1
|
Huang S, Zhang Y, Shu H, Liu W, Zhou X, Zhou X. Advances of the MAPK pathway in the treatment of spinal cord injury. CNS Neurosci Ther 2024; 30:e14807. [PMID: 38887853 PMCID: PMC11183187 DOI: 10.1111/cns.14807] [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: 02/05/2024] [Revised: 04/23/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Spinal cord injury (SCI) represents a complex pathology within the central nervous system (CNS), leading to severe sensory and motor impairments. It activates various signaling pathways, notably the mitogen-activated protein kinase (MAPK) pathway. Present treatment approaches primarily focus on symptomatic relief, lacking efficacy in addressing the underlying pathophysiological mechanisms. Emerging research underscores the significance of the MAPK pathway in neuronal differentiation, growth, survival, axonal regeneration, and inflammatory responses post-SCI. Modulating this pathway post-injury has shown promise in attenuating inflammation, minimizing apoptosis, alleviating neuropathic pain, and fostering neural regeneration. Given its pivotal role, the MAPK pathway emerges as a potential therapeutic target in SCI management. This review synthesizes current knowledge on SCI pathology, delineates the MAPK pathway's characteristics, and explores its dual roles in SCI pathology and therapeutic interventions. Furthermore, it addresses the existing challenges in MAPK research in the context of SCI, proposing solutions to overcome these hurdles. Our aim is to offer a comprehensive reference for future research on the MAPK pathway and SCI, laying the groundwork for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Shixue Huang
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Yinuo Zhang
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Haoming Shu
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Wei Liu
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Xin Zhou
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
- Translational Research Centre of Orthopedics, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Wang W, Gao R, Yan X, Shu W, Zhang X, Zhang W, Zhang L. Relationship between plasma brain-derived neurotrophic factor levels and neurological disorders: An investigation using Mendelian randomisation. Heliyon 2024; 10:e30415. [PMID: 38707431 PMCID: PMC11068855 DOI: 10.1016/j.heliyon.2024.e30415] [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: 10/05/2023] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024] Open
Abstract
Background Altered brain-derived neurotrophic factor (BDNF) concentrations have been detected in the central nervous system tissues and peripheral blood. These alterations are associated with a series of neurological disorders. Objective To investigate the potential causal relationships between genetically determined plasma BDNF levels and various neurological diseases using a two-sample Mendelian randomisation study. Methods We selected single nucleotide polymorphisms strongly related to plasma BDNF levels as instrumental variables. Within the Mendelian randomisation framework, we used summary-level statistics for exposure (plasma BDNF levels) and outcomes (neurological disorders). Results We observed suggestive evidence of a relation between higher plasma BDNF levels and less risk of nontraumatic intracranial haemorrhage (nITH) (odds ratio [OR] = 0.861, 95 % confidence interval [CI]: 0.774-0.958, P = 0.006, PFDR = 0.078), epilepsy (OR = 0.927, 95 % CI: 0.880-0.976, P = 0.004, PFDR = 0.078), focal epilepsy (OR = 0.928, 95 % CI: 0.874-0.986, P = 0.016, PFDR = 0.139), and non-lesional focal epilepsy (OR = 0.981, 95 % CI: 0.964-0.999, P = 0.041, PFDR = 0.267). Combined with the UK Biobank dataset, the association of plasma BDNF levels with nITH remained significant (OR = 0.88, 95 % CI: 0.81-0.96, P < 0.01). The combined analysis of three consortium datasets demonstrated a considerable impact of plasma BDNF on epilepsy (OR = 0.94, 95 % CI: 0.90-0.98, P < 0.01) and a suggestive impact on focal epilepsy (OR = 0.94, 95 % CI: 0.89-0.99, P = 0.02). However, there was no apparent correlation between plasma BDNF levels and other neurological disorders or related subtypes. Conclusions Our study supports a possible causal relationship between elevated plasma BDNF levels and a reduced risk of nITH, epilepsy, and focal epilepsy.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Runshi Gao
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoming Yan
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Shu
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xi Zhang
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenjie Zhang
- Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Nordengen K, Cappelletti C, Bahrami S, Frei O, Pihlstrøm L, Henriksen SP, Geut H, Rozemuller AJM, van de Berg WDJ, Andreassen OA, Toft M. Pleiotropy with sex-specific traits reveals genetic aspects of sex differences in Parkinson's disease. Brain 2024; 147:858-870. [PMID: 37671566 PMCID: PMC10907091 DOI: 10.1093/brain/awad297] [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: 03/01/2023] [Revised: 08/01/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
Parkinson's disease is an age-related neurodegenerative disorder with a higher incidence in males than females. The causes for this sex difference are unknown. Genome-wide association studies (GWAS) have identified 90 Parkinson's disease risk loci, but the genetic studies have not found sex-specific differences in allele frequency on autosomal chromosomes or sex chromosomes. Genetic variants, however, could exert sex-specific effects on gene function and regulation of gene expression. To identify genetic loci that might have sex-specific effects, we studied pleiotropy between Parkinson's disease and sex-specific traits. Summary statistics from GWASs were acquired from large-scale consortia for Parkinson's disease (n cases = 13 708; n controls = 95 282), age at menarche (n = 368 888 females) and age at menopause (n = 69 360 females). We applied the conditional/conjunctional false discovery rate (FDR) method to identify shared loci between Parkinson's disease and these sex-specific traits. Next, we investigated sex-specific gene expression differences in the superior frontal cortex of both neuropathologically healthy individuals and Parkinson's disease patients (n cases = 61; n controls = 23). To provide biological insights to the genetic pleiotropy, we performed sex-specific expression quantitative trait locus (eQTL) analysis and sex-specific age-related differential expression analysis for genes mapped to Parkinson's disease risk loci. Through conditional/conjunctional FDR analysis we found 11 loci shared between Parkinson's disease and the sex-specific traits age at menarche and age at menopause. Gene-set and pathway analysis of the genes mapped to these loci highlighted the importance of the immune response in determining an increased disease incidence in the male population. Moreover, we highlighted a total of nine genes whose expression or age-related expression in the human brain is influenced by genetic variants in a sex-specific manner. With these analyses we demonstrated that the lack of clear sex-specific differences in allele frequencies for Parkinson's disease loci does not exclude a genetic contribution to differences in disease incidence. Moreover, further studies are needed to elucidate the role that the candidate genes identified here could have in determining a higher incidence of Parkinson's disease in the male population.
Collapse
Affiliation(s)
- Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Chiara Cappelletti
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Faculty of Technology, Art and Design, OsloMet—Oslo Metropolitan University, 0130 Oslo, Norway
- Department of Research, Innovation and Education, Oslo University Hospital, 0424 Oslo, Norway
| | - Shahram Bahrami
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Oleksandr Frei
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | | | - Hanneke Geut
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Ole A Andreassen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
4
|
Nguyen-Thi PT, Vo TK, Pham THT, Nguyen TT, Van Vo G. Natural flavonoids as potential therapeutics in the management of Alzheimer's disease: a review. 3 Biotech 2024; 14:68. [PMID: 38357675 PMCID: PMC10861420 DOI: 10.1007/s13205-024-03925-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/05/2024] [Indexed: 02/16/2024] Open
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disorder which is associated with the accumulation of proteotoxic Aβ peptides, and pathologically characterized by the deposition of Aβ-enriched plaques and neurofibrillary tangles. Given the social and economic burden caused by the rising frequency of AD, there is an urgent need for the development of appropriate therapeutics. Natural compounds are gaining popularity as alternatives to synthetic drugs due to their neuroprotective properties and higher biocompatibility. While natural compound's therapeutic effects for AD have been recently investigated in numerous in vitro and in vivo studies, only few have developed to clinical trials. The present review aims to provide a brief overview of the therapeutic effects, new insights, and upcoming perspectives of the preclinical and clinical trials of flavonoids for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
| | - Tuong Kha Vo
- Department of Sports Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, 100000 Vietnam
| | - Thi Hong Trang Pham
- Institute for Global Health Innovations, Duy Tan University, Da Nang, 550000 Vietnam
- Faculty of Pharmacy, Duy Tan University, Da Nang, 550000 Vietnam
| | - Thuy Trang Nguyen
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 71420 Vietnam
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University – Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000 Vietnam
- Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University, Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 70000 Vietnam
- Vietnam National University – Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000 Vietnam
| |
Collapse
|
5
|
Edvinsson L, Krause DN. Switching Off Vascular MAPK Signaling: A Novel Strategy to Prevent Delayed Cerebral Ischemia Following Subarachnoid Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01234-z. [PMID: 38334872 DOI: 10.1007/s12975-024-01234-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Patients who initially survive the rupture and repair of a brain aneurysm often take a devastating turn for the worse some days later and die or suffer permanent neurologic deficits. This catastrophic sequela is attributed to a delayed phase of global cerebral ischemia (DCI) following aneurysmal subarachnoid hemorrhage (aSAH), but we lack effective treatment. Here we present our view, based on 20 years of research, that the initial drop in blood flow at the time of rupture triggers genomic responses throughout the brain vasculature that manifest days later as increased vasoconstriction and decreased cerebral blood flow. We propose a novel treatment strategy to prevent DCI by early inhibition of the vascular mitogen-activated protein kinase (MAPK) pathway that triggers expression of vasoconstrictor and inflammatory mediators. We summarize evidence from experimental SAH models showing early treatment with MAPK inhibitors "switches off" these detrimental responses, maintains flow, and improves neurological outcome. This promising therapy is currently being evaluated in clinical trials.
Collapse
Affiliation(s)
- Lars Edvinsson
- Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University, Sölvegatan 19, 22100, Lund, Sweden.
- Department of Experimental Research, Glostrup Research Institute, CopenhagenUniversity, Copenhagen, Denmark.
| | - Diana N Krause
- Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University, Sölvegatan 19, 22100, Lund, Sweden
- Department of Pharmaceutical Sciences, SchoolofPharmacy&PharmaceuticalSciences, University of California at Irvine, Irvine, CA, USA
| |
Collapse
|
6
|
Lin S, Chen Z, Wu Z, Fei F, Xu Z, Tong Y, Sun W, Wang P. Involvement of PI3K/AKT Pathway in the Rapid Antidepressant Effects of Crocetin in Mice with Depression-Like Phenotypes. Neurochem Res 2024; 49:477-491. [PMID: 37935859 DOI: 10.1007/s11064-023-04051-2] [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: 06/08/2023] [Revised: 09/22/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023]
Abstract
The current first-line antidepressants have the drawback of slow onset, which greatly affects the treatment of depression. Crocetin, one of the main active ingredients in saffron (Crocus sativus L.), has been demonstrated to have antidepressant activities, but whether it has a rapid antidepressant effect remains unclear. This study aimed to investigate the onset, duration, and mechanisms of the rapid antidepressant activity of crocetin (20, 40 and 80 mg/kg, intraperitoneal injection) in male mice subjected to chronic restraint stress (CRS). The results of behavioral tests showed that crocetin exerted rapid antidepressant-like effect in mice with depression-like phenotypes, including rapid normalization of depressive-like behaviors within 3 h, and the effects could be maintained for 2 days. Hematoxylin-eosin (HE) and Nissl staining showed that crocetin ameliorated hippocampal neuroinflammation and nerve injuries in mice with depression-like phenotypes. The levels of inflammatory factors, corticosterone and pro brain-derived neurotrophic factor in crocetin-administrated mice serum were significantly reduced compared with those in the CRS group, as well as the levels of inflammatory factors in hippocampus. What's more, Western blot analyses showed that, compared to CRS-induced mice, the relative levels of mitogen-activated kinase phosphatase 1 and toll-like receptor 4 were significantly reduced after the administration of crocetin, and the relative expressions of extracellular signal-regulated kinase 1/2 (ERK1/2), cAMP-response element binding protein, phosphorylated phosphoinositide 3 kinase (p-PI3K)/PI3K, phosphorylated protein kinase B (p-AKT)/AKT, phosphorylated glycogen synthase kinase 3β (p-GSK3β)/GSK3β, phosphorylated mammalian target of rapamycin (p-mTOR)/mTOR were markedly upregulated. In conclusion, crocetin exerted rapid antidepressant effects via suppressing the expression of inflammatory cytokines and the apoptosis of neuronal cells through PI3K/AKT signaling pathways. The rapid antidepressant effect of crocetin (40 mg/kg) could be maintained for at least 2 days after single treatment.
Collapse
Affiliation(s)
- Susu Lin
- The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, People's Republic of China
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
| | - Ziwei Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
| | - Zhaoruncheng Wu
- School of Biomedical engineering, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Fei Fei
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
| | - Zijin Xu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China
- College of Pharmacy, Jiangxi Medical College, Shangrao, 334000, Jiangxi, People's Republic of China
| | - Yingpeng Tong
- Institute of Natural Medicine and Health Product, School of Advanced Study, Taizhou University, Taizhou, 318000, People's Republic of China
| | - Wenyu Sun
- The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, 314001, People's Republic of China.
| | - Ping Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, No. 18 Chaowang Road, Hangzhou, 310014, People's Republic of China.
| |
Collapse
|
7
|
Zhou S, Nao J. Nesfatin-1: A Biomarker and Potential Therapeutic Target in Neurological Disorders. Neurochem Res 2024; 49:38-51. [PMID: 37740893 DOI: 10.1007/s11064-023-04037-0] [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: 06/23/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Nesfatin-1 is a novel adipocytokine consisting of 82 amino acids with anorexic and anti-hyperglycemic properties. Further studies of nesfatin-1 have shown it to be closely associated with neurological disorders. Changes in nesfatin-1 levels are closely linked to the onset, progression and severity of neurological disorders. Nesfatin-1 may affect the development of neurological disorders and can indicate disease evolution and prognosis, thus informing the choice of treatment options. In addition, regulation of the expression or level of nesfatin-1 can improve the level of neuroinflammation, apoptosis, oxidative damage and other indicators. It is demonstrated that nesfatin-1 is involved in neuroprotection and may be a therapeutic target for neurological disorders. In this paper, we will also discuss the role of nesfatin-1 as a biomarker in neurological diseases and its potential mechanism of action in neurological diseases, providing new ideas for the diagnosis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Siyu Zhou
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
8
|
Park JS, Choe K, Lee HJ, Park TJ, Kim MO. Neuroprotective effects of osmotin in Parkinson's disease-associated pathology via the AdipoR1/MAPK/AMPK/mTOR signaling pathways. J Biomed Sci 2023; 30:66. [PMID: 37568205 PMCID: PMC10422754 DOI: 10.1186/s12929-023-00961-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most frequent age-related neurodegenerative disorder and is characterized by the loss of dopaminergic neurons. Both environmental and genetic aspects are involved in the pathogenesis of PD. Osmotin is a structural and functional homolog of adiponectin, which regulates the phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK) via adiponectin receptor 1 (AdipoR1), thus attenuating PD-associated pathology. Therefore, the current study investigated the neuroprotective effects of osmotin using in vitro and in vivo models of PD. METHODS The study used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced and neuron-specific enolase promoter human alpha-synuclein (NSE-hαSyn) transgenic mouse models and 1-methyl-4-phenylpyridinium (MPP+)- or alpha-synuclein A53T-treated cell models. MPTP was injected at a dose of 30 mg/kg/day for five days, and osmotin was injected twice a week at a dose of 15 mg/kg for five weeks. We performed behavioral tests and analyzed the biochemical and molecular changes in the substantia nigra pars compacta (SNpc) and the striatum. RESULTS Based on our study, osmotin mitigated MPTP- and α-synuclein-induced motor dysfunction by upregulating the nuclear receptor-related 1 protein (Nurr1) transcription factor and its downstream markers tyrosine hydroxylase (TH), dopamine transporter (DAT), and vesicular monoamine transporter 2 (VMAT2). From a pathological perspective, osmotin ameliorated neuronal cell death and neuroinflammation by regulating the mitogen-activated protein kinase (MAPK) signaling pathway. Additionally, osmotin alleviated the accumulation of α-synuclein by promoting the AMPK/mammalian target of rapamycin (mTOR) autophagy signaling pathway. Finally, in nonmotor symptoms of PD, such as cognitive deficits, osmotin restored synaptic deficits, thereby improving cognitive impairment in MPTP- and α-synuclein-induced mice. CONCLUSIONS Therefore, our findings indicated that osmotin significantly rescued MPTP/α-synuclein-mediated PD neuropathology. Altogether, these results suggest that osmotin has potential neuroprotective effects in PD neuropathology and may provide opportunities to develop novel therapeutic interventions for the treatment of PD.
Collapse
Affiliation(s)
- Jun Sung Park
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
| | - Kyonghwan Choe
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229ER Maastricht, the Netherlands
| | - Hyeon Jin Lee
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, G12 0ZD UK
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, 52828 Republic of Korea
- Alz-Dementia Korea Co., Jinju, 52828 Republic of Korea
| |
Collapse
|
9
|
Ren XQ, Huang X, Xing SY, Long Y, Yuan DH, Hong H, Tang SS. Neuroprotective effects of novel compound FMDB on cognition, neurogenesis and apoptosis in APP/PS1 transgenic mouse model of Alzheimer's disease. Neurochem Int 2023; 165:105510. [PMID: 36893915 DOI: 10.1016/j.neuint.2023.105510] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 02/02/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023]
Abstract
Clinical and experimental studies have shown that the sharp reduction of estrogen is one of the important reasons for the high incidence of Alzheimer's disease (AD) in elderly women, but there is currently no such drug for treatment of AD. Our group first designed and synthesized a novel compound R-9-(4fluorophenyl)-3-methyl-10,10,-Hydrogen-6-hydrogen-benzopyran named FMDB. In this study, our aim is to investigate the neuroprotective effects and mechanism of FMDB in APP/PS1 transgenic mice. 6 months old APP/PS1 transgenic mice were intragastrical administered with FMDB (1.25, 2.5 and 5 mg/kg) every other day for 8 weeks. LV-ERβ-shRNA was injected bilaterally into the hippocampus of APP/PS1 mice to knockdown estrogen receptor β (ERβ). We found that FMDB ameliorated cognitive impairment in the Morris water maze and novel object recognition tests, increased hippocampal neurogenesis and prevented hippocampal apoptotic responses in APP/PS1 mice. Importantly, FMDB activated nuclear ERβ mediated CBP/p300, CREB and brain-derived neurotrophic factor (BDNF) signaling, and membrane ERβ mediated PI3K/Akt, CREB and BDNF signaling in the hippocampus. Our study demonstrated the contributions and mechanism of FMDB to cognition, neurogenesis and apoptosis in APP/PS1 mice. These lay the experimental foundation for the development of new anti-AD drugs.
Collapse
Affiliation(s)
- Xiao-Qian Ren
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xin Huang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Shu-Yun Xing
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yan Long
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Dan-Hua Yuan
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Su-Su Tang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
10
|
Paschou M, Liaropoulou D, Kalaitzaki V, Efthimiopoulos S, Papazafiri P. Knockdown of Amyloid Precursor Protein Increases Ion Channel Expression and Alters Ca 2+ Signaling Pathways. Int J Mol Sci 2023; 24:ijms24032302. [PMID: 36768625 PMCID: PMC9917207 DOI: 10.3390/ijms24032302] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
Although the physiological role of the full-length Amyloid Precursor Protein (APP) and its proteolytic fragments remains unclear, they are definitively crucial for normal synaptic function. Herein, we report that the downregulation of APP in SH-SY5Y cells, using short hairpin RNA (shRNA), alters the expression pattern of several ion channels and signaling proteins that are involved in synaptic and Ca2+ signaling. Specifically, the levels of GluR2 and GluR4 subunits of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors (AMPAR) were significantly increased with APP knockdown. Similarly, the expression of the majority of endoplasmic reticulum (ER) residing proteins, such as the ER Ca2+ channels IP3R (Inositol 1,4,5-triphosphate Receptor) and RyR (Ryanodine Receptor), the Ca2+ pump SERCA2 (Sarco/endoplasmic reticulum Ca2+ ATPase 2) and the ER Ca2+ sensor STIM1 (Stromal Interaction Molecule 1) was upregulated. A shift towards the upregulation of p-AKT, p-PP2A, and p-CaMKIV and the downregulation of p-GSK, p-ERK1/2, p-CaMKII, and p-CREB was observed, interconnecting Ca2+ signal transduction from the plasma membrane and ER to the nucleus. Interestingly, we detected reduced responses to several physiological stimuli, with the most prominent being the ineffectiveness of SH-SY5Y/APP- cells to mobilize Ca2+ from the ER upon carbachol-induced Ca2+ release through IP3Rs and RyRs. Our data further support an emerging yet perplexing role of APP within a functional molecular network of membrane and cytoplasmic proteins implicated in Ca2+ signaling.
Collapse
Affiliation(s)
- Maria Paschou
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Danai Liaropoulou
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Vasileia Kalaitzaki
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
- Institute of Laboratory Animal Science, University of Zurich, 8952 Schlieren, Switzerland
| | - Spiros Efthimiopoulos
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
- Correspondence: (S.E.); (P.P.)
| | - Panagiota Papazafiri
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
- Correspondence: (S.E.); (P.P.)
| |
Collapse
|
11
|
Changes in the histopathology and in the proteins related to the MAPK pathway in the brains of rats exposed to pre and postnatal radiofrequency radiation over four generations. J Chem Neuroanat 2022; 126:102187. [DOI: 10.1016/j.jchemneu.2022.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
|
12
|
Tortosa E, Sengupta Ghosh A, Li Q, Wong WR, Hinkle T, Sandoval W, Rose CM, Hoogenraad CC. Stress-induced vesicular assemblies of dual leucine zipper kinase are signaling hubs involved in kinase activation and neurodegeneration. EMBO J 2022; 41:e110155. [PMID: 35611591 PMCID: PMC9289706 DOI: 10.15252/embj.2021110155] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/09/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) drive key signaling cascades during neuronal survival and degeneration. The localization of kinases to specific subcellular compartments is a critical mechanism to locally control signaling activity and specificity upon stimulation. However, how MAPK signaling components tightly control their localization remains largely unknown. Here, we systematically analyzed the phosphorylation and membrane localization of all MAPKs expressed in dorsal root ganglia (DRG) neurons, under control and stress conditions. We found that MAP3K12/dual leucine zipper kinase (DLK) becomes phosphorylated and palmitoylated, and it is recruited to sphingomyelin-rich vesicles upon stress. Stress-induced DLK vesicle recruitment is essential for kinase activation; blocking DLK-membrane interaction inhibits downstream signaling, while DLK recruitment to ectopic subcellular structures is sufficient to induce kinase activation. We show that the localization of DLK to newly formed vesicles is essential for local signaling. Inhibition of membrane internalization blocks DLK activation and protects against neurodegeneration in DRG neurons. These data establish vesicular assemblies as dynamically regulated platforms for DLK signaling during neuronal stress responses.
Collapse
Affiliation(s)
- Elena Tortosa
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, USA
| | | | - Qingling Li
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South San Francisco, CA, USA
| | - Weng Ruh Wong
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South San Francisco, CA, USA
| | - Trent Hinkle
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South San Francisco, CA, USA
| | - Wendy Sandoval
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South San Francisco, CA, USA
| | - Christopher M Rose
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South San Francisco, CA, USA
| | | |
Collapse
|
13
|
Ischemic Brain Stroke and Mesenchymal Stem Cells: An Overview of Molecular Mechanisms and Therapeutic Potential. Stem Cells Int 2022; 2022:5930244. [PMID: 35663353 PMCID: PMC9159823 DOI: 10.1155/2022/5930244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/12/2021] [Accepted: 05/04/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic brain injury is associated with a high rate of mortality and disability with no effective therapeutic strategy. Recently, a growing number of studies are focusing on mesenchymal stem cell-based therapies for neurodegenerative disorders. However, despite having the promising outcome of preclinical studies, the clinical application of stem cell therapy remained elusive due to little or no progress in clinical trials. The objective of this study was to provide a generalized critique for the role of mesenchymal stem cell therapy in ischemic stroke injury, its underlying mechanisms, and constraints on its preclinical and clinical applications. Thus, we attempted to present an overview of previously published reports to evaluate the progress and provide molecular basis of mesenchymal stem cells (MSCs) therapy and its application in preclinical and clinical settings, which could aid in designing an effective regenerative therapeutic strategy in the future.
Collapse
|
14
|
Dai X, Wang Y, Li Y, Zhong Y, Pei M, Long J, Dong X, Chen YL, Wang Q, Wang G, Gold BG, Vandenbark AA, Neve KA, Offner H, Wang C. Tyrphostin A9 protects axons in experimental autoimmune encephalomyelitis through activation of ERKs. Life Sci 2022; 294:120383. [PMID: 35143827 PMCID: PMC8920308 DOI: 10.1016/j.lfs.2022.120383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022]
Abstract
AIMS Small molecule compound tyrphostin A9 (A9), an inhibitor of platelet-derived growth factor (PDGF) receptor, was previously reported by our group to stimulate extracellular signal-regulated kinase 1 (ERK1) and 2 (ERK2) in neuronal cells in a PDGF receptor-irrelevant manner. The study aimed to investigate whether A9 could protect axons in experimental autoimmune encephalomyelitis through activation of ERKs. MAIN METHODS A9 treatment on the protection on neurite outgrowth in SH-SY5Y neuroblastoma cells and primary substantia nigra neuron cultures from the neurotoxin MPP+ were analyzed. Then, clinical symptoms as well as ERK1/2 activation, axonal protection induction, and the abundance increases of the regeneration biomarker GAP-43 in the CNS in the relapsing-remitting experimental autoimmune encephalomyelitis (EAE) model were verified. KEY FINDINGS A9 treatment could stimulate neurite outgrowth in SH-SY5Y neuroblastoma cells and protect primary substantia nigra neuron cultures from the neurotoxin MPP+. In the relapsing-remitting EAE model, oral administration of A9 successfully ameliorated clinical symptoms, activated ERK1/2, induced axonal protection, and increased the abundance of the regeneration biomarker GAP-43 in the CNS. Interestingly, gene deficiency of ERK1 or ERK2 disrupted the beneficial effects of A9 in MOG-35-55-induced EAE. SIGNIFICANCE These results demonstrated that small molecule compounds that stimulate persistent ERK activation in vitro and in vivo may be useful in protective or restorative treatment for neurodegenerative diseases.
Collapse
MESH Headings
- Animals
- Axons/drug effects
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Extracellular Signal-Regulated MAP Kinases/genetics
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Gene Expression Regulation/drug effects
- Humans
- Mice
- Mice, Inbred C57BL
- Neuroblastoma/drug therapy
- Neuroblastoma/metabolism
- Neuroblastoma/pathology
- Rats
- Rats, Sprague-Dawley
- Tyrphostins/pharmacology
Collapse
Affiliation(s)
- Xiaodong Dai
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yongmei Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yuexin Li
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, United States of America; Research Service, VA Portland Health Care System, Portland, OR 97239, United States of America
| | - Yongping Zhong
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, United States of America
| | - Min Pei
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jing Long
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xingchen Dong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yi-Li Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qi Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guifeng Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Bruce G Gold
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, United States of America
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, United States of America; Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, United States of America; Research Service, VA Portland Health Care System, Portland, OR 97239, United States of America
| | - Kim A Neve
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, United States of America; Research Service, VA Portland Health Care System, Portland, OR 97239, United States of America
| | - Halina Offner
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, United States of America; Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239, United States of America; Research Service, VA Portland Health Care System, Portland, OR 97239, United States of America
| | - Chunhe Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200126, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; Department of Neurology, Oregon Health & Science University, Portland, OR 97239, United States of America; Research Service, VA Portland Health Care System, Portland, OR 97239, United States of America.
| |
Collapse
|
15
|
Pathophysiology of neurodegenerative diseases: An interplay among axonal transport failure, oxidative stress, and inflammation? Semin Immunol 2022; 59:101628. [PMID: 35779975 PMCID: PMC9807734 DOI: 10.1016/j.smim.2022.101628] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Neurodegenerative diseases (NDs) are heterogeneous neurological disorders characterized by a progressive loss of selected neuronal populations. A significant risk factor for most NDs is aging. Considering the constant increase in life expectancy, NDs represent a global public health burden. Axonal transport (AT) is a central cellular process underlying the generation and maintenance of neuronal architecture and connectivity. Deficits in AT appear to be a common thread for most, if not all, NDs. Neuroinflammation has been notoriously difficult to define in relation to NDs. Inflammation is a complex multifactorial process in the CNS, which varies depending on the disease stage. Several lines of evidence suggest that AT defect, axonopathy and neuroinflammation are tightly interlaced. However, whether these impairments play a causative role in NDs or are merely a downstream effect of neuronal degeneration remains unsettled. We still lack reliable information on the temporal relationship between these pathogenic mechanisms, although several findings suggest that they may occur early during ND pathophysiology. This article will review the latest evidence emerging on whether the interplay between AT perturbations and some aspects of CNS inflammation can participate in ND etiology, analyze their potential as therapeutic targets, and the urge to identify early surrogate biomarkers.
Collapse
|
16
|
Mesa-Infante V, Afonso-Oramas D, Salas-Hernández J, Rodríguez-Núñez J, Barroso-Chinea P. Long-term exposure to GDNF induces dephosphorylation of Ret, AKT, and ERK1/2, and is ineffective at protecting midbrain dopaminergic neurons in cellular models of Parkinson's disease. Mol Cell Neurosci 2021; 118:103684. [PMID: 34826608 DOI: 10.1016/j.mcn.2021.103684] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 12/01/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) promotes differentiation, proliferation, and survival in different cell types, including dopaminergic neurons. Thus, GDNF has been proposed as a promising neuroprotective therapy in Parkinson's disease. Although findings from cellular and animal models of Parkinson's disease were encouraging, results emerging from clinical trials were not as good as expected, probably due to the inappropriate administration protocols. Despite the growing information on GDNF action mechanisms, many aspects of its pharmacological effects are still unclear and data from different studies are still contradictory. Considering that GDNF action mechanisms are mediated by its receptor tyrosine kinase Ret, which activates PI3K/AKT and MAPK/ERK signaling pathways, we aimed to investigate Ret activation and its effect over both signaling pathways in midbrain cell cultures treated with GDNF at different doses (0.3, 1, and 10 ng/ml) and times (15 min, 24 h, 24 h (7 days), and 7 continuous days). The results showed that short-term or acute (15 min, 24 h, and 24 h (7 days)) GDNF treatment in rat midbrain neurons increases Tyrosine hydroxylase (TH) expression and the phosphorylation levels of Ret (Tyr 1062), AKT (Ser 473), ERK1/2 (Thr202/Tyr204), S6 (Ser 235/236), and GSK3-β (Ser 9). However, the phosphorylation level of these kinases, TH expression, and dopamine uptake, decreased below basal levels after long-term or prolonged treatment with 1 and 10 ng/ml GDNF (7 continuous days). Our data suggest that long-term GDNF treatment inactivates the receptor by an unknown mechanism, affecting its neuroprotective capacity against degeneration caused by 6-OHDA or rotenone, while short-term exposure to GDNF promoted dopaminergic cell survival. These findings highlight the need to find new and more effective long-acting therapeutic approaches for disorders in which GDNF plays a beneficial role, including Parkinson's disease. In this regard, it is necessary to propose new GDNF treatment guidelines to regulate and control its long-term expression levels and optimize the clinical use of this trophic factor in patients with Parkinson's disease.
Collapse
Affiliation(s)
- V Mesa-Infante
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - D Afonso-Oramas
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain.
| | - J Salas-Hernández
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - J Rodríguez-Núñez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - P Barroso-Chinea
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
17
|
Tang SS, Xing SY, Zhang XJ, Ren XQ, Hong H, Long Y. Neuroprotective effects of novel compound Tozan on cognition, neurogenesis and apoptosis in diabetes. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1471. [PMID: 34734023 PMCID: PMC8506716 DOI: 10.21037/atm-21-4439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/10/2021] [Indexed: 11/06/2022]
Abstract
Background Cognitive impairment is a serious complication of diabetes that manifests as an impairment of spatial memory and learning ability. Its pathogenesis is unclear, and effective therapeutic drugs are very limited. Our group designed and synthesized a novel compound named 3-p-tolyl-9H-xanthen-9-one (Tozan). In this study, we sought to investigate the effects and mechanism of Tozan on diabetic cognitive impairment. Methods Methylglyoxal (MG)-induced SH-SY5Y cells and streptozotocin (STZ)-induced type 1 diabetic mice were treated with Tozan. Methyl thiazolul tetrazolium (MTT) and lactate dehydrogenase (LDH) were used to test cytotoxicity. Morris water maze (MWM) and Y-maze tests were used to evaluate cognitive function. Immunofluorescence and western blot analyses were used to evaluate neurogenesis, apoptosis, and signal transduction pathway-related proteins. In addition, Lentivirus (LV)-estrogen receptor beta (ERβ)-ribonucleic acid interference (RNAi) was used to knockdown the ERβ gene in SH-SY5Y cells. Results We found that Tozan ameliorated MG-induced cytotoxicity in SH-SY5Y cells, improved cognitive dysfunction in STZ-induced type 1 diabetic mice, increased neurogenesis, and prevented apoptotic responses in vitro and in vivo. Importantly, Tozan (2, 4, and 8 mg/kg) mediated phosphatidylinositol-3-kinase and protein kinase B cAMP-response element binding protein (PI3K/Akt-CREB) signaling by activating membrane ERβ, and a high dose of Tozan (8 mg/kg) mediated CREB signaling by activating nuclear ERβ in the hippocampus. Notably, Tozan did not have an anti-apoptosis and regeneration protective role in ERβ gene knockdown cells. Conclusions Our study demonstrates Tozan’s contributions to and role in cognition, neurogenesis, and apoptosis in diabetes, and lays an experimental foundation for the development of new anti-diabetic cognitive impairment drugs.
Collapse
Affiliation(s)
- Su-Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Shu-Yun Xing
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Xue-Jiao Zhang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Xiao-Qian Ren
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| | - Yan Long
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
18
|
Berezovskaya AS, Tyganov SA, Nikolaeva SD, Naumova AA, Merkulyeva NS, Shenkman BS, Glazova MV. Dynamic Foot Stimulations During Short-Term Hindlimb Unloading Prevent Dysregulation of the Neurotransmission in the Hippocampus of Rats. Cell Mol Neurobiol 2021; 41:1549-1561. [PMID: 32683580 PMCID: PMC11448613 DOI: 10.1007/s10571-020-00922-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022]
Abstract
Spaceflight and simulated microgravity both affect learning and memory, which are mostly controlled by the hippocampus. However, data about molecular alterations in the hippocampus in real or simulated microgravity conditions are limited. Adult Wistar rats were recruited in the experiments. Here we analyzed whether short-term simulated microgravity caused by 3-day hindlimb unloading (HU) will affect the glutamatergic and GABAergic systems of the hippocampus and how dynamic foot stimulation (DFS) to the plantar surface applied during HU can contribute in the regulation of hippocampus functioning. The results demonstrated a decreased expression of vesicular glutamate transporters 1 and 2 (VGLUT1/2) in the hippocampus after 3 days of HU, while glutamate decarboxylase 67 (GAD67) expression was not affected. HU also significantly induced Akt signaling and transcriptional factor CREB that are supposed to activate the neuroprotective mechanisms. On the other hand, DFS led to normalization of VGLUT1/2 expression and activity of Akt and CREB. Analysis of exocytosis proteins revealed the inhibition of SNAP-25, VAMP-2, and syntaxin 1 expression in DFS group proposing attenuation of excitatory neurotransmission. Thus, we revealed that short-term HU causes dysregulation of glutamatergic system of the hippocampus, but, at the same time, stimulates neuroprotective Akt-dependent mechanism. In addition, most importantly, we demonstrated positive effect of DFS on the hippocampus functioning that probably depends on the regulation of neurotransmitter exocytosis.
Collapse
Affiliation(s)
- Anna S Berezovskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez pr., 194223, St.Petersburg, Russia
| | - Sergey A Tyganov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Svetlana D Nikolaeva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez pr., 194223, St.Petersburg, Russia
| | - Alexandra A Naumova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez pr., 194223, St.Petersburg, Russia
| | - Natalia S Merkulyeva
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Boris S Shenkman
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Margarita V Glazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez pr., 194223, St.Petersburg, Russia.
| |
Collapse
|
19
|
Sun Y, Zhou Y, Shi X, Ma X, Peng X, Xie Y, Cao X. CircTYW1 serves as a sponge for microRNA-380 in accelerating neurological recovery following spinal cord injury via regulating FGF9. Cell Cycle 2021; 20:1828-1844. [PMID: 34375168 DOI: 10.1080/15384101.2021.1962634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
As one of the most severe kinds of neurological damage, spinal cord injury (SCI) contributes to persistent motor dysfunction and involves a large repertoire of gene alterations. The participation of circular RNAs (circRNAs) in neurological recovery following SCI needs to be clarified. In the current work, we attempted to assess the function of hsa_circRNA_0003962/circTYW1 and its underlying mechanism in SCI. By accessing the GEO repository, the expression of circTYW1, microRNA-380 (miR-380), and FGF9 in SCI and sham-operated rats was evaluated. PC12 cells after oxygen-glucose deprivation (OGD) treatment were prepared to mimic the SCI model. circTYW1 and FGF9 were poorly expressed, whereas miR-380 was highly expressed in the spinal cord tissues of SCI rats. circTYW1 promoted neurological recovery in SCI rats and inhibited apoptosis in spinal cord tissues. In PC12 cells exposed to OGD, circTYW1 suppressed PC12 cell apoptosis; however, miR-380 overexpression reversed the protective effect of circTYW1 on PC12 cells. Also, circTYW1 promoted FGF9 expression through competitively binding to miR-380, which activated the ERK1/2 signaling. In summary, our results demonstrated that declines in circTYW1 prevented SCI rats from neurological recovery by regulating the miR-380/FGF9/ERK1/2 axis, which might provide new understanding for SCI treatment.
Collapse
Affiliation(s)
- Yanpeng Sun
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province, Luoyang Henan, P.R. China
| | - Yingjie Zhou
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province, Luoyang Henan, P.R. China
| | - Xiangqin Shi
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province, Luoyang Henan, P.R. China
| | - Xiaoran Ma
- Faculty of Graduate Studies, Tianjin University of Traditional Chinese Medicine, Tianjin P.R. China
| | - Xiaodong Peng
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province, Luoyang Henan, P.R. China
| | - Yan Xie
- Biomedical Engineering Laboratory, Luoyang Orthopedic Hospital of Henan Province, Luoyang Henan, P.R. China
| | - Xiangyang Cao
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province, Luoyang Henan, P.R. China
| |
Collapse
|
20
|
Yu W, Fang H, Zhang L, Hu M, He S, Li H, Zhu H. Reversible Changes in BDNF Expression in MK-801-Induced Hippocampal Astrocytes Through NMDAR/PI3K/ERK Signaling. Front Cell Neurosci 2021; 15:672136. [PMID: 34054433 PMCID: PMC8160225 DOI: 10.3389/fncel.2021.672136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
Dizocilpine (MK-801), a non-competitive N-methyl-D-aspartic acid receptor (NMDA-R) antagonist, can induce schizophrenia-like symptoms in healthy individuals, implicating NMDA-R hypofunction in disease pathogenesis. Brain-derived neurotrophic factor (BDNF) is also implicated in schizophrenia, and expression is regulated by NMDA-R activity, suggesting a functional link. We previously found that BDNF signaling was upregulated by MK-801 in cultured hippocampal astrocytes, but the underlying mechanism is not clear. To address this issue, the levels of BDNF expression and secretion were evaluated in hippocampal astrocytes incubated with MK-801 by ELISA and qPCR, with and without NMDA co-incubation or pretreatment of either the ERK1/2 inhibitor, PD98059 or the PI3K inhibitor, LY294002. The apoptosis, viability, and proliferation of the astrocytes were also examined. In the current study, we demonstrate that MK-801 treatment (20 μM for 5 days) enhances the proliferation of rat cultured hippocampal astrocytes. Expression of BDNF mRNA was enhanced after 24 h in MK-801, but returned to near baseline over the next 24 h in the continued presence of MK-801. However, two successive 24-h treatments enhanced BDNF expression. These application regimens had no effect on apoptosis or proliferation rate. Co-addition of NMDA significantly inhibited MK-801-induced upregulation of BDNF. Similarly, MK-801-induced BDNF upregulation was blocked by pretreatment with inhibitors of PI3K and ERK1/2, but not by inhibitors of p38 and JNK. These findings suggested that astrocytes may contribute to the acute neurological and behavioral response to MK-801 treatment via a transient increase in BDNF expression involving NMDA-R–PI3K–ERK signaling.
Collapse
Affiliation(s)
- Wenjuan Yu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongwei Fang
- Department of Anesthesiology and Intensive Care Unit, Dongfang Hospital, Tongji University, Shanghai, China
| | - Lei Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miaowen Hu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sidi He
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huafang Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Clinical Research Center for Mental Health, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Hao Zhu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Sharma V, Fernando V, Letson J, Walia Y, Zheng X, Fackelman D, Furuta S. S-Nitrosylation in Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22094600. [PMID: 33925645 PMCID: PMC8124305 DOI: 10.3390/ijms22094600] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
S-nitrosylation is a selective and reversible post-translational modification of protein thiols by nitric oxide (NO), which is a bioactive signaling molecule, to exert a variety of effects. These effects include the modulation of protein conformation, activity, stability, and protein-protein interactions. S-nitrosylation plays a central role in propagating NO signals within a cell, tissue, and tissue microenvironment, as the nitrosyl moiety can rapidly be transferred from one protein to another upon contact. This modification has also been reported to confer either tumor-suppressing or tumor-promoting effects and is portrayed as a process involved in every stage of cancer progression. In particular, S-nitrosylation has recently been found as an essential regulator of the tumor microenvironment (TME), the environment around a tumor governing the disease pathogenesis. This review aims to outline the effects of S-nitrosylation on different resident cells in the TME and the diverse outcomes in a context-dependent manner. Furthermore, we will discuss the therapeutic potentials of modulating S-nitrosylation levels in tumors.
Collapse
|
22
|
The Role of CaMKII and ERK Signaling in Addiction. Int J Mol Sci 2021; 22:ijms22063189. [PMID: 33804804 PMCID: PMC8004038 DOI: 10.3390/ijms22063189] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Nicotine is the predominant addictive compound of tobacco and causes the acquisition of dependence through its interactions with nicotinic acetylcholine receptors and various neurotransmitter releases in the central nervous system. The Ca2+/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) play a pivotal role in synaptic plasticity in the hippocampus. CaMKII is involved in long-term potentiation induction, which underlies the consolidation of learning and memory; however, the roles of CaMKII in nicotine and other psychostimulant-induced addiction still require further investigation. This article reviews the molecular mechanisms and crucial roles of CaMKII and ERK in nicotine and other stimulant drug-induced addiction. We also discuss dopamine (DA) receptor signaling involved in nicotine-induced addiction in the brain reward circuitry. In the last section, we introduce the association of polyunsaturated fatty acids and cellular chaperones of fatty acid-binding protein 3 in the context of nicotine-induced addiction in the mouse nucleus accumbens and provide a novel target for the treatment of drug abuse affecting dopaminergic systems.
Collapse
|
23
|
Roque C, Pinto N, Vaz Patto M, Baltazar G. Astrocytes contribute to the neuronal recovery promoted by high-frequency repetitive magnetic stimulation in in vitro models of ischemia. J Neurosci Res 2021; 99:1414-1432. [PMID: 33522025 DOI: 10.1002/jnr.24792] [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: 07/15/2020] [Revised: 12/11/2020] [Accepted: 12/23/2020] [Indexed: 11/07/2022]
Abstract
After decades of effort, there are no effective clinical treatments to induce the recovery of ischemia-injured tissues, and among the several strategies that have been explored, repetitive transcranial magnetic stimulation has proven to be one of the most promising, with beneficial effects in limb motor function, aphasia, hemispatial neglect, or dysphagia. Despite the clinical evidences, little is known about the mechanisms underlying those effects. The present study aimed to explore the cellular and molecular effects of high-frequency repetitive magnetic stimulation (HF-rMS) on an in vitro model of ischemia. Using primary cortical cultures exposed to oxygen and glucose deprivation followed by reperfusion, we observed that HF-rMS treatment prevents the ischemia-induced neuronal death by 21.2%, and the neurite degeneration triggered by ischemia. Our results also demonstrate that with this treatment there is an increase of 89.2% on the number cells expressing ERK1/2, of 20.1% on the number of cells expressing c-Fos, and a synaptogenic effect, through an increase of 62.9% in the number of synaptic puncta as well as of 49.4% in their intensity. Interestingly, our results indicate that astrocytes are crucial to the beneficial effects triggered by HF-rMS after ischemia, thus suggesting a direct effect of HF-rMS on these cells. The modulation of astrocytes with this non-invasive brain stimulation technique is a promising approach to promote the recovery of ischemia-induced injured tissues; however, it is essential to understand how these effects can be modulated in order to optimize the protocols and enhance the beneficial outcomes.
Collapse
Affiliation(s)
- Cláudio Roque
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal.,Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Nuno Pinto
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Maria Vaz Patto
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal.,Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Graça Baltazar
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal.,Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| |
Collapse
|
24
|
Sharma HS, Sahib S, Tian ZR, Muresanu DF, Nozari A, Castellani RJ, Lafuente JV, Wiklund L, Sharma A. Protein kinase inhibitors in traumatic brain injury and repair: New roles of nanomedicine. PROGRESS IN BRAIN RESEARCH 2020; 258:233-283. [PMID: 33223036 DOI: 10.1016/bs.pbr.2020.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) causes physical injury to the cell membranes of neurons, glial and axons causing the release of several neurochemicals including glutamate and cytokines altering cell-signaling pathways. Upregulation of mitogen associated protein kinase (MAPK) and extracellular signal-regulated kinase (ERK) occurs that is largely responsible for cell death. The pharmacological blockade of these pathways results in cell survival. In this review role of several protein kinase inhibitors on TBI induced oxidative stress, blood-brain barrier breakdown, brain edema formation, and resulting brain pathology is discussed in the light of current literature.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bilbao, Spain
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| |
Collapse
|
25
|
Huang YH, Chung CL, Tsai HP, Tzou RD, Wu SC, Chai CY, Lee TC, Kwan AL. Impact of hyperglycemia on neuronal apoptosis after subarachnoid hemorrhage in rodent brain: An experimental research. Int J Surg 2020; 83:246-252. [PMID: 32739549 DOI: 10.1016/j.ijsu.2020.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Hyperglycemia, a derangement after subarachnoid hemorrhage (SAH), is known to be associated with unfavorable outcomes. Whether the connection between hyperglycemia and poor prognosis results from severe neuronal apoptosis is unknown, and we aim at investigating their relationship. MATERIAL AND METHODS Streptozotocin (STZ) was administrated to trigger hyperglycemia before SAH induction in Sprague-Dawley rats that were assigned to one of four groups: control, SAH only, hyperglycemia only, and SAH with hyperglycemia. The severity of neuronal apoptosis was analyzed by terminal deoxynucleotidyl transferase-mediated dUTP nickend labelling (TUNEL) staining of cerebral cortex. RESULTS When subjected to SAH, hyperglycemic animals had worse neurobehavioral functions than normoglycemic ones. Hyperglycemia-exacerbated apoptosis was evident by greater increases in cleaved caspase-3 expression and TUNEL-positive cell density in the SAH with hyperglycemia group than those in the SAH only group, whereas there was no significant difference in cleaved caspase-9 expression and Bax/Bcl-2 ratio between the two groups. Furthermore, there was a remarkable decrease in the ratio of phosphorylated extracellular regulated kinase (ERK)/total ERK in the hyperglycemic rats after SAH. CONCLUSION Hyperglycemia aggravated neuronal apoptosis after SAH and was associated with impaired neurological outcomes. Activation of the extrinsic caspase cascade through the ERK signal pathway may contribute to hyperglycemia-mediated apoptosis.
Collapse
Affiliation(s)
- Yu-Hua Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Li Chung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Surgery, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Hung-Pei Tsai
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Rong-Dar Tzou
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shu-Chuan Wu
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tao-Chen Lee
- Department of Neurosurgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Aij-Lie Kwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA; Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
26
|
Nagy EE, Frigy A, Szász JA, Horváth E. Neuroinflammation and microglia/macrophage phenotype modulate the molecular background of post-stroke depression: A literature review. Exp Ther Med 2020; 20:2510-2523. [PMID: 32765743 PMCID: PMC7401670 DOI: 10.3892/etm.2020.8933] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022] Open
Abstract
Increasing evidence hints to the central role of neuroinflammation in the development of post-stroke depression. Danger signals released in the acute phase of ischemia trigger microglial activation, along with the infiltration of neutrophils and macrophages. The increased secretion of proinflammatory cytokines interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor α (TNFα) provokes neuronal degeneration and apoptosis, whereas IL-6, interferon γ (IFNγ), and TNFα induce aberrant tryptophane degradation with the accumulation of the end-product quinolinic acid in resident glial cells. This promotes glutamate excitotoxicity via hyperexcitation of N-methyl-D-aspartate receptors and antagonizes 5-hydroxy-tryptamine, reducing synaptic plasticity and neuronal survival, thus favoring depression. In the post-stroke period, CX3CL1 and the CD200-CD200R interaction mediates the activation of glial cells, whereas CCL-2 attracts infiltrating macrophages. CD206 positive cells grant the removal of excessive danger signals; the high number of regulatory T cells, IL-4, IL-10, transforming growth factor β (TGFβ), and intracellular signaling via cAMP response element-binding protein (CREB) support the M2 type differentiation. In favorable conditions, these cells may exert efficient clearance, mediate tissue repair, and might be essential players in the downregulation of molecular pathways that promote post-stroke depression.
Collapse
Affiliation(s)
- Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Attila Frigy
- Department of Internal Medicine IV, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - József Attila Szász
- Neurology Clinic II, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540136 Targu Mures, Romania
| | - Emőke Horváth
- Department of Pathology, 'George Emil Palade' University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| |
Collapse
|
27
|
Bendová Z, Pačesová D, Novotný J. The day-night differences in ERK1/2, GSK3β activity and c-Fos levels in the brain, and the responsiveness of various brain structures to morphine. J Comp Neurol 2020; 528:2471-2495. [PMID: 32170720 DOI: 10.1002/cne.24906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/17/2020] [Accepted: 03/09/2020] [Indexed: 11/12/2022]
Abstract
As with other drugs or pharmaceuticals, opioids differ in their rewarding or analgesic effects depending on when they are applied. In the previous study, we have demonstrated the day/night difference in the sensitivity of the major circadian clock in the suprachiasmatic nucleus to a low dose of morphine, and showed the bidirectional effect of morphine on pERK1/2 and pGSK3β levels in the suprachiasmatic nucleus depending on the time of administration. The main aim of this study was to identify other brain structures that respond differently to morphine depending on the time of its administration. Using immunohistochemistry, we identified 44 structures that show time-of-day specific changes in c-Fos level and activity of ERK1/2 and GSK3β kinases in response to a single dose of 1 mg/kg morphine. Furthermore, comparison among control groups revealed the differences in the spontaneous levels of all markers with a generally higher level during the night, that is, in the active phase of the day. We thus provide further evidence for diurnal variations in the activity of brain regions outside the suprachiasmatic nucleus indicated by the temporal changes in the molecular substrate. We suggest that these changes are responsible for generating diurnal variation in the reward behavior or analgesic effect of opioid administration.
Collapse
Affiliation(s)
- Zdeňka Bendová
- Faculty of Science, Charles University, Prague, Czech Republic.,Department of Sleep Medicine and Chronobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Dominika Pačesová
- Faculty of Science, Charles University, Prague, Czech Republic.,Department of Sleep Medicine and Chronobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Jiří Novotný
- Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
28
|
Spritzer MD, Roy EA. Testosterone and Adult Neurogenesis. Biomolecules 2020; 10:biom10020225. [PMID: 32028656 PMCID: PMC7072323 DOI: 10.3390/biom10020225] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
It is now well established that neurogenesis occurs throughout adulthood in select brain regions, but the functional significance of adult neurogenesis remains unclear. There is considerable evidence that steroid hormones modulate various stages of adult neurogenesis, and this review provides a focused summary of the effects of testosterone on adult neurogenesis. Initial evidence came from field studies with birds and wild rodent populations. Subsequent experiments with laboratory rodents have tested the effects of testosterone and its steroid metabolites upon adult neurogenesis, as well as the functional consequences of induced changes in neurogenesis. These experiments have provided clear evidence that testosterone increases adult neurogenesis within the dentate gyrus region of the hippocampus through an androgen-dependent pathway. Most evidence indicates that androgens selectively enhance the survival of newly generated neurons, while having little effect on cell proliferation. Whether this is a result of androgens acting directly on receptors of new neurons remains unclear, and indirect routes involving brain-derived neurotrophic factor (BDNF) and glucocorticoids may be involved. In vitro experiments suggest that testosterone has broad-ranging neuroprotective effects, which will be briefly reviewed. A better understanding of the effects of testosterone upon adult neurogenesis could shed light on neurological diseases that show sex differences.
Collapse
Affiliation(s)
- Mark D. Spritzer
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA
- Correspondence: ; Tel.: 802-443-5676
| | - Ethan A. Roy
- Graduate School of Education, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
29
|
|
30
|
Calabrò M, Mandelli L, Crisafulli C, Lee SJ, Jun TY, Wang SM, Patkar AA, Masand PS, Han C, Pae CU, Serretti A. Genes Involved in Neurodevelopment, Neuroplasticity and Major Depression: No Association for CACNA1C, CHRNA7 and MAPK1. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2019; 17:364-368. [PMID: 31352702 PMCID: PMC6705106 DOI: 10.9758/cpn.2019.17.3.364] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 01/08/2023]
Abstract
Objective Genetics factors are likely to play a role in the risk, clinical presentation and treatment outcome in major depressive disorder (MDD). In this study, we investigated the role of three candidate genes for MDD; calcium voltage-gated channel subunit alpha1 C (CACNA1C ), cholinergic receptor nicotinic alpha 7 subunit (CHRNA7 ), and mitogen-activated protein kinase 1 (MAPK1). Methods Two-hundred forty-two MDD patients and 326 healthy controls of Korean ancestry served as samples for the analyses. Thirty-nine single nucleotide polymorphisms (SNPs) within CACNA1C, CHRNA7, and MAPK1 genes were genotyped and subsequently tested for association with MDD (primary analysis) and other clinical features (symptoms’ severity, age of onset, history of suicide attempt, treatment outcome) (secondary analyses). Single SNPs, haplotypes and epistatic analyses were performed. Results Single SNPs were not associated with disease risk and clinical features. However, a combination of alleles (haplotype) within MAPK1 was found associated with MDD-status. Secondary analyses detected a possible involvement of CACNA1C haplotype in resistance to antidepressant treatment. Conclusion These data suggest a role for MAPK1 and CACNA1C in MDD risk and treatment resistance, respectively. However, since many limitations characterize the analysis, the results must be considered with great caution and verified.
Collapse
Affiliation(s)
| | - Laura Mandelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina
| | - Soo-Jung Lee
- Department of Psychiatry and 8Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Tae-Youn Jun
- Department of Psychiatry and 8Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sheng-Min Wang
- Department of Psychiatry and 8Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ashwin A Patkar
- Department of Psychiatry and Behavioural Sciences, Duke University Medical Center, Durham, NC, USA
| | - Prakash S Masand
- Academic Medicine Education Institute, Duke-NUS Medical School, Singapore, Singapore.,Global Medical Education, New York, NY, USA
| | - Changsu Han
- Department of Psychiatry, Korea University College of Medicine, Seoul, Korea
| | - Chi-Un Pae
- Department of Psychiatry and 8Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Department of Psychiatry and Behavioural Sciences, Duke University Medical Center, Durham, NC, USA
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
31
|
Pardo E, Barake F, Godoy JA, Oyanadel C, Espinoza S, Metz C, Retamal C, Massardo L, Tapia-Rojas C, Inestrosa NC, Soza A, González A. GALECTIN-8 Is a Neuroprotective Factor in the Brain that Can Be Neutralized by Human Autoantibodies. Mol Neurobiol 2019; 56:7774-7788. [PMID: 31119556 DOI: 10.1007/s12035-019-1621-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/23/2019] [Indexed: 12/19/2022]
Abstract
Galectin-8 (Gal-8) is a glycan-binding protein that modulates a variety of cellular processes interacting with cell surface glycoproteins. Neutralizing anti-Gal-8 antibodies that block Gal-8 functions have been described in autoimmune and inflammatory disorders, likely playing pathogenic roles. In the brain, Gal-8 is highly expressed in the choroid plexus and accordingly has been detected in human cerebrospinal fluid. It protects against central nervous system autoimmune damage through its immune-suppressive potential. Whether Gal-8 plays a direct role upon neurons remains unknown. Here, we show that Gal-8 protects hippocampal neurons in primary culture against damaging conditions such as nutrient deprivation, glutamate-induced excitotoxicity, hydrogen peroxide (H2O2)-induced oxidative stress, and β-amyloid oligomers (Aβo). This protective action is manifested even after 2 h of exposure to the harmful condition. Pull-down assays demonstrate binding of Gal-8 to selected β1-integrins, including α3 and α5β1. Furthermore, Gal-8 activates β1-integrins, ERK1/2, and PI3K/AKT signaling pathways that mediate neuroprotection. Hippocampal neurons in primary culture produce and secrete Gal-8, and their survival decreases upon incubation with human function-blocking Gal-8 autoantibodies obtained from lupus patients. Despite the low levels of Gal-8 expression detected by real-time PCR in hippocampus, compared with other brain regions, the complete lack of Gal-8 in Gal-8 KO mice determines higher levels of apoptosis upon H2O2 stereotaxic injection in this region. Therefore, endogenous Gal-8 likely contributes to generate a neuroprotective environment in the brain, which might be eventually counteracted by human function-blocking autoantibodies.
Collapse
Affiliation(s)
- Evelyn Pardo
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisca Barake
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Juan A Godoy
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia Oyanadel
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Sofía Espinoza
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Claudia Metz
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Claudio Retamal
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Loreto Massardo
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Cheril Tapia-Rojas
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Fundación Ciencia y Vida, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Center for Healthy Brain Ageing, University of New South Wales, Sydney, NSW, Australia
- Center of Excellence in Biomedicine of Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Andrea Soza
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.
| | - Alfonso González
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.
- Fundación Ciencia y Vida, Santiago, Chile.
| |
Collapse
|
32
|
Effect of the HDAC Inhibitor, Sodium Butyrate, on Neurogenesis in a Rat Model of Neonatal Hypoxia-Ischemia: Potential Mechanism of Action. Mol Neurobiol 2019; 56:6341-6370. [PMID: 30767185 PMCID: PMC6682584 DOI: 10.1007/s12035-019-1518-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/29/2019] [Indexed: 12/16/2022]
Abstract
Neonatal hypoxic-ischemic (HI) brain injury likely represents the major cause of long-term neurodevelopmental disabilities in surviving babies. Despite significant investigations, there is not yet any known reliable treatment to reduce brain damage in suffering infants. Our recent studies in an animal model of HI revealed the therapeutic potential of a histone deacetylase inhibitor (HDACi). The neuroprotective action was connected with the stimulation of neurogenesis in the dentate gyrus subgranular zone. In the current study, we investigated whether HDACi-sodium butyrate (SB)-would also lead to neurogenesis in the subventricular zone (SVZ). By using a neonatal rat model of hypoxia-ischemia, we found that SB treatment stimulated neurogenesis in the damaged ipsilateral side, based on increased DCX labeling, and restored the number of neuronal cells in the SVZ ipsilateral to lesioning. The neurogenic effect was associated with inhibition of inflammation, expressed by a transition of microglia to the anti-inflammatory phenotype (M2). In addition, the administration of SB increased the activation of the TrkB receptor and the phosphorylation of the transcription factor-CREB-in the ipsilateral hemisphere. In contrast, SB administration reduced the level of HI-induced p75NTR. Together, these results suggest that BDNF-TrkB signaling plays an important role in SB-induced neurogenesis after HI. These findings provide the basis for clinical approaches targeted at protecting the newborn brain damage, which may prove beneficial for treating neonatal hypoxia-ischemia.
Collapse
|
33
|
Miras-Portugal MT, Queipo MJ, Gil-Redondo JC, Ortega F, Gómez-Villafuertes R, Gualix J, Delicado EG, Pérez-Sen R. P2 receptor interaction and signalling cascades in neuroprotection. Brain Res Bull 2018; 151:74-83. [PMID: 30593879 DOI: 10.1016/j.brainresbull.2018.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/27/2018] [Accepted: 12/19/2018] [Indexed: 02/08/2023]
Abstract
Nucleotides can contribute to the survival of different glial and neuronal models at the nervous system via activation of purinergic P2X and P2Y receptors. Their activation counteracts different proapoptotic events, such as excitotoxicity, mitochondrial impairment, oxidative stress and DNA damage, which concur to elicit cell loss in different processes of neurodegeneration and brain injury. Thus, it is frequent to find that different neuroprotective mediators converge in the activation of the same intracellular survival pathways to protect cells from death. The present review focuses on the role of P2Y1 and P2Y13 metabotropic receptors, and P2X7 ionotropic receptors to regulate the balance between survival and apoptosis. In particular, we analyze the intracellular pathways involved in the signaling of these nucleotide receptors to elicit survival, including calcium/PLC, PI3K/Akt/GSK3, MAPK cascades, and the expression of antioxidant and antiapoptotic genes. This review emphasizes the novel contribution of nucleotide receptors to maintain cell homeostasis through the regulation of MAP kinases and phosphatases. Unraveling the different roles found for nucleotide receptors in different models and cellular contexts may be crucial to delineate future therapeutic applications based on targeting nucleotide receptors for neuroprotection.
Collapse
Affiliation(s)
- Mª Teresa Miras-Portugal
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Mª José Queipo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Juan Carlos Gil-Redondo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Felipe Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Rosa Gómez-Villafuertes
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Javier Gualix
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain
| | - Esmerilda G Delicado
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Raquel Pérez-Sen
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| |
Collapse
|
34
|
Colle D, Santos DB, de Souza V, Lopes MW, Leal RB, de Souza Brocardo P, Farina M. Sodium selenite protects from 3-nitropropionic acid-induced oxidative stress in cultured primary cortical neurons. Mol Biol Rep 2018; 46:751-762. [PMID: 30511305 DOI: 10.1007/s11033-018-4531-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
Abstract
Selenium (Se) is an essential trace element for humans; its intake is needed to allow the proper synthesis of 25 different selenoproteins that are necessary to the normal functioning of several organs, including the brain. Accordingly, decreased Se levels have been associated with neurological disorders. In the present study, we investigated the potential beneficial effects of Se, as sodium selenite, against 3-nitropropionic acid (3-NP)-induced oxidative stress in primary cultures of mouse cortical neurons. 3-NP treatment caused a significant decrease in cellular viability, which was accompanied by decreases in mitochondrial complex II activity and reduced glutathione (GSH) content, as well as increases in reactive oxygen species (ROS) generation and oxidized glutathione (GSSG) levels. Sodium selenite pretreatment (6 days) attenuated 3-NP-induced decrease in cell viability. In addition, sodium selenite pretreatment significantly protected against 3-NP-induced increase in ROS generation and decrease in GSH/GSSG ratio. Of note, sodium selenite pretreatment did not change 3-NP-induced decrease of mitochondrial complex II activity, suggesting that Se modulates secondary events resultant from 3-NP-induced mitochondrial dyshomeostasis. In addition, sodium selenite pretreatment significantly increased glutathione peroxidase (GPx) activity. Our data provide insights into the mechanism of protection by sodium selenite, which is related, at least in part, to GPx induction.
Collapse
Affiliation(s)
- Dirleise Colle
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil. .,Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, CEP 88040-900, Brazil.
| | - Danúbia Bonfanti Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Viviane de Souza
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Mark William Lopes
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Rodrigo Bainy Leal
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil
| | - Patricia de Souza Brocardo
- Departamento de Ciências Morfológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Bloco C, Campus, Universitário Trindade, Florianópolis, Santa Catarina, CEP 88040-900, Brazil.
| |
Collapse
|
35
|
Chandwani MN, Creisher PS, O'Donnell LA. Understanding the Role of Antiviral Cytokines and Chemokines on Neural Stem/Progenitor Cell Activity and Survival. Viral Immunol 2018; 32:15-24. [PMID: 30307795 DOI: 10.1089/vim.2018.0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Viral infections of the central nervous system are accompanied by the expression of cytokines and chemokines that can be critical for the control of viral replication in the brain. The outcomes of cytokine/chemokine signaling in neural cells vary widely, with cell-specific effects on cellular activity, proliferation, and survival. Neural stem/progenitor cells (NSPCs) are often altered during viral infections, through direct infection by the virus or by the influence of immune cell activity or cytokine/chemokine signaling. However, it has been challenging to dissect the contribution of the virus and specific inflammatory mediators during an infection. In addition to initiating an antiviral program in infected NSPCs, cytokines/chemokines can induce multiple changes in NSPC behavior that can perturb NSPC numbers, differentiation into other neural cells, and migration to sites of injury, and ultimately brain development and repair. The focus of this review was to dissect the effects of common antiviral cytokines and chemokines on NSPC activity, and to consider the subsequent pathological consequences for the host from changes in NSPC function.
Collapse
Affiliation(s)
- Manisha N Chandwani
- Department of Pharmaceutical, Administrative, and Social Sciences, Graduate School of Pharmaceutical Sciences, Duquesne University School of Pharmacy , Pittsburgh, Pennsylvania
| | - Patrick S Creisher
- Department of Pharmaceutical, Administrative, and Social Sciences, Graduate School of Pharmaceutical Sciences, Duquesne University School of Pharmacy , Pittsburgh, Pennsylvania
| | - Lauren A O'Donnell
- Department of Pharmaceutical, Administrative, and Social Sciences, Graduate School of Pharmaceutical Sciences, Duquesne University School of Pharmacy , Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Abdel-Aleem GA, Khaleel EF. Rutin hydrate ameliorates cadmium chloride-induced spatial memory loss and neural apoptosis in rats by enhancing levels of acetylcholine, inhibiting JNK and ERK1/2 activation and activating mTOR signalling. Arch Physiol Biochem 2018; 124:367-377. [PMID: 29214892 DOI: 10.1080/13813455.2017.1411370] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study aimed at studying the potential neuroprotective effect of Rutin hydrate (RH) alone or in conjugation with α-tocopherol against cadmium chloride (CdCl2)-induced neurotoxicity and cognitive impairment in rats and to investigate the mechanisms of action. Rats intoxicated with CdCl2 were treated with the vehicle, RH, α-tocopherol or combined treatment were examined, and compared to control rats received vehicle or individual doses of either drug. Data confirmed that RH improves spatial memory function by increasing acetylcholine availability, boosting endogenous antioxidant potential, activating cell survival and inhibiting apoptotic pathways, an effect that is more effective when RH was conjugated with α-tocopherol. Mechanism of RH action includes activation of PP2A mediated inhibiting of ERK1/2 and JNK apoptotic pathways and inhibition of PTEN mediated activation of mTOR survival pathway. In conclusion, RH affords a potent neuroprotection against CdCl2-induced brain damage and memory dysfunction and co-administration of α-tocopherol enhances its activity.
Collapse
Affiliation(s)
- Ghada A Abdel-Aleem
- a Department of Medical Biochemistry, College of Medicine , King Khalid University , Abha , Saudi Arabia
- b Department of Medical Biochemistry, Faculty of Medicine , Tanta University , Tanta , Egypt
| | - Eman F Khaleel
- c Department of Medical Physiology, College of Medicine , King Khalid University , Abha , Saudi Arabia
- d Department of Medical Physiology, Faculty of Medicine , Cairo University , Cairo , Egypt
| |
Collapse
|
37
|
ERα and/or ERβ activation ameliorates cognitive impairment, neurogenesis and apoptosis in type 2 diabetes mellitus mice. Exp Neurol 2018; 311:33-43. [PMID: 30201537 DOI: 10.1016/j.expneurol.2018.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/30/2018] [Accepted: 09/05/2018] [Indexed: 12/17/2022]
Abstract
Estrogen receptors (ERs) are thought to be associated with the onset and progression of neurodegenerative injuries and diseases, but the relationship and mechanisms underlying between ERs and cognition in type 2 diabetes remain elusive. In the current study, we investigated the effects of ERα and ERβ on the cognition, neurogenesis and apoptosis in high-fat diet and streptozocin-induced diabetic mice. We found that ERα and/or ERβ activation using their agonists (0.5 mg/kg E2, PPT or DPN) ameliorate memory impairment in the Morris water maze and Y-maze tests, increase hippocampal neurogenesis and prevent hippocampal apoptotic responses. Importantly, treatment with the pharmacologic ERs agonists caused significant increases in the membrane ERα and ERβ expression and subsequent PI3K/Akt, CREB and BDNF activation in the hippocampus of type 2 diabetes mellitus mice. Our data indicate that ERα and ERβ are involved in the cognitive impairment in type 2 diabetes, and that activated ERs, such as application of ERs agonists, could be a novel and promising strategy for the treatment of diabetic cognitive impairment.
Collapse
|
38
|
Tang SS, Ren Y, Xu LJ, Cao JR, Hong H, Ji H, Hu QH. Activation of ERα and/or ERβ ameliorates cognitive impairment and apoptosis in streptozotocin-induced diabetic mice. Horm Behav 2018; 105:95-103. [PMID: 30096284 DOI: 10.1016/j.yhbeh.2018.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/18/2018] [Accepted: 08/07/2018] [Indexed: 02/02/2023]
Abstract
Estrogen receptors (ERs) are thought to be associated with the onset and progression of neurodegenerative injuries and diseases, but the relationship and mechanisms underlying between ERs and cognition in type 1 diabetes remain elusive. In the current study, we investigated the effects of ERα and ERβ on the memory impairment and apoptosis in streptozotocin-induced diabetic mice. We found that ERα and/or ERβ activation using their agonists (0.5 mg/kg E2, PPT or DPN) ameliorate memory impairment in the Morris water maze (MWM) and Y-maze tests and suppress apoptosis as evidenced by decreased caspase-3 activity and increased ratio of Bcl-2/Bax. Importantly, treatment with the pharmacologic ERs agonists caused significant increases in the membrane ERα and ERβ expression and subsequent PI3K/Akt, CREB and BDNF activation in the hippocampus of diabetic mice. Our data indicate that ERα and ERβ are involved in the cognitive impairment of type 1 diabetes and that activation of ERs via administration of ERs agonists could be a novel and promising strategy for the treatment of diabetic cognitive impairment.
Collapse
Affiliation(s)
- Su-Su Tang
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China.
| | - Yi Ren
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Li-Jie Xu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-Ran Cao
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Hong
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Ji
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Qing-Hua Hu
- Department of Pharmacology, Key Laboratory of Neuropsychiatric Diseases, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
39
|
Hung PL, Hsu MH, Yu HR, Wu KLH, Wang FS. Thyroxin Protects White Matter from Hypoxic-Ischemic Insult in the Immature Sprague⁻Dawley Rat Brain by Regulating Periventricular White Matter and Cortex BDNF and CREB Pathways. Int J Mol Sci 2018; 19:ijms19092573. [PMID: 30158497 PMCID: PMC6164053 DOI: 10.3390/ijms19092573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Periventricular white-matter (WM) injury is a prominent feature of brain injury in preterm infants. Thyroxin (T4) treatment reduces the severity of hypoxic-ischemic (HI)-mediated WM injury in the immature brain. This study aimed to delineate molecular events underlying T4 protection following periventricular WM injury in HI rats. Methods: Right common-carotid-artery ligation, followed by hypoxia, was performed on seven-day-old rat pups. The HI pups were injected with saline, or 0.2 or 1 mg/kg of T4 at 48–96 h postoperatively. Cortex and periventricular WM were dissected for real-time (RT)-quantitative polymerase chain reactions (PCRs), immunoblotting, and for immunofluorescence analysis of neurotrophins, myelin, oligodendrocyte precursors, and neointimal. Results: T4 significantly mitigated hypomyelination and oligodendrocyte death in HI pups, whereas angiogenesis of periventricular WM, observed using antiendothelium cell antibody (RECA-1) immunofluorescence and vascular endothelium growth factor (VEGF) immunoblotting, was not affected. T4 also increased the brain-derived neurotrophic factors (BDNFs), but not the nerve growth factor (NGF) expression of injured periventricular WM. However, phosphorylated extracellular signal regulated kinase (p-ERK) and phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB) concentrations, but not the BDNF downstream pathway kinases, p38, c-Jun amino-terminal kinase (c-JNK), or Akt, were reduced in periventricular WM with T4 treatment. Notably, T4 administration significantly increased BDNF and phosphorylated CREB in the overlying cortex of the HI-induced injured cortex. Conclusion: Our findings reveal that T4 reversed BNDF signaling to attenuate HI-induced WM injury by activating ERK and CREB pathways in the cortex, but not directly in periventricular WM. This study offers molecular insight into the neuroprotective actions of T4 in HI-mediated WM injury in the immature brain.
Collapse
Affiliation(s)
- Pi-Lien Hung
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 33303, Taiwan.
| | - Mei-Hsin Hsu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 33303, Taiwan.
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 33303, Taiwan.
| | - Kay L H Wu
- Center for Translational Research in Biomedical Sciences, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung 33303, Taiwan.
| | - Feng-Sheng Wang
- Core facility for Phenomics & Diagnostics, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, No123, Rd Ta-Pei, Niao-Song District, Kaohsiung 33303, Taiwan.
| |
Collapse
|
40
|
Sustained Activation of JNK Induced by Quinolinic Acid Alters the BDNF/TrkB Axis in the Rat Striatum. Neuroscience 2018; 383:22-32. [DOI: 10.1016/j.neuroscience.2018.04.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 04/10/2018] [Accepted: 04/24/2018] [Indexed: 12/26/2022]
|
41
|
Wattanathorn J, Kirisattayakul W, Suriharn B, Lertrat K. Functional Drink Containing the Extracts of Purple Corn Cob and Pandan Leaves, the Novel Cognitive Enhancer, Increases Spatial Memory and Hippocampal Neuron Density Through the Improvement of Extracellular Signal Regulated Protein Kinase Expression, Cholinergic Function, and Oxidative Status in Ovariectomized Rats. Rejuvenation Res 2018; 21:431-441. [PMID: 29847217 DOI: 10.1089/rej.2017.2009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Due to requirement of novel memory enhancer for menopausal women, this study aimed to determine safety and effect of the functional drink containing the extracts of purple corn cob and pandan leaves (PCP) on memory and brain changes in experimental menopause induced by bilateral ovariectomy (OVX). Acute toxicity of PCP was carried out in female Wistar rats. The results showed that LD50 was more than 2000 mg/kg BW. To determine the cognitive enhancing effect of PCP, OVX rats were orally treated with PCP at the doses of 20, 40, and 80 mg/kg BW for 28 days. The spatial memory was assessed every 7 days throughout the study period. At the end of the study, oxidative stress status, acetylcholinesterase (AChE) activity, monoamine oxidase (MAO) activity, neuronal density, and extracellular signal regulated protein kinase 1 and 2 (ERK1/2) signaling in hippocampus were measured. The improved spatial memory, ERK1/2 expression, and neuron density in dentate gyrus of hippocampus were observed in PCP-treated rats. In addition, a reduction of AChE activity was also observed. Unfortunately, no improved oxidative stress status was observed. Taken altogether, PCP exerts the memory-enhancing effect partly through the suppression of AChE and the increase in ERK signaling in the hippocampus.
Collapse
Affiliation(s)
- Jintanaporn Wattanathorn
- 1 Department of Physiology, Faculty of Medicine, Khon Kaen University , Khon Kaen, Thailand .,2 Integrative Complementary Alternative Medicine Research and Development Center, Khon Kaen University , Khon Kaen, Thailand
| | - Woranan Kirisattayakul
- 2 Integrative Complementary Alternative Medicine Research and Development Center, Khon Kaen University , Khon Kaen, Thailand .,3 Department of Physiology and Graduate School (Neuroscience Program), Faculty of Medicine, Khon Kaen University , Khon Kaen, Thailand
| | - Bhalang Suriharn
- 4 Department of Plant Science and Agricultural Resources, Faculty of Agriculture, Khon Kaen University , Khon Kaen, Thailand
| | - Kamol Lertrat
- 4 Department of Plant Science and Agricultural Resources, Faculty of Agriculture, Khon Kaen University , Khon Kaen, Thailand
| |
Collapse
|
42
|
Region-Dependent Alterations in Cognitive Function and ERK1/2 Signaling in the PFC in Rats after Social Defeat Stress. Neural Plast 2018; 2018:9870985. [PMID: 29849577 PMCID: PMC5925180 DOI: 10.1155/2018/9870985] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 02/26/2018] [Indexed: 12/23/2022] Open
Abstract
Cognitive dysfunctions are highly comorbid with depression. Impairments of cognitive flexibility, which are modulated by the monoaminergic system of the prefrontal cortex (PFC), are increasingly recognized as an important component of the pathophysiology and treatment of depression. However, the downstream molecular mechanisms remain unclear. Using a classical model of depression, this study investigated the effects of social defeat stress on emotional behaviors, on cognitive flexibility in the attentional set-shifting task (AST), and on the expression of extracellular signal-regulated kinase 1 and 2 (ERK1 and ERK2) and their downstream signaling molecules cAMP-response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in two subregions of the PFC, the medial prefrontal cortex (mPFC), and the orbitofrontal cortex (OFC). The results showed that stress induced emotional and cognitive alterations associated with depression, including a decreased sucrose intake ratio and impaired reversal learning and set-shifting performance in the AST. Additionally, rats in the stress group showed a significant decrease only in ERK2 signaling in the mPFC, while more extensive decreases in both ERK1 signaling and ERK2 signaling were observed in the OFC. Along with the decreased ERK signaling, compared to controls, stressed rats showed downregulation of CREB phosphorylation and BDNF expression in both the OFC and the mPFC. Further analysis showed that behavioral changes were differentially correlated with several molecules in subregions of the PFC. These results suggested that social defeat stress was an effective animal model to induce both emotional and cognitive symptoms of depression and that the dysfunction of ERK signaling activities in the PFC might be a potential underlying biological mechanism.
Collapse
|
43
|
Mammone T, Chidlow G, Casson RJ, Wood JPM. Expression and activation of mitogen-activated protein kinases in the optic nerve head in a rat model of ocular hypertension. Mol Cell Neurosci 2018; 88:270-291. [PMID: 29408550 DOI: 10.1016/j.mcn.2018.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 01/03/2018] [Accepted: 01/11/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glaucoma is a leading cause of irreversible blindness manifesting as an age-related, progressive optic neuropathy with associated retinal ganglion cell (RGC) loss. Mitogen-activated protein kinases (MAPKs: p42/44 MAPK, SAPK/JNK, p38 MAPK) are activated in various retinal disease models and likely contribute to the mechanisms of RGC death. Although MAPKs play roles in the development of retinal pathology, their action in the optic nerve head (ONH), where the initial insult to RGC axons likely resides in glaucoma, remains unexplored. METHODS An experimental paradigm representing glaucoma was established by induction of chronic ocular hypertension (OHT) via laser-induced coagulation of the trabecular meshwork in Sprague-Dawley rats. MAPKs were subsequently investigated over the following days for expression and activity alterations, using RT-PCR, immunohistochemistry and Western immunoblot. RESULTS p42/44 MAPK expression was unaltered after intraocular pressure (IOP) elevation, but there was a significant activation of this enzyme in ONH astrocytes after 6-24 h. Activated SAPK/JNK isoforms were present throughout healthy RGC axons but after IOP elevation or optic nerve crush, they both accumulated at the ONH, likely due to RGC axon transport disruption, and were subject to additional activation. p38 MAPK was expressed by a population of microglia which were significantly more populous following IOP elevation. However it was only significantly activated in microglia after 3 days, and then only in the ONH and optic nerve; in the retina it was solely activated in RGC perikarya. CONCLUSIONS In conclusion, each of the MAPKs showed a specific spatio-temporal expression and activation pattern in the retina, ONH and optic nerve as a result of IOP elevation. These findings likely reflect the roles of the individual enzymes, and the cells in which they reside, in the developing pathology following IOP elevation. These data have implications for understanding the mechanisms of ocular pathology in diseases such as glaucoma.
Collapse
Affiliation(s)
- Teresa Mammone
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Glyn Chidlow
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Robert J Casson
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - John P M Wood
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
44
|
Caballero-Gallardo K, Wirbisky-Hershberger SE, Olivero-Verbel J, de la Rosa J, Freeman JL. Embryonic exposure to an aqueous coal dust extract results in gene expression alterations associated with the development and function of connective tissue and the hematological system, immunological and inflammatory disease, and cancer in zebrafish. Metallomics 2018; 10:463-473. [PMID: 29485154 DOI: 10.1039/c7mt00300e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Coal mining is one of the economic activities with the greatest impact on environmental quality. At all stages contaminants are released as particulates such as coal dust. The first aim of this study was to obtain an aqueous coal dust extract and characterize its composition in terms of trace elements by ICP-MS. In addition, the developmental toxicity of the aqueous coal extract was evaluated using zebrafish (Danio rerio) after exposure to different concentrations (0-1000 ppm; μg mL-1) to establish acute toxicity, morphology and transcriptome changes. Trace elements within the aqueous coal dust extract present at the highest concentrations (>10 ppb) included Sr, Zn, Ba, As, Cu and Se. In addition, Cd and Pb were found in lower concentrations. No significant difference in mortality was observed (p > 0.05), but a delay in hatching was found at 0.1 and 1000 ppm (p < 0.05). No significant differences in morphological characteristics were observed in any of the treatment groups (p > 0.05). Transcriptomic results of zebrafish larvae revealed alterations in 77, 61 and 1376 genes in the 1, 10, and 100 ppm groups, respectively. Gene ontology analysis identified gene alterations associated with the development and function of connective tissue and the hematological system, as well as pathways associated with apoptosis, the cell cycle, transcription, and oxidative stress including the MAPK signaling pathway. In addition, altered genes were associated with cancer; connective tissue, muscular, and skeletal disorders; and immunological and inflammatory diseases. Overall, this is the first study to characterize gene expression alterations in response to developmental exposure to aqueous coal dust residue from coal mining with transcriptome results signifying functions and systems to target in future studies.
Collapse
Affiliation(s)
- Karina Caballero-Gallardo
- Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, Zaragocilla Campus, University of Cartagena, Cartagena 130015, Colombia
| | | | - Jesus Olivero-Verbel
- Environmental and Computational Chemistry Group, School of Pharmaceutical Sciences, Zaragocilla Campus, University of Cartagena, Cartagena 130015, Colombia
| | - Jesus de la Rosa
- Associate Unit CSIC - University of Huelva ''Atmospheric Pollution'', Center for Research in Sustainable Chemistry (CIQSO), University of Huelva, E21071 Huelva, Spain
| | - Jennifer L Freeman
- School of Health Sciences, 550 Stadium Mall Drive Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
45
|
Torra A, Parent A, Cuadros T, Rodríguez-Galván B, Ruiz-Bronchal E, Ballabio A, Bortolozzi A, Vila M, Bové J. Overexpression of TFEB Drives a Pleiotropic Neurotrophic Effect and Prevents Parkinson's Disease-Related Neurodegeneration. Mol Ther 2018; 26:1552-1567. [PMID: 29628303 DOI: 10.1016/j.ymthe.2018.02.022] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/16/2018] [Accepted: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
The possible implication of transcription factor EB (TFEB) as a therapeutic target in Parkinson's disease has gained momentum since it was discovered that TFEB controls lysosomal biogenesis and autophagy and that its activation might counteract lysosomal impairment and protein aggregation. However, the majority of putative direct targets of TFEB described to date is linked to a range of biological processes that are not related to the lysosomal-autophagic system. Here, we assessed the effect of overexpressing TFEB with an adeno-associated viral vector in mouse substantia nigra dopaminergic neurons. We demonstrate that TFEB overexpression drives a previously unknown bona fide neurotrophic effect, giving rise to cell growth, higher tyrosine hydroxylase levels, and increased dopamine release in the striatum. TFEB overexpression induces the activation of the mitogen-activated protein kinase 1/3 (MAPK1/3) and AKT pro-survival pathways, phosphorylation of mTORC1 effectors 4E-binding protein 1 (4E-BP1) and S6 kinase B1 (S6K1), and increased protein synthesis. We show that TFEB overexpression prevents dopaminergic cell loss and counteracts atrophy and the associated protein synthesis decline in the MPTP mouse model of Parkinson's disease. Our results suggest that increasing TFEB activity might prevent neuronal death and restore neuronal function in Parkinson's disease and other neurodegenerative diseases through different mechanisms.
Collapse
Affiliation(s)
- Albert Torra
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Annabelle Parent
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Thais Cuadros
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Beatriz Rodríguez-Galván
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain
| | - Esther Ruiz-Bronchal
- Department of Neurochemistry and Neuropharmacology, IIBB-CSIC, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Center for Networked Biomedical Research on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Analía Bortolozzi
- Department of Neurochemistry and Neuropharmacology, IIBB-CSIC, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Center for Networked Biomedical Research on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain
| | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain; Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, Barcelona, Catalonia, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain.
| | - Jordi Bové
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute, Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Catalonia, Spain.
| |
Collapse
|
46
|
Joo MC, Jang CH, Park JT, Choi SW, Ro S, Kim MS, Lee MY. Effect of electrical stimulation on neural regeneration via the p38-RhoA and ERK1/2-Bcl-2 pathways in spinal cord-injured rats. Neural Regen Res 2018; 13:340-346. [PMID: 29557386 PMCID: PMC5879908 DOI: 10.4103/1673-5374.226404] [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] [Accepted: 01/19/2018] [Indexed: 11/26/2022] Open
Abstract
Although electrical stimulation is therapeutically applied for neural regeneration in patients, it remains unclear how electrical stimulation exerts its effects at the molecular level on spinal cord injury (SCI). To identify the signaling pathway involved in electrical stimulation improving the function of injured spinal cord, 21 female Sprague-Dawley rats were randomly assigned to three groups: control (no surgical intervention, n = 6), SCI (SCI only, n = 5), and electrical simulation (ES; SCI induction followed by ES treatment, n = 10). A complete spinal cord transection was performed at the 10th thoracic level. Electrical stimulation of the injured spinal cord region was applied for 4 hours per day for 7 days. On days 2 and 7 post SCI, the Touch-Test Sensory Evaluators and the Basso-Beattie-Bresnahan locomotor scale were used to evaluate rat sensory and motor function. Somatosensory-evoked potentials of the tibial nerve of a hind paw of the rat were measured to evaluate the electrophysiological function of injured spinal cord. Western blot analysis was performed to measure p38-RhoA and ERK1/2-Bcl-2 pathways related protein levels in the injured spinal cord. Rat sensory and motor functions were similar between SCI and ES groups. Compared with the SCI group, in the ES group, the latencies of the somatosensory-evoked potential of the tibial nerve of rats were significantly shortened, the amplitudes were significantly increased, RhoA protein level was significantly decreased, protein gene product 9.5 expression, ERK1/2, p38, and Bcl-2 protein levels in the spinal cord were significantly increased. These data suggest that ES can promote the recovery of electrophysiological function of the injured spinal cord through regulating p38-RhoA and ERK1/2-Bcl-2 pathway-related protein levels in the injured spinal cord.
Collapse
Affiliation(s)
- Min Cheol Joo
- Department of Rehabilitation Medicine and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, South Korea
| | - Chul Hwan Jang
- Department of Rehabilitation Medicine and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, South Korea
| | - Jong Tae Park
- Department of Neurosurgery, School of Medicine, Wonkwang University, Iksan, South Korea
| | - Seung Won Choi
- Department of Anesthesiology and Pain Medicine, School of Medicine, Wonkwang University, Iksan, South Korea
| | - Seungil Ro
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, USA
| | - Min Seob Kim
- Department of Physiology and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, South Korea
| | - Moon Young Lee
- Department of Physiology and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, South Korea
| |
Collapse
|
47
|
Malik J, Choudhary S. The Molecular Basis for Protective Effect of Mediterranean Diet in Neurodegenerative Disorders. ROLE OF THE MEDITERRANEAN DIET IN THE BRAIN AND NEURODEGENERATIVE DISEASES 2018:53-76. [DOI: 10.1016/b978-0-12-811959-4.00004-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
48
|
Kovács V, Tóth-Szűki V, Németh J, Varga V, Remzső G, Domoki F. Active forms of Akt and ERK are dominant in the cerebral cortex of newborn pigs that are unaffected by asphyxia. Life Sci 2017; 192:1-8. [PMID: 29138115 DOI: 10.1016/j.lfs.2017.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 11/03/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023]
Abstract
AIMS Perinatal asphyxia (PA) often results in hypoxic-ischemic encephalopathy (HIE) in term neonates. Introduction of therapeutic hypothermia improved HIE outcome, but further neuroprotective therapies are still warranted. The present study sought to determine the feasibility of the activation of the cytoprotective PI-3-K/Akt and the MAPK/ERK signaling pathways in the subacute phase of HIE development in a translational newborn pig PA/HIE model. MAIN METHODS Phosphorylated and total levels of Akt and ERK were determined by Western blotting in brain samples obtained from untreated naive, time control, and PA/HIE animals at 24-48h survival (n=3-3-6,respectively). PA (20min) was induced in anesthetized piglets by ventilation with a hypoxic/hypercapnic (6%O220%CO2) gas mixture. Furthermore, we studied the effect of topically administered specific Akt1/2 and MAPK/ERK kinase inhibitors on Akt and ERK phosphorylation (n=4-4) in the cerebral cortex under normoxic conditions. KEY FINDINGS PA resulted in significant neuronal injury shown by neuropathology assessment of haematoxylin/eosin stained sections. However, there were no significant differences among the groups in the high phosphorylation levels of both ERK and Akt in the cerebral cortex, hippocampus and subcortical structures. However, the Akt1/2 and MAPK/ERK kinase inhibitors significantly reduced cerebrocortical Akt and ERK phosphorylation within 30min. SIGNIFICANCE The major finding of the present study is that the PI-3-K/Akt and the MAPK/ERK signaling pathways appear to be constitutively active in the piglet brain, and this activation remains unaltered during HIE development. Thus, neuroprotective strategies aiming to activate these pathways to limit apoptotic neuronal death may offer limited efficacy in this translational model.
Collapse
Affiliation(s)
- Viktória Kovács
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary.
| | - Valéria Tóth-Szűki
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - János Németh
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - Viktória Varga
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - Gábor Remzső
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| | - Ferenc Domoki
- Department of Physiology, University of Szeged, School of Medicine, Szeged, Hungary
| |
Collapse
|
49
|
Wang H, Zhang Y, Li H, Zeng W, Qiao M. Shuyu capsules relieve liver- qi depression by regulating ERK-CREB-BDNF signal pathway in central nervous system of rat. Exp Ther Med 2017; 14:4831-4838. [PMID: 29201187 PMCID: PMC5704346 DOI: 10.3892/etm.2017.5125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/11/2017] [Indexed: 01/26/2023] Open
Abstract
The purpose of this study was to investigate the possible therapeutic mechanism of Shuyu capsules in liver-qi depression. Liver-qi depression rats were prepared based on chronic unpredictable mild stress (CUMS) and delayed constraint. Rats were gavaged with Shuyu capsule, fluoxetine, Radix Bupleuri and Radix Paeoniae Alba to constrct rat models. Body weight test, sucrose preference test and open-field test were applied to test rat models. Western blot analysis and quantitative real-time PCR was applied to determine the relative expression of extracellular signal-regulated protein kinase (ERK), cyclic AMP response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in hippocampus and frontal lobe tissues. ELISA was used to detect the content of BDNF in serum. Body weight, sugar intake and total distance were significantly decreased in depression group compared with control. The four drugs significantly increased levels of these factors. Compared with control group, ERK, CREB and BDNF expression were significantly decreased in depression group in both hippocampus and frontal lobe tissues at both mRNA and protein level. Shuyu capsule and fluoxetine group showed a significant increase in the expression of ERK, CREB and BDNF at mRNA, p-ERK and p-BDNF at protein level. Compared with Radix Paeoniae Alba, Radix Bupleuri were better in the rescue of ERK, CREB and BDNF expression. In conclusion, the pathogenesis of liver-qi depression associated with lower expression of ERK, CREB and BDNF in hippocampus and frontal. Shuyu capsule and main constitution alleviated the depressive-like behaviors and reversed the disruptions of the p-ERK, p-CREB and BDNF in stressed rats.
Collapse
Affiliation(s)
- Hongyan Wang
- Department of Traditional Chinese Medicine, Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Yingquan Zhang
- Department of Internal Medicine, The Hospital of Traditional Chinese Medicine of Tai'an, Tai'an, Shandong 271000, P.R. China
| | - Helou Li
- Department of Laboratory, Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Wei Zeng
- Department of Cardiology, Taishan Medical University, Tai'an, Shandong 271000, P.R. China
| | - Mingqi Qiao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| |
Collapse
|
50
|
Chen L, Serdyuk T, Yang B, Wang S, Chen S, Chu X, Zhang X, Song J, Bao H, Zhou C, Wang X, Dong S, Song L, Chen F, He G, He L, Zhou Y, Li W. Abnormal circadian oscillation of hippocampal MAPK activity and power spectrums in NF1 mutant mice. Mol Brain 2017; 10:29. [PMID: 28673309 PMCID: PMC5496334 DOI: 10.1186/s13041-017-0309-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/15/2017] [Indexed: 11/10/2022] Open
Abstract
Studies have implied that the circadian oscillation of mitogen-activated protein kinase (MAPK) signal pathways is crucial for hippocampus-dependent memory. NF1 mouse models (Nf1 heterozygous null mutants; Nf1 +/-) displayed enhanced MAPK activity in the hippocampus and resulted in memory deficits. We assumed a link between MAPK pathways and hippocampal rhythmic oscillations, which have never been explored in Nf1 +/- mice. We demonstrated that the level of extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation in Nf1 +/- mice were significantly higher at nighttime than at daytime. Moreover, the in vivo recording revealed that for the Nf1 +/- group, the power spectral density of theta rhythm significantly decreased and the firing rates of pyramidal neurons increased. Our results indicated that the hippocampal MAPK oscillation and theta rhythmic oscillations in Nf1 +/- mice were disturbed and hinted about a possible mechanism for the brain dysfunction in Nf1 +/- mice.
Collapse
Affiliation(s)
- Lei Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Tatiana Serdyuk
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Beimeng Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Shuai Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Shiqing Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Xixia Chu
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Xu Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Jinjing Song
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Hechen Bao
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Chengbin Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Xiang Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Shuangle Dong
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Lulu Song
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Fujun Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Ying Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| |
Collapse
|