1
|
Guo J, Xu Y, Liu J, Hou X. The involvement of lidocaine in amyloid-β1-42-dependent mitochondrial dysfunction and apoptosis in hippocampal neurons via nerve growth factor-protein kinase B pathway. Neuroreport 2024:00001756-990000000-00297. [PMID: 39445521 DOI: 10.1097/wnr.0000000000002105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
This project is conceived to reveal the role of lidocaine in the process of Alzheimer's disease (AD) and its possible downstream targets. After the employment of AD cell model in mice hippocampal neuronal HT-22 cells in the presence of amyloid-β1-42 (Aβ1-42), Cell Counting Kit-8 method investigated cell viability. Oxidative damage was assayed based on a dichloro-dihydro-fluorescein diacetate fluorescent probe and commercially available kits. The 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolocarbocyanine iodide fluorescent probe estimated mitochondrial function. Terminal-deoxynucleotidyl transferase mediated nick end labeling, western blotting, and immunofluorescence appraised the apoptotic level. Western blot also ascertained the alternations of nerve growth factors (NGF)-protein kinase B (Akt) pathway-related proteins. Aβ1-42 concentration dependently triggered the viability loss, oxidative damage, and apoptosis in HT-22 cells. Lidocaine promoted the viability and reduced the mitochondrial impairment and mitochondria-dependent apoptosis in Aβ1-42-treated HT-22 cells in a concentration-dependent manner. Besides, lidocaine activated the NGF-Akt pathway and NGF absence blocked NGF-Akt pathway, aggravated mitochondrial dysfunction as well as mitochondria-dependent apoptosis in lidocaine-administrated HT-22 cells in response to Aβ1-42. Altogether, these observations concluded that lidocaine might stimulate NGF-Akt pathway to confer protection against mitochondrial impairment and apoptosis in Aβ1-42-mediated cellular model of AD.
Collapse
Affiliation(s)
- Jianlian Guo
- Department of Surgical Anesthesiology, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | | | | | | |
Collapse
|
2
|
Soroudi S, Mousavi G, Jafari F, Elyasi S. Prevention of colistin-induced neurotoxicity: a narrative review of preclinical data. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3709-3727. [PMID: 38091077 DOI: 10.1007/s00210-023-02884-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/01/2023] [Indexed: 05/23/2024]
Abstract
Polymyxin E or colistin is an effective antibiotic against MDR Gram-negative bacteria. Due to unwanted side effects, the use of this antibiotic has been limited for a long time, but in recent years, the widespread of MDR Gram-negative bacteria infections has led to its reintroduction. Neurotoxicity and nephrotoxicity are the significant dose-limiting adverse effects of colistin. Several agents with anti-inflammatory and antioxidant properties have been used for the prevention of colistin-induced neurotoxicity. This study aims to review the preclinical studies in this field to prepare guidance for future human studies. The data was achieved by searching PubMed, Scopus, and Google Scholar databases. All eligible pre-clinical studies performed on neuroprotective agents against colistin-induced neurotoxicity, which were published up to September 2023, were included. Finally, 16 studies (ten in vitro and eight in vivo) are reviewed. Apoptosis (in 13 studies), inflammatory (in four studies), and oxidative stress (in 14 studies) pathways are the most commonly reported pathways involved in colistin-induced neurotoxicity. The assessed compounds include non-herbal (e.g., ascorbic acid, rapamycin, and minocycline) and herbal (e.g., curcumin, rutin, baicalein, salidroside, and ginsenoside) agents. Besides these compounds, some other measures like transplantation of mitochondria and the use of nerve growth factor and mesenchymal stem cells could be motivating subjects for future research. Based on the data from experimental (in vitro and animal) studies, a combination of colistin with neuroprotective agents could prevent or decrease colistin-induced neurotoxicity. However, well-designed randomized clinical trials and human studies are essential for demonstrating efficacy.
Collapse
Affiliation(s)
- Setareh Soroudi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box, Mashhad, 91775-1365, Iran
| | - Ghazal Mousavi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box, Mashhad, 91775-1365, Iran
| | - Fatemeh Jafari
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box, Mashhad, 91775-1365, Iran
| | - Sepideh Elyasi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, P.O. Box, Mashhad, 91775-1365, Iran.
| |
Collapse
|
3
|
Wei M, Wu T, Chen N. Bridging neurotrophic factors and bioactive peptides to Alzheimer's disease. Ageing Res Rev 2024; 94:102177. [PMID: 38142891 DOI: 10.1016/j.arr.2023.102177] [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: 10/26/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder. As the demographic shifting towards an aging population, AD has emerged as a prominent public health concern. The pathogenesis of AD is complex, and there are no effective treatment methods for AD until now. In recent years, neurotrophic factors and bioactive peptides including brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), irisin, melatonin, have been discovered to exert neuroprotective functions for AD. Bioactive peptides can be divided into two categories based on their sources: endogenous and exogenous. This review briefly elaborates on the pathogenesis of AD and analyzes the regulatory effects of endogenous and exogenous peptides on the pathogenesis of AD, thereby providing new therapeutic targets for AD and a theoretical basis for the application of bioactive peptides as adjunctive therapies for AD.
Collapse
Affiliation(s)
- Minhui Wei
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Tong Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
4
|
Tonev D, Momchilova A. Oxidative Stress and the Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) Pathway in Multiple Sclerosis: Focus on Certain Exogenous and Endogenous Nrf2 Activators and Therapeutic Plasma Exchange Modulation. Int J Mol Sci 2023; 24:17223. [PMID: 38139050 PMCID: PMC10743556 DOI: 10.3390/ijms242417223] [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: 10/26/2023] [Revised: 11/18/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
The pathogenesis of multiple sclerosis (MS) suggests that, in genetically susceptible subjects, T lymphocytes undergo activation in the peripheral compartment, pass through the BBB, and cause damage in the CNS. They produce pro-inflammatory cytokines; induce cytotoxic activities in microglia and astrocytes with the accumulation of reactive oxygen species, reactive nitrogen species, and other highly reactive radicals; activate B cells and macrophages and stimulate the complement system. Inflammation and neurodegeneration are involved from the very beginning of the disease. They can both be affected by oxidative stress (OS) with different emphases depending on the time course of MS. Thus, OS initiates and supports inflammatory processes in the active phase, while in the chronic phase it supports neurodegenerative processes. A still unresolved issue in overcoming OS-induced lesions in MS is the insufficient endogenous activation of the Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) pathway, which under normal conditions plays an essential role in mitochondria protection, OS, neuroinflammation, and degeneration. Thus, the search for approaches aiming to elevate endogenous Nrf2 activation is capable of protecting the brain against oxidative damage. However, exogenous Nrf2 activators themselves are not without drawbacks, necessitating the search for new non-pharmacological therapeutic approaches to modulate OS. The purpose of the present review is to provide some relevant preclinical and clinical examples, focusing on certain exogenous and endogenous Nrf2 activators and the modulation of therapeutic plasma exchange (TPE). The increased plasma levels of nerve growth factor (NGF) in response to TPE treatment of MS patients suggest their antioxidant potential for endogenous Nrf2 enhancement via NGF/TrkA/PI3K/Akt and NGF/p75NTR/ceramide-PKCζ/CK2 signaling pathways.
Collapse
Affiliation(s)
- Dimitar Tonev
- Department of Anesthesiology and Intensive Care, University Hospital “Tzaritza Yoanna—ISUL”, Medical University of Sofia, 1527 Sofia, Bulgaria
| | - Albena Momchilova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Science, 1113 Sofia, Bulgaria;
| |
Collapse
|
5
|
Single and repeated bisphenol A treatment induces ROS, Aβ and hyperphosphorylated-tau accumulation, and insulin pathways disruption, through HDAC2 and PTP1B overexpression, leading to SN56 cholinergic apoptotic cell death. Food Chem Toxicol 2022; 170:113500. [DOI: 10.1016/j.fct.2022.113500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
|
6
|
Maeda Y, Takeda M, Mitsuhara T, Okazaki T, Shimizu K, Kuwabara M, Hosogai M, Yuge L, Horie N. Longitudinal electrophysiological changes after mesenchymal stem cell transplantation in a spinal cord injury rat model. PLoS One 2022; 17:e0272526. [PMID: 35930554 PMCID: PMC9355172 DOI: 10.1371/journal.pone.0272526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/21/2022] [Indexed: 11/19/2022] Open
Abstract
Transcranial electrically stimulated motor-evoked potentials (tcMEPs) are widely used to evaluate motor function in humans and animals. However, the relationship between tcMEPs and the recovery of paralysis remains unclear. We previously reported that transplantation of mesenchymal stem cells to a spinal cord injury (SCI) rat model resulted in various degrees of recovery from paraplegia. As a continuation of this work, in the present study, we aimed to establish the longitudinal electrophysiological changes in this SCI rat model after mesenchymal stem cell transplantation. SCI rats were established using the weight-drop method. The model rats were transvenously transplanted with two types of mesenchymal stem cells (MSCs), one derived from rat cranial bones and the other from the bone marrow of the femur and tibia bone, 24 h after SCI. A phosphate-buffered saline (PBS) group that received only PBS was also created for comparison. The degree of paralysis was evaluated over 28 days using the Basso–Beattie–Bresnahan (BBB) scale and inclined plane task score. Extended tcMEPs were recorded using a previously reported bone-thinning technique, and the longitudinal electrophysiological changes in tcMEPs were investigated. In addition, the relationship between the time course of recovery from paralysis and reappearance of tcMEPs was revealed. The appearance of the tcMEP waveform was earlier in MSC-transplanted rats than in PBS-administered rats (earliest date was 7 days after SCI). The MEP waveforms also appeared at approximately the same level on the BBB scale (average score, 11 points). Ultimately, this study can help enhance our understanding of the relationship between neural regeneration and tcMEP recording. Further application of tcMEP in regenerative medicine research is expected.
Collapse
Affiliation(s)
- Yuyo Maeda
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail:
| | - Masaaki Takeda
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takafumi Mitsuhara
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takahito Okazaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kiyoharu Shimizu
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masashi Kuwabara
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masahiro Hosogai
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Louis Yuge
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobutaka Horie
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
7
|
Liu H, Li Q, Zhang X, Shi Y, Li J. Effect of ginkgolide K on calcium channel activity in Alzheimer's disease. Exp Ther Med 2022; 23:426. [PMID: 35607377 PMCID: PMC9121205 DOI: 10.3892/etm.2022.11353] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/12/2022] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative dementia with the key pathological hallmark of amyloid deposits that may induce mitochondrial dysfunction. Ginkgolide K (GK) has been proven to have neuroprotective effects. The present study sought to explore the neuroprotective effect of GK through regulation of the expression of mitochondrial Ca2+ uniporter (MCU) in the pathology of AD. SH-SY5Y cells were cultured and the expression of MCU was enhanced by transfection of MCU recombinant vectors or knockdown by MCU small interfering RNA. The cells were treated with GK and amyloid β (Aβ). Thereafter, the effects of GK, MCU expression and Aβ on viability and apoptosis of SH-SY5Y cells were examined via a WST-1 assay, flow cytometry and Caspase-3/8 activity assays, respectively. The effects of GK, MCU expression and Aβ on the calcium levels in mitochondria were also examined. The regulatory effect of GK on MCU expression was examined by reverse transcription-quantitative PCR and western blot analysis. Furthermore, APP/PS1 mice received supplementation with GK and their cognitive ability was then examined through water maze tests, while the expression of MCU was examined using immunohistochemistry. The results indicated that enhancing the expression of MCU inhibited cell viability and promoted apoptosis. GK protected cells from amyloid-induced cytotoxicity by promoting cell viability and preventing cell apoptosis. The neuroprotective effect of GK was abolished when MCU expression was knocked down. GK decreased the expression of MCU in vitro and downregulation of MCU decreased the calcium level in mitochondria. Treatment with GK in APP/PS1 mice downregulated the expression of MCU in the brains and alleviated cognitive impairment. In conclusion, the present study demonstrated that the administration of GK protected neurons by preventing apoptosis. Furthermore, the neuroprotective effect of GK in neuronal cells was indicated to be related to the inhibition of MCU expression. Therefore, administration of GK may be a promising strategy for treating AD.
Collapse
Affiliation(s)
- Hongbin Liu
- No. 2 Department of Geriatrics, Beijing Geriatric Hospital, Beijing 100095, P.R. China
| | - Qinyun Li
- No. 2 Department of Geriatrics, Beijing Geriatric Hospital, Beijing 100095, P.R. China
| | - Xiaodan Zhang
- No. 2 Department of Geriatrics, Beijing Geriatric Hospital, Beijing 100095, P.R. China
| | - Yun Shi
- No. 2 Department of Geriatrics, Beijing Geriatric Hospital, Beijing 100095, P.R. China
| | - Jinyi Li
- Dolu Health Consultant Co., Ltd., Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
8
|
Gong Y, Jiang X, Yang S, Huang Y, Hong J, Ma Y, Fang X, Fang Y, Wu J. The Biological Activity of 3-O-Acetyl-11-keto-β-Boswellic Acid in Nervous System Diseases. Neuromolecular Med 2022; 24:374-384. [PMID: 35303275 PMCID: PMC8931781 DOI: 10.1007/s12017-022-08707-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/02/2022] [Indexed: 12/29/2022]
Abstract
Frankincense is a hard gelatinous resin exuded by Boswellia serrata. It contains a complex array of components, of which acetyl-11-keto-beta-boswellic acid (AKBA), a pentacyclic triterpenoid of the resin class, is the main active component. AKBA has a variety of physiological actions, including anti-infection, anti-tumor, and antioxidant effects. The use of AKBA for the treatment of mental diseases has been documented as early as ancient Greece. Recent studies have found that AKBA has anti-aging and other neurological effects, suggesting its potential for the treatment of neurological diseases. This review focuses on nervous system-related diseases, summarizes the functions and mechanisms of AKBA in promoting nerve repair and regeneration after injury, protecting against ischemic brain injury and aging, inhibiting neuroinflammation, ameliorating memory deficits, and alleviating neurotoxicity, as well as having anti-glioma effects and relieving brain edema. The mechanisms by which AKBA functions in different diseases and the relationships between dosage and biological effects are discussed in depth with the aim of increasing understanding of AKBA and guiding its use for the treatment of nervous system diseases.
Collapse
Affiliation(s)
- Yuqing Gong
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Xinyi Jiang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Suibi Yang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Yue Huang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Jinhui Hong
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Yanxiu Ma
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Xin Fang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Yong Fang
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, 150081, China.
| | - Jing Wu
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China.
| |
Collapse
|
9
|
Gohil K, Kazmi MZH, Williams FJ. Structure-activity relationship and bioactivity studies of neurotrophic trans-banglene. Org Biomol Chem 2022; 20:2187-2193. [PMID: 35229853 DOI: 10.1039/d2ob00016d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The synthesis and bioactivity of neurotrophic banglenes and derivatives is described, establishing a structure-activity relationship which enables future mechanistic studies. Neuritogenesis assays indicate that (-) trans-banglene is the active enantiomer. Assays performed with and without NGF protein suggest that neurotrophic activity and potentiation of NGF activity by (-) trans-banglene might be distinct unassociated processes. Interestingly, (-) trans-banglene potentiation of NGF-induced neuritogenesis is unaffected by the presence of Erk1/2, Akt and Pkc inhibitors.
Collapse
Affiliation(s)
- Khyati Gohil
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - M Zain H Kazmi
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | | |
Collapse
|
10
|
Song Y, Du Z, Chen X, Zhang W, Zhang G, Li H, Chang L, Wu Y. Astrocytic N-Methyl-D-Aspartate Receptors Protect the Hippocampal Neurons Against Amyloid-β142-Induced Synaptotoxicity by Regulating Nerve Growth Factor. J Alzheimers Dis 2021; 85:167-178. [PMID: 34776441 DOI: 10.3233/jad-210730] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Soluble oligomeric amyloid-β (Aβ)-induced synaptic dysfunction is an early event in Alzheimer's disease (AD) pathogenesis. Mounting evidence has suggested N-methyl-D-aspartate receptors (NMDARs) play an important role in Aβ-induced synaptotoxicity. Originally NMDARs were believed to be expressed exclusively in neurons; however, recent two decades studies have demonstrated functional NMDARs present on astrocytes. Neuronal NMDARs are modulators of neurodegeneration, while our previous initial study found that astrocytic NMDARs mediated synaptoprotection and identified nerve growth factor (NGF) secreted by astrocytes, as a likely mediator, but how astrocytic NMDARs protect neurons against Aβ-induced synaptotoxicity through regulating NGF remains unclear. OBJECTIVE To achieve further insight into the mechanism of astrocytic NMDARs oppose Aβ-induced synaptotoxicity through regulating NGF. METHODS With the primary hippocampal neuronal and astrocytic co-cultures, astrocytes were pretreated with agonist or antagonist of NMDARs before Aβ142 oligomers application to neuron-astrocyte co-cultures. Western blot, RT-PCR, etc., were used for the related proteins evaluation. RESULTS Activation of astrocytic NMDARs can significantly mitigate Aβ142-induced loss of PSD-95 and synaptophysin through increasing NGF release. Blockade of astrocytic NMDARs inhibited Aβ-induced compensatory protective NGF increase in protein and mRNA levels through modulating NF-κB of astrocytes. Astrocytic NMDARs activation can enhance Aβ-induced Furin increase, and blockade of astrocytic NMDARs inhibited Aβ-induced immunofluorescent intensity elevation of vesicle trafficking protein VAMP3 and NGF double-staining. CONCLUSION Astrocytic NMDARs oppose Aβ-induced synaptotoxicity through modulating the synthesis, maturation, and secretion of NGF in astrocytes. This new information may contribute to the quest for specific targeted strategy of intervention to delay the onset of AD.
Collapse
Affiliation(s)
- Yizhi Song
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Zunshu Du
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xinyue Chen
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Wanning Zhang
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Guitao Zhang
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Hui Li
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Lirong Chang
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Anatomy, School of Basic MedicalSciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Transplantation of rat cranial bone-derived mesenchymal stem cells promotes functional recovery in rats with spinal cord injury. Sci Rep 2021; 11:21907. [PMID: 34754046 PMCID: PMC8578570 DOI: 10.1038/s41598-021-01490-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Cell-based therapy using mesenchymal stem cells (MSCs) is a novel treatment strategy for spinal cord injury (SCI). MSCs can be isolated from various tissues, and their characteristics vary based on the source. However, reports demonstrating the effect of transplanted rat cranial bone-derived MSCs (rcMSCs) on rat SCI models are lacking. In this study, we determined the effect of transplanting rcMSCs in rat SCI models. MSCs were established from collected bone marrow and cranial bones. SCI rats were established using the weight-drop method and transplanted intravenously with MSCs at 24 h post SCI. The recovery of motor function and hindlimb electrophysiology was evaluated 4 weeks post transplantation. Electrophysiological recovery was evaluated by recording the transcranial electrical stimulation motor-evoked potentials. Tissue repair after SCI was assessed by calculating the cavity ratio. The expression of genes involved in the inflammatory response and cell death in the spinal cord tissue was assessed by real-time polymerase chain reaction. The transplantation of rcMSCs improved motor function and electrophysiology recovery, and reduced cavity ratio. The expression of proinflammatory cytokines was suppressed in the spinal cord tissues of the rats that received rcMSCs. These results demonstrate the efficacy of rcMSCs as cell-based therapy for SCI.
Collapse
|
12
|
The Role of HO-1 and Its Crosstalk with Oxidative Stress in Cancer Cell Survival. Cells 2021; 10:cells10092401. [PMID: 34572050 PMCID: PMC8471703 DOI: 10.3390/cells10092401] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenases (HOs) act on heme degradation to produce carbon monoxide (CO), free iron, ferritin, and biliverdin. Upregulation of cellular HO-1 levels is signature of oxidative stress for its downstream effects particularly under pro-oxidative status. Subcellular traffics of HO-1 to different organelles constitute a network of interactions compromising a variety of effectors such as pro-oxidants, ROS, mitochondrial enzymes, and nucleic transcription factors. Some of the compartmentalized HO-1 have been demonstrated as functioning in the progression of cancer. Emerging data show the multiple roles of HO-1 in tumorigenesis from pathogenesis to the progression to malignancy, metastasis, and even resistance to therapy. However, the role of HO-1 in tumorigenesis has not been systematically addressed. This review describes the crosstalk between HO-1 and oxidative stress, and following redox regulation in the tumorigenesis. HO-1-regulated signaling pathways are also summarized. This review aims to integrate basic information and current progress of HO-1 in cancer research in order to enhance the understandings and facilitate following studies.
Collapse
|
13
|
Tzeng HE, Lin SL, Thadevoos LA, Ko CY, Liu JF, Huang YW, Lin CY, Fong YC, Tang CH. The mir-423-5p/MMP-2 Axis Regulates the Nerve Growth Factor-Induced Promotion of Chondrosarcoma Metastasis. Cancers (Basel) 2021; 13:cancers13133347. [PMID: 34283074 PMCID: PMC8268073 DOI: 10.3390/cancers13133347] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary A chondrosarcoma is a common tumor of the bone that has a high propensity to metastasize to distant organs. The effects of NGF in a chondrosarcoma are not confirmed although NGF is capable of promoting the progression and metastasis of several different types of tumors. Here, we found that NGF promotes the chondrosarcoma migration and metastasis in vitro and in vivo. The levels of NGF and MMP-2 in human chondrosarcoma tumor tissues correlated strongly with the tumor stage. We identified that NGF induces the MMP-2 synthesis and chondrosarcoma cell motility by inhibiting miR-423-5p expression through the FAK and c-Src pathways. We suggest that NGF is a worthwhile therapeutic target in the treatment of a metastatic chondrosarcoma. Abstract A chondrosarcoma is a common tumor of the soft tissue and bone that has a high propensity to metastasize to distant organs. Nerve growth factor (NGF) is capable of promoting the progression and metastasis of several different types of tumors although the effects of NGF in a chondrosarcoma are not confirmed. Here, we found that the levels of NGF and matrix metalloproteinase-2 (MMP-2) correlated with the tumor stage in patients with a chondrosarcoma. NGF facilitated the MMP-2-dependent cellular migration in human chondrosarcoma JJ012 cells while the overexpression of NGF enhanced the lung metastasis in a mouse model of a chondrosarcoma. NGF promoted the MMP-2 synthesis and cell migration by inhibiting miR-423-5p expression through the FAK and c-Src signaling cascades. NGF appears to be a worthwhile therapeutic target in the treatment of a metastatic chondrosarcoma.
Collapse
Affiliation(s)
- Huey-En Tzeng
- Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Division of Hematology/Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Syuan-Ling Lin
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40402, Taiwan;
| | - Louis-Anoop Thadevoos
- International Master Program of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan;
| | - Chih-Yuan Ko
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 40402, Taiwan;
| | - Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yu-Wen Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan;
| | - Chih-Yang Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan;
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung 40402, Taiwan
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin 65152, Taiwan
- Correspondence: (Y.-C.F.); (C.-H.T.)
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan;
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan;
- Department of Biotechnology, College of Health Science, Asia University, Taichung 41354, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Correspondence: (Y.-C.F.); (C.-H.T.)
| |
Collapse
|
14
|
Ma WH, Chen AF, Xie XY, Huang YS. Sigma ligands as potent inhibitors of Aβ and AβOs in neurons and promising therapeutic agents of Alzheimer's disease. Neuropharmacology 2021; 190:108342. [PMID: 33045243 DOI: 10.1016/j.neuropharm.2020.108342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/14/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease and characterized by dementia, memory decline, loss of learning and cognitive disorder. The main pathological features of AD are the deposition of amyloid plaques and the formation of neurofibrillary tangles (NFTs) in the brain. The current anti-AD drugs have shown unsatisfactory therapeutic results. Due to the complications and unclear pathogenesis, AD is still irreversible and incurable. Among several hypotheses proposed by the academic community, the amyloid cascade is widely recognized by scholars and supported by a large amount of evidences. However, controversy over pathogenic factors has also been ongoing. Increasing evidence has shown that amyloid-β (Aβ) and especially amyloid-β oligomers (AβOs) are highly neurotoxic and pathogenic agents that damage neurons, mediate various receptors in the downstream pathways, and ultimately lead to learning and cognitive dysfunction. However, efforts in developing inhibitors of Aβ or amyloid-β precursor protein (APP) have all failed to yield good clinical results. More recently, it has been demonstrated that sigma receptors, including sigma-1 and sigma-2 subtypes, may play critical roles in the regulation of binding and metabolism of AβOs in neuron cells and the pathophysiology of AD. Thus, sigma receptor ligands are being recognized as promising therapeutic agents for treating or ameliorating AD. This article will review the pathophysiology of AD and highlight the sigma ligands that display the capability of preventing or even reversing Aβ- and AβOs-induced neurotoxicity and blocking the signal transduction caused by AβOs.
Collapse
Affiliation(s)
- Wen-Hui Ma
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China; Dongguan Key Laboratory for Drug Design & Formulation, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Ai-Fang Chen
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China; Dongguan Key Laboratory for Drug Design & Formulation, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Xiao-Yang Xie
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China; Dongguan Key Laboratory for Drug Design & Formulation, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Yun-Sheng Huang
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China; Dongguan Key Laboratory for Drug Design & Formulation, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China.
| |
Collapse
|
15
|
Colardo M, Martella N, Pensabene D, Siteni S, Di Bartolomeo S, Pallottini V, Segatto M. Neurotrophins as Key Regulators of Cell Metabolism: Implications for Cholesterol Homeostasis. Int J Mol Sci 2021; 22:5692. [PMID: 34073639 PMCID: PMC8198482 DOI: 10.3390/ijms22115692] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
Neurotrophins constitute a family of growth factors initially characterized as predominant mediators of nervous system development, neuronal survival, regeneration and plasticity. Their biological activity is promoted by the binding of two different types of receptors, leading to the generation of multiple and variegated signaling cascades in the target cells. Increasing evidence indicates that neurotrophins are also emerging as crucial regulators of metabolic processes in both neuronal and non-neuronal cells. In this context, it has been reported that neurotrophins affect redox balance, autophagy, glucose homeostasis and energy expenditure. Additionally, the trophic support provided by these secreted factors may involve the regulation of cholesterol metabolism. In this review, we examine the neurotrophins' signaling pathways and their effects on metabolism by critically discussing the most up-to-date information. In particular, we gather experimental evidence demonstrating the impact of these growth factors on cholesterol metabolism.
Collapse
Affiliation(s)
- Mayra Colardo
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Noemi Martella
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Daniele Pensabene
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Silvia Siteni
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Sabrina Di Bartolomeo
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Valentina Pallottini
- Department of Science, University Roma Tre, Viale Marconi 446, 00146 Rome, Italy;
- Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Via del Fosso Fiorano 64, 00143 Rome, Italy
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| |
Collapse
|
16
|
Nerve Growth Factor Peptides Bind Copper(II) with High Affinity: A Thermodynamic Approach to Unveil Overlooked Neurotrophin Roles. Int J Mol Sci 2021; 22:ijms22105085. [PMID: 34064906 PMCID: PMC8150721 DOI: 10.3390/ijms22105085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
Nerve growth factor (NGF) is a protein essential to neurons survival, which interacts with its receptor as a non-covalent dimer. Peptides belonging to NGF N-terminal domain are able to mimic the activity of the whole protein. Such activity is affected by the presence of copper ions. The metal is released in the synaptic cleft where proteins, not yet identified, may bind and transfer to human copper transporter 1 (hCtr1), for copper uptake in neurons. The measurements of the stability constants of copper complexes formed by amyloid beta and hCtr1 peptide fragments suggest that beta-amyloid (Aβ) can perform this task. In this work, the stability constant values of copper complex species formed with the dimeric form of N-terminal domain, sequence 1–15 of the protein, were determined by means of potentiometric measurements. At physiological pH, NGF peptides bind one equivalent of copper ion with higher affinity of Aβ and lower than hCtr1 peptide fragments. Therefore, in the synaptic cleft, NGF may act as a potential copper chelating molecule, ionophore or chaperone for hCtr1 for metal uptake. Copper dyshomeostasis and mild acidic environment may modify the balance between metal, NGF, and Aβ, with consequences on the metal cellular uptake and therefore be among causes of the Alzheimer’s disease onset.
Collapse
|
17
|
Liu BW, Zhang J, Hong YS, Li NB, Liu Y, Zhang M, Wu WY, Zheng H, Lampert A, Zhang XW. NGF-Induced Nav1.7 Upregulation Contributes to Chronic Post-surgical Pain by Activating SGK1-Dependent Nedd4-2 Phosphorylation. Mol Neurobiol 2021; 58:964-982. [PMID: 33063281 DOI: 10.1007/s12035-020-02156-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/29/2020] [Indexed: 01/07/2023]
Abstract
At present, chronic post-surgical pain (CPSP) is difficult to prevent and cure clinically because of our lack of understanding of its mechanisms. Surgical injury induces the upregulation of voltage-gated sodium channel Nav1.7 in dorsal root ganglion (DRG) neurons, suggesting that Nav1.7 is involved in the development of CPSP. However, the mechanism leading to persistent dysregulation of Nav1.7 is largely unknown. Given that nerve growth factor (NGF) induces a long-term increase in the neuronal hyperexcitability after injury, we hypothesized that NGF might cause the long-term dysregulation of Nav1.7. In this study, we aimed to investigate whether Nav1.7 regulation by NGF is involved in CPSP and thus contributes to the specific mechanisms involved in the development of CPSP. Using conditional nociceptor-specific Nav1.7 knockout mice, we confirmed the involvement of Nav1.7 in NGF-induced pain and identified its role in the maintenance of pain behavior during long-term observations (up to 14 days). Using western blot analyses and immunostaining, we showed that NGF could trigger the upregulation of Nav1.7 expression and thus support the development of CPSP in rats. Using pharmacological approaches, we showed that the increase of Nav1.7 might be partly regulated by an NGF/TrkA-SGK1-Nedd4-2-mediated pathway. Furthermore, reversing the upregulation of Nav1.7 in DRG could alleviate spinal sensitization. Our results suggest that the maintained upregulation of Nav1.7 triggered by NGF contributes to the development of CPSP. Attenuating the dysregulation of Nav1.7 in peripheral nociceptors may be a strategy to prevent the transition from acute post-surgical pain to CPSP.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Behavior, Animal/drug effects
- Benzamides/pharmacology
- Brain-Derived Neurotrophic Factor/metabolism
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Hydrazines/pharmacology
- Immediate-Early Proteins/antagonists & inhibitors
- Immediate-Early Proteins/metabolism
- Indoles/pharmacology
- Male
- Mice, Knockout
- Models, Biological
- NAV1.7 Voltage-Gated Sodium Channel/genetics
- NAV1.7 Voltage-Gated Sodium Channel/metabolism
- Nedd4 Ubiquitin Protein Ligases/metabolism
- Nerve Growth Factor/pharmacology
- Pain, Postoperative/genetics
- Pain, Postoperative/pathology
- Phosphorylation/drug effects
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Rats, Sprague-Dawley
- Receptor, trkA/antagonists & inhibitors
- Receptor, trkA/metabolism
- Spinal Cord/pathology
- Ubiquitination/drug effects
- Up-Regulation/drug effects
- Vesicular Glutamate Transport Protein 2/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Bao-Wen Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jin Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi-Shun Hong
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ning-Bo Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mi Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Yao Wu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hua Zheng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Angelika Lampert
- Institute of Physiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Xian-Wei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
18
|
Dai C, Xiong J, Wang Y, Shen J, Velkov T, Xiao X. Nerve Growth Factor Confers Neuroprotection against Colistin-Induced Peripheral Neurotoxicity. ACS Infect Dis 2020; 6:1451-1459. [PMID: 32422040 DOI: 10.1021/acsinfecdis.0c00107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neurotoxicity is an unwanted side effect for patients when receiving parenteral colistin therapy. The development of effective neuroprotective agents that can be coadministered during colistin therapy remains a priority area in antimicrobial chemotherapy. The present study aimed to investigate the protective effect of nerve growth factor (NGF) against colistin-induced peripheral neurotoxicity using a murine model. C57BL/6 mice were randomly divided into the following 6 groups: (i) untreated control, (ii) NGF alone (36 μg/kg/day administered intraperitoneally), (iii) colistin alone (18 mg/kg/day administered intraperitoneally), and (iv-vi) colistin (18 mg/kg/day) plus NGF (9, 18, and 36 μg/kg/day). After treatment for 7 days, neurobehavioral and electrophysiology changes, histopathological assessments of sciatic nerve damage, and oxidative stress biomarkers were examined. The mRNA expression levels of Nrf2, HO-1, Akt, Bax, and caspase-3 and -9 were assessed using quantitative RT-PCR. The results showed that, across all the groups wherein NGF was coadministered with colistin, a marked attenuation of colistin-induced sciatic nerve damage and improved sensory and motor function were observed. In comparison to the colistin only treatment group, animals that received NGF displayed upregulated Nrf2 and HO-1 mRNA expression levels and downregulated Bax and caspase-3 and -9 mRNA expression levels. In summary, our study reveals that NGF coadministration protects against colistin-induced peripheral neurotoxicity via the activation of Akt and Nrf2/HO-1 pathways and inhibition of oxidative stress. This study highlights the potential clinical application of NGF as a neuroprotective agent for coadministration during colistin therapy.
Collapse
Affiliation(s)
- Chongshan Dai
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Jianli Xiong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P. R. China
| | - Yang Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Jianzhong Shen
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xilong Xiao
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
19
|
Su R, Su W, Jiao Q. Corrigendum to: NGF protects neuroblastoma cells against β-amyloid induced apoptosis via the Nrf2/HO-1 pathway. FEBS Open Bio 2020; 10:288. [PMID: 32025477 PMCID: PMC6996377 DOI: 10.1002/2211-5463.12782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Rui Su
- Department of Neurosurgery, Luoyang Central Hospital Affiliated to Zhengzhou University, China
| | - Wei Su
- Department of Intensive Care Unit, Sir Run Run Shaw Hospital Affiliated by Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Jiao
- Department of Anesthesia Surgery, Sanmenxia Central Hospital, China
| |
Collapse
|