factor-binding sites (TFBS) in our dataset, including NF-kBP65. Using N9 microglial cells, we observed that NRG-1 significantly inhibited LPS-induced TNFα and IL-6 release. LPS increased the phosphorylation and degradation of IkB-α which was blocked by NRG-1. NRG-1 also prevented the nuclear translocation of the NF-kB p65 subunit following LPS administration. However, NRG-1 increased production of the neuroprotective cytokine granulocyte colony-stimulating factor (G-CSF) and the nuclear translocation of the NF-kB p52 subunit, which is associated with the induction of anti-apoptotic and suppression of pro-inflammatory gene expression. Conclusions Neuroprotective and anti-inflammatory effects of NRG-1 are associated with the differential regulation of NF-kB signaling pathways in microglia. Taken together, these findings suggest that NRG-1 may be a potential therapeutic treatment for treating stroke and other neuroinflammatory disorders.
Collapse
|
32
|
Lasiene J, Komine O, Fujimori-Tonou N, Powers B, Endo F, Watanabe S, Shijie J, Ravits J, Horner P, Misawa H, Yamanaka K. Neuregulin 1 confers neuroprotection in SOD1-linked amyotrophic lateral sclerosis mice via restoration of C-boutons of spinal motor neurons. Acta Neuropathol Commun 2016; 4:15. [PMID: 26891847 PMCID: PMC4758105 DOI: 10.1186/s40478-016-0286-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 12/25/2022] Open
Abstract
Introduction Increasing evidence implicates the role of the cell types surrounding motor neurons, such as interneurons and glial cells, in non-cell autonomous neurodegeneration of amyotrophic lateral sclerosis (ALS). C-boutons, the large cholinergic synapses that innervate spinal α-motor neurons to control their excitability, are progressively lost from motor neurons in both human ALS and mutant Cu/Zn superoxide dismutase 1 (SOD1)-ALS mice. Neuregulin-1 (NRG1), a trophic factor implicated in neural development, transmission, and synaptic plasticity, has been reported to localize in the synapse of C-boutons. However, the roles of NRG1 in maintenance of motor neuron health and activity, as well as the functional consequences of its alteration in motor neuron disease, are not fully understood. Results NRG1 was localized to the post-synaptic face of C-boutons and its expression was significantly lost in SOD1-ALS mice and human ALS patients. Losses of NRG1 expression and C-boutons occured almost contemporaneously in SOD1-ALS mice. In addition, expressions of ErbB3 and ErbB4, receptors for NRG1, were reduced in the motor neurons of SOD1-ALS mice. Furthermore, viral-mediated delivery of type III-NRG1 to the spinal cord restored the number of C-boutons and extended the survival time of SOD1-ALS mice. Conclusions These results suggest that maintenance of NRG1-ErbB4/3 axis by supplementation of NRG1 confers neuroprotection in motor neuron disease, partly through the maintenance of C-boutons of spinal motor neurons. Electronic supplementary material The online version of this article (doi:10.1186/s40478-016-0286-7) contains supplementary material, which is available to authorized users.
Collapse
|