101
|
Sharma K, Pannu V, Sayal N, Bhalla A, Anand A. Effects of one month of Common Yoga Protocol practice appear to be mediated by the angiogenic and neurogenic pathway: A pilot study. Explore (NY) 2020; 17:451-457. [PMID: 33059980 DOI: 10.1016/j.explore.2020.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 09/03/2020] [Accepted: 09/13/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To examine the molecular effects of mindful activities such as yoga and meditation DESIGN: This was an open label single arm exploratory yoga intervention study. STUDY PARTICIPANTS 64 healthy individuals within the age of 18-60 years were recruited for this one month yoga intervention study. INTERVENTION Common Yoga Protocol (CYP) is a standardized yoga protocol released by Ministry of AYUSH, India for International Yoga Day. It includes all aspects of yoga i.e. asanas, pranayama and meditation. It is designed for adoption by all age groups for the health of community. OUTCOME MEASURES The participants were assessed for biochemical parameters including Fasting Sugar and Lipid profile. The molecular markers of neurogenesis (i.e. Brain derived Neurotropic Factor, BDNF) and Angiogenesis (i.e. Vascular Endothelial Growth Factor, VEGF and Angiogenin) along with Amyloid β (marker related to neuro-degenerative diseases) were assessed. All the assessments were made at baseline and after one month of the intervention. RESULTS After one month of CYP practice High Density Lipoprotein (HDL) levels increased significantly (p<0.001), although other biochemical parameters i.e. fasting sugar and other lipid assessments were found to be unaltered. Angiogenesis marker, angiogenin was increased significantly (p<0.002), other angiogenesis marker VEGF did not show any change along with BDNF, marker of neurogenesis. Amyloid β levels were also unaltered. Even though individual levels of VEGF and Amyloid β did not show any change, proportion of VEGF to Amyloid β showed a significant increase (p<0.001) after one month of CYP intervention indicating that the change in VEGF levels was significantly higher than the change in Amyloid β levels. CONCLUSION CYP practice may influence cell survival pathways mediated by angiogenic and neurogenic cross talk. Hence, CYP can be considered as a preventive measure for diseases associated with impaired angiogenic and neurogenic mechanism. This is the first study to examine the effects of CYP at the molecular level.
Collapse
Affiliation(s)
- Kanupriya Sharma
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Viraaj Pannu
- Government Medical College and Hospital, Sector-32, Chandigarh, India
| | - Natasha Sayal
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashish Bhalla
- Department of Internal Medicine,, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
102
|
Basu A, Mestres I, Sahu SK, Tiwari N, Khongwir B, Baumgart J, Singh A, Calegari F, Tiwari VK. Phf21b imprints the spatiotemporal epigenetic switch essential for neural stem cell differentiation. Genes Dev 2020; 34:1190-1209. [PMID: 32820037 PMCID: PMC7462064 DOI: 10.1101/gad.333906.119] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
Cerebral cortical development in mammals involves a highly complex and organized set of events including the transition of neural stem and progenitor cells (NSCs) from proliferative to differentiative divisions to generate neurons. Despite progress, the spatiotemporal regulation of this proliferation-differentiation switch during neurogenesis and the upstream epigenetic triggers remain poorly known. Here we report a cortex-specific PHD finger protein, Phf21b, which is highly expressed in the neurogenic phase of cortical development and gets induced as NSCs begin to differentiate. Depletion of Phf21b in vivo inhibited neuronal differentiation as cortical progenitors lacking Phf21b were retained in the proliferative zones and underwent faster cell cycles. Mechanistically, Phf21b targets the regulatory regions of cell cycle promoting genes by virtue of its high affinity for monomethylated H3K4. Subsequently, Phf21b recruits the lysine-specific demethylase Lsd1 and histone deacetylase Hdac2, resulting in the simultaneous removal of monomethylation from H3K4 and acetylation from H3K27, respectively. Intriguingly, mutations in the Phf21b locus associate with depression and mental retardation in humans. Taken together, these findings establish how a precisely timed spatiotemporal expression of Phf21b creates an epigenetic program that triggers neural stem cell differentiation during cortical development.
Collapse
Affiliation(s)
- Amitava Basu
- Institute of Molecular Biology, 55128 Mainz, Germany
| | - Iván Mestres
- Center for Regenerative Therapies Dresden (CRTD), School of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | | | - Neha Tiwari
- Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg-University Mainz, 55128 Mainz, Germany
| | | | - Jan Baumgart
- Translational Animal Research Center (TARC), University Medical Centre, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Aditi Singh
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queens University Belfast, Belfast BT9 7BL, United Kingdom
| | - Federico Calegari
- Center for Regenerative Therapies Dresden (CRTD), School of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queens University Belfast, Belfast BT9 7BL, United Kingdom
| |
Collapse
|
103
|
Systems biology reveals reprogramming of the S-nitroso-proteome in the cortical and striatal regions of mice during aging process. Sci Rep 2020; 10:13913. [PMID: 32807865 PMCID: PMC7431412 DOI: 10.1038/s41598-020-70383-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/28/2020] [Indexed: 12/26/2022] Open
Abstract
Cell aging depends on the rate of cumulative oxidative and nitrosative damage to DNA and proteins. Accumulated data indicate the involvement of protein S-nitrosylation (SNO), the nitric oxide (NO)-mediated posttranslational modification (PTM) of cysteine thiols, in different brain disorders. However, the changes and involvement of SNO in aging including the development of the organism from juvenile to adult state is still unknown. In this study, using the state-of-the-art mass spectrometry technology to identify S-nitrosylated proteins combined with large-scale computational biology, we tested the S-nitroso-proteome in juvenile and adult mice in both cortical and striatal regions. We found reprogramming of the S-nitroso-proteome in adult mice of both cortex and striatum regions. Significant biological processes and protein–protein clusters associated with synaptic and neuronal terms were enriched in adult mice. Extensive quantitative analysis revealed a large set of potentially pathological proteins that were significantly upregulated in adult mice. Our approach, combined with large scale computational biology allowed us to perform a system-level characterization and identification of the key proteins and biological processes that can serve as drug targets for aging and brain disorders in future studies.
Collapse
|
104
|
Li W, Wu M, Zhang Y, Wei X, Zang J, Liu Y, Wang Y, Gong CX, Wei W. Intermittent fasting promotes adult hippocampal neuronal differentiation by activating GSK-3β in 3xTg-AD mice. J Neurochem 2020; 155:697-713. [PMID: 32578216 DOI: 10.1111/jnc.15105] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 12/26/2022]
Abstract
Moderate dietary restriction can ameliorate age-related chronic diseases such as Alzheimer's disease (AD) by increasing the expression of neurotrophic factors and promoting neurogenesis in the brain. Glycogen synthase kinase-3β (GSK-3β) signaling is essential for the coordination of progenitor cell proliferation and differentiation during brain development. The mechanisms by which GSK-3β is involved in dietary restriction-induced neurogenesis and cognitive improvement remain unclear. Six-month-old male 3xTg-AD and wild-type mice were fed on alternate days (intermittent fasting, IF) or ad libitum (AL) for 3 months. GSK-3β activity was regulated by bilaterally infusing lentiviral vectors carrying siRNA targeting GSK-3β into the dentate gyrus region of the hippocampus. Intermittent fasting promoted neuronal differentiation and maturation in the dentate gyrus and ameliorated recognized dysfunction in 3xTg-AD mice. These effects were reversed by siRNA targeting GSK-3β. After intermittent fasting, the insulin and protein kinase A signaling pathways were inhibited, while the adenosine monophosphate-activated protein kinase and brain-derived neurotrophic factor pathways were activated. These findings suggest that intermittent fasting can promote neuronal differentiation and maturation in the hippocampus by activating GSK-3β, thus improving learning and memory.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China.,Department of Pathology, The first people's hospital of foshan, Foshan, Guangdong, P. R. China
| | - Meijian Wu
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Yilin Zhang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Xuemin Wei
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Jiankun Zang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Yinghua Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, P. R. China
| | - Yanping Wang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wei Wei
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| |
Collapse
|
105
|
Liu Q, Jiao Y, Yang W, Gao B, Hsu DK, Nolta J, Russell M, Lyeth B, Zanto TP, Zhao M. Intracranial alternating current stimulation facilitates neurogenesis in a mouse model of Alzheimer's disease. Alzheimers Res Ther 2020; 12:89. [PMID: 32703308 PMCID: PMC7376967 DOI: 10.1186/s13195-020-00656-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/15/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Neurogenesis is significantly impaired in the brains of both human patients and experimental animal models of Alzheimer's disease (AD). Although deep brain stimulation promotes neurogenesis, it is an invasive technique that may damage neural circuitry along the path of the electrode. To circumvent this problem, we assessed whether intracranial electrical stimulation to the brain affects neurogenesis in a mouse model of Alzheimer's disease (5xFAD). METHODS AND RESULTS We used Ki67, Nestin, and doublecortin (DCX) as markers and determined that neurogenesis in both the subventricular zone (SVZ) and hippocampus were significantly reduced in the brains of 4-month-old 5xFAD mice. Guided by a finite element method (FEM) computer simulation to approximately estimate current and electric field in the mouse brain, electrodes were positioned on the skull that were likely to deliver stimulation to the SVZ and hippocampus. After a 4-week program of 40-Hz intracranial alternating current stimulation (iACS), neurogenesis indicated by expression of Ki67, Nestin, and DCX in both the SVZ and hippocampus were significantly increased compared to 5xFAD mice who received sham stimulation. The magnitude of neurogenesis was close to the wild-type (WT) age-matched unmanipulated controls. CONCLUSION Our results suggest that iACS is a promising, less invasive technique capable of effectively stimulating the SVZ and hippocampus regions in the mouse brain. Importantly, iACS can significantly boost neurogenesis in the brain and offers a potential treatment for AD.
Collapse
Affiliation(s)
- Qian Liu
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
- Center for Neuroscience, Department of Neurological Surgery, School of Medicine, University of California at Davis, Sacramento, CA, 95817, USA
| | - Yihang Jiao
- Department of Electrical and Computer Engineering, University of California at Davis, Davis, CA, 95616, USA
| | - Weijian Yang
- Department of Electrical and Computer Engineering, University of California at Davis, Davis, CA, 95616, USA
| | - Beiyao Gao
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
- Present location: Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, 200041, P. R. China
| | - Daniel K Hsu
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Jan Nolta
- Stem Cell Program and Gene Therapy Center, Institute for Regenerative Cures, Department of Internal Medicine, University of California at Davis, Sacramento, 95817, CA, USA
| | - Michael Russell
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA
| | - Bruce Lyeth
- Center for Neuroscience, Department of Neurological Surgery, School of Medicine, University of California at Davis, Sacramento, CA, 95817, USA
| | - Theodore P Zanto
- Neuroscape, Department of Neurology, University of California San Francisco - Mission Bay, Sandler Neuroscience Center MC 0444, San Francisco, CA, 94158, USA.
| | - Min Zhao
- Department of Dermatology, Institute for Regenerative Cures, University of California at Davis, School of Medicine, Sacramento, CA, 95817, USA.
- Center for Neuroscience, Department of Neurological Surgery, School of Medicine, University of California at Davis, Sacramento, CA, 95817, USA.
- Department of Ophthalmology and Vision Science, University of California at Davis, Sacramento, CA, 95616, USA.
| |
Collapse
|
106
|
Nguyen LD, Ehrlich BE. Cellular mechanisms and treatments for chemobrain: insight from aging and neurodegenerative diseases. EMBO Mol Med 2020; 12:e12075. [PMID: 32346964 PMCID: PMC7278555 DOI: 10.15252/emmm.202012075] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/09/2020] [Accepted: 04/01/2020] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy is a life-saving treatment for cancer patients, but also causes long-term cognitive impairment, or "chemobrain", in survivors. However, several challenges, including imprecise diagnosis criteria, multiple confounding factors, and unclear and heterogeneous molecular mechanisms, impede effective investigation of preventions and treatments for chemobrain. With the rapid increase in the number of cancer survivors, chemobrain is an urgent but unmet clinical need. Here, we leverage the extensive knowledge in various fields of neuroscience to gain insights into the mechanisms for chemobrain. We start by outlining why the post-mitotic adult brain is particularly vulnerable to chemotherapy. Next, through drawing comparisons with normal aging, Alzheimer's disease, and traumatic brain injury, we identify universal cellular mechanisms that may underlie the cognitive deficits in chemobrain. We further identify existing neurological drugs targeting these cellular mechanisms that can be repurposed as treatments for chemobrain, some of which were already shown to be effective in animal models. Finally, we briefly describe future steps to further advance our understanding of chemobrain and facilitate the development of effective preventions and treatments.
Collapse
Affiliation(s)
- Lien D Nguyen
- Department of Pharmacology and Interdepartmental Neuroscience ProgramYale UniversityNew HavenCTUSA
| | - Barbara E Ehrlich
- Department of Pharmacology and Interdepartmental Neuroscience ProgramYale UniversityNew HavenCTUSA
| |
Collapse
|
107
|
Li IC, Chang HH, Lin CH, Chen WP, Lu TH, Lee LY, Chen YW, Chen YP, Chen CC, Lin DPC. Prevention of Early Alzheimer's Disease by Erinacine A-Enriched Hericium erinaceus Mycelia Pilot Double-Blind Placebo-Controlled Study. Front Aging Neurosci 2020; 12:155. [PMID: 32581767 PMCID: PMC7283924 DOI: 10.3389/fnagi.2020.00155] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To investigate the efficacy and safety of three H. erinaceus mycelia (EAHE) capsules (350 mg/capsule; containing 5 mg/g erinacine A active ingredient) per day for the treatment of patients with mild Alzheimer's Disease (AD). METHODS This study comprised a 3-week no-drug screening period, followed by a 49-week double-blind treatment period with 2-parallel groups in which eligible patients were randomized to either three 5 mg/g EAHE mycelia capsules per day or identical appearing placebo capsules. Cognitive assessments, ophthalmic examinations, biomarker collection, and neuroimaging were followed throughout the study period. RESULTS After 49 weeks of EAHE intervention, a significant decrease in Cognitive Abilities Screening Instrument score was noted in the placebo group, a significant improvement in Mini-Mental State Examination score was observed in the EAHE group and a significant Instrumental Activities of Daily Living score difference were found between the two groups. In addition, EAHE group achieved a significantly better contrast sensitivity when compared to the placebo group. Moreover, only the placebo group observed significantly lowered biomarkers such as calcium, albumin, apolipoprotein E4, hemoglobin, and brain-derived neurotrophic factor and significantly elevated alpha1-antichymotrypsin and amyloid-beta peptide 1-40 over the study period. Using diffusion tensor imaging, the mean apparent diffusion coefficient (ADC) values from the arcuate fasciculus region in the dominant hemisphere significantly increased in the placebo group while no significant difference was found in the EAHE group in comparison to their baselines. Moreover, ADC values from the parahippocampal cingulum region in the dominant hemisphere significantly decreased in the EAHE group whereas no significant difference was found in the placebo group when compared to their baselines. Lastly, except for four subjects who dropped out of the study due to abdominal discomfort, nausea, and skin rash, no other adverse events were reported. CONCLUSION Three 350 mg/g EAHE capsules intervention for 49 weeks demonstrated higher CASI, MMSE, and IADL scores and achieved a better contrast sensitivity in patients with mild AD when compared to the placebo group, suggesting that EAHE is safe, well-tolerated, and may be important in achieving neurocognitive benefits. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, identifier NCT04065061.
Collapse
Affiliation(s)
- I-Chen Li
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Han-Hsin Chang
- Department of Nutrition, Chung Shan Medical University, Taichung City, Taiwan
| | - Chuan-Han Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
| | - Wan-Ping Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Tsung-Han Lu
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
| | - Li-Ya Lee
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Yu-Wen Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Yen-Po Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei City, Taiwan
- Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei City, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City, Taiwan
| | - David Pei-Cheng Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|
108
|
Redecker TM, Kisko TM, Wöhr M, Schwarting RKW. Cacna1c haploinsufficiency lacks effects on adult hippocampal neurogenesis and volumetric properties of prefrontal cortex and hippocampus in female rats. Physiol Behav 2020; 223:112974. [PMID: 32473156 DOI: 10.1016/j.physbeh.2020.112974] [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] [Received: 03/13/2020] [Revised: 04/30/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
The cross-disorder risk gene CACNA1C is strongly involved in the etiology of all major neuropsychiatric disorders, with women often being more affected by CACNA1C mutations than men. Human neuroimaging studies provided evidence that CACNA1C variants are associated with anatomical and functional brain alterations, such as decreased prefrontal volumes, microstructural changes in the hippocampus, and reduced hippocampal activity during memory tasks. In mouse models, Cacna1c alterations were repeatedly linked to disorder-like behavioral phenotypes and reduced adult hippocampal neurogenesis, which has been implicated in the pathology of neuropsychiatric disorders. Here, we applied a recently developed rat model and conducted two studies to investigate the effects of partial Cacna1c depletion on adult hippocampal neurogenesis and volumetric properties of the hippocampus and the prefrontal cortex in adult female constitutive heterozygous (Cacna1c+/-) rats and wildtype (Cacna1c+/+) littermate controls. In study 1, we analyzed proliferation versus survival of adult-born hippocampal cells based on a 5-bromodeoxyuridine assay ensuring neuronal cell-type specificity through applying an immunofluorescent multiple staining approach. In study 2, we performed a detailed volumetric analysis with high structural resolution of the dorsal hippocampus and the medial prefrontal cortex, including their major substructures. Our results indicate comparable levels of cell proliferation and neuronal survival in Cacna1c+/- rats and Cacna1c+/+ controls. Additionally, we found similar volumes of the dorsal hippocampus and the medial prefrontal cortex across major substructures irrespective of genotype, indicating that Cacna1c haploinsufficiency has no prominent effects on these brain features in female rats.
Collapse
Affiliation(s)
- Tobias M Redecker
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of Psychology, Philipps-Universität Marburg, Gutenbergstr. 18, D-35037 Marburg, Germany
| | - Theresa M Kisko
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of Psychology, Philipps-Universität Marburg, Gutenbergstr. 18, D-35037 Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of Psychology, Philipps-Universität Marburg, Gutenbergstr. 18, D-35037 Marburg, Germany; Center for Mind, Brain, and Behavior (CMBB), Hans-Meerwein-Str. 6, D-35032 Marburg, Germany; Laboratory for Behavioral Neuroscience, Department of Biology, Faculty of Science, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Rainer K W Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Department of Psychology, Philipps-Universität Marburg, Gutenbergstr. 18, D-35037 Marburg, Germany; Center for Mind, Brain, and Behavior (CMBB), Hans-Meerwein-Str. 6, D-35032 Marburg, Germany.
| |
Collapse
|
109
|
Yousefi N, Abdollahii S, Kouhbanani MAJ, Hassanzadeh A. Induced pluripotent stem cells (iPSCs) as game-changing tools in the treatment of neurodegenerative disease: Mirage or reality? J Cell Physiol 2020; 235:9166-9184. [PMID: 32437029 DOI: 10.1002/jcp.29800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/02/2020] [Accepted: 05/02/2020] [Indexed: 12/14/2022]
Abstract
Based on investigations, there exist tight correlations between neurodegenerative diseases' incidence and progression and aberrant protein aggregreferates in nervous tissue. However, the pathology of these diseases is not well known, leading to an inability to find an appropriate therapeutic approach to delay occurrence or slow many neurodegenerative diseases' development. The accessibility of induced pluripotent stem cells (iPSCs) in mimicking the phenotypes of various late-onset neurodegenerative diseases presents a novel strategy for in vitro disease modeling. The iPSCs provide a valuable and well-identified resource to clarify neurodegenerative disease mechanisms, as well as prepare a promising human stem cell platform for drug screening. Undoubtedly, neurodegenerative disease modeling using iPSCs has established innovative opportunities for both mechanistic types of research and recognition of novel disease treatments. Most important, the iPSCs have been considered as a novel autologous cell origin for cell-based therapy of neurodegenerative diseases following differentiation to varied types of neural lineage cells (e.g. GABAergic neurons, dopamine neurons, cortical neurons, and motor neurons). In this review, we summarize iPSC-based disease modeling in neurodegenerative diseases including Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, and Huntington's disease. Moreover, we discuss the efficacy of cell-replacement therapies for neurodegenerative disease.
Collapse
Affiliation(s)
- Niloufar Yousefi
- Department of Physiology and Pharmacology, Pasteur Instittableute of Iran, Tehran, Iran.,Stem Cell and Regenerative Medicine Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahla Abdollahii
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Stem Cell and Regenerative Medicine Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Stem Cell and Regenerative Medicine Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
110
|
Tripathi S, Verma A, Jha SK. Training on an Appetitive Trace-Conditioning Task Increases Adult Hippocampal Neurogenesis and the Expression of Arc, Erk and CREB Proteins in the Dorsal Hippocampus. Front Cell Neurosci 2020; 14:89. [PMID: 32362814 PMCID: PMC7181388 DOI: 10.3389/fncel.2020.00089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) plays an essential role in hippocampal-dependent memory consolidation. Increased neurogenesis enhances learning, whereas its ablation causes memory impairment. In contrast, few reports suggest that neurogenesis reduces after learning. Although the interest in exploring the role of adult neurogenesis in learning has been growing, the evidence is still limited. The role of the trace- and delay-appetitive-conditioning on AHN and its underlying mechanism are not known. The consolidation of trace-conditioned memory requires the hippocampus, but delay-conditioning does not. Moreover, the dorsal hippocampus (DH) and ventral hippocampus (VH) may have a differential role in these two conditioning paradigms. Here, we have investigated the changes in: (A) hippocampal cell proliferation and their progression towards neuronal lineage; and (B) expression of Arc, Erk1, Erk2, and CREB proteins in the DH and VH after trace- and delay-conditioning in the rat. The number of newly generated cells significantly increased in the trace-conditioned but did not change in the delay-conditioned animals compared to the control group. Similarly, the expression of Arc protein significantly increased in the DH but not in the VH after trace-conditioning. Nonetheless, it remains unaltered in the delay-conditioned group. The expression of pErk1, pErk2, and pCREB also increased in the DH after trace-conditioning. Whereas, the expression of only pErk1 pErk2 and pCREB proteins increased in the VH after delay-conditioning. Our results suggest that appetitive trace-conditioning enhances AHN. The increased DH neuronal activation and pErk1, pErk2, and pCREB in the DH may be playing an essential role in learning-induced cell-proliferation after appetitive trace-conditioning.
Collapse
Affiliation(s)
- Shweta Tripathi
- School of Life Science, Jawaharlal Nehru University, New Delhi, India
| | - Anita Verma
- School of Life Science, Jawaharlal Nehru University, New Delhi, India
| | - Sushil K Jha
- School of Life Science, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
111
|
Rodríguez-Barrera R, Flores-Romero A, García E, Fernández-Presas AM, Incontri-Abraham D, Navarro-Torres L, García-Sánchez J, Juárez-Vignon Whaley JJ, Madrazo I, Ibarra A. Immunization with neural-derived peptides increases neurogenesis in rats with chronic spinal cord injury. CNS Neurosci Ther 2020; 26:650-658. [PMID: 32352656 PMCID: PMC7248545 DOI: 10.1111/cns.13368] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/13/2020] [Accepted: 03/07/2020] [Indexed: 02/06/2023] Open
Abstract
Aims Immunization with neural‐derived peptides (INDP) has demonstrated to be a promising therapy to achieve a regenerative effect in the chronic phase of the spinal cord injury (SCI). Nevertheless, INDP‐induced neurogenic effects in the chronic stage of SCI have not been explored. Methods and Results In this study, we analyzed the effect of INDP on both motor and sensitive function recovery; afterward, we assessed neurogenesis and determined the production of cytokines (IL‐4, IL‐10, and TNF alpha) and neurotrophic factors (BDNF and GAP‐43). During the chronic stage of SCI, rats subjected to INDP showed a significant increase in both motor and sensitive recovery when compared to the control group. Moreover, we found a significant increase in neurogenesis, mainly at the central canal and at both the dorsal and ventral horns of INDP‐treated animals. Finally, INDP induced significant production of antiinflammatory and regeneration‐associated proteins in the chronic stages of SCI. Conclusions These findings suggest that INDP has a neurogenic effect that could improve motor and sensitive recovery in the chronic stage of SCI. Moreover, our results also envision the use of INDP as a possible therapeutic strategy for other trauma‐related disorders like traumatic brain injury.
Collapse
Affiliation(s)
- Roxana Rodríguez-Barrera
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Adrián Flores-Romero
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Elisa García
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Ana Maria Fernández-Presas
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Col. Universidad Nacional Autónoma de México, Coyoacan, Mexico
| | - Diego Incontri-Abraham
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Lisset Navarro-Torres
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Julián García-Sánchez
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Juan José Juárez-Vignon Whaley
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Ignacio Madrazo
- Proyecto CAMINA A.C, Tlalpan, Mexico.,Unidad de Investigación Médica en Enfermedades Neurológicas, CMN Siglo XXI, IMSS, Ciudad de México, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico.,Proyecto CAMINA A.C, Tlalpan, Mexico
| |
Collapse
|
112
|
Sahab-Negah S, Hajali V, Moradi HR, Gorji A. The Impact of Estradiol on Neurogenesis and Cognitive Functions in Alzheimer's Disease. Cell Mol Neurobiol 2020; 40:283-299. [PMID: 31502112 DOI: 10.1007/s10571-019-00733-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is described as cognitive and memory impairments with a sex-related epidemiological profile, affecting two times more women than men. There is emerging evidence that alternations in the hippocampal neurogenesis occur at the early stage of AD. Therapies that may effectively slow, stop, or regenerate the dying neurons in AD are being extensively investigated in the last few decades, but none has yet been found to be effective. The regulation of endogenous neurogenesis is one of the main therapeutic targets for AD. Mounting evidence indicates that the neurosteroid estradiol (17β-estradiol) plays a supporting role in neurogenesis, neuronal activity, and synaptic plasticity of AD. This effect may provide preventive and/or therapeutic approaches for AD. In this article, we discuss the molecular mechanism of potential estradiol modulatory action on endogenous neurogenesis, synaptic plasticity, and cognitive function in AD.
Collapse
Affiliation(s)
- Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Vahid Hajali
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Moradi
- Histology and Embryology Group, Basic Science Department, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Domagkstr. 11, Münster, Germany.
| |
Collapse
|
113
|
Ginsenoside Compound K Induces Adult Hippocampal Proliferation and Survival of Newly Generated Cells in Young and Elderly Mice. Biomolecules 2020; 10:biom10030484. [PMID: 32210026 PMCID: PMC7175218 DOI: 10.3390/biom10030484] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/20/2020] [Accepted: 03/21/2020] [Indexed: 12/14/2022] Open
Abstract
Cognitive impairment can be associated with reduced adult hippocampal neurogenesis, and it may contribute to age-associated neurodegenerative diseases such as Alzheimer’s (AD). Compound K (CK) is produced from the protopanaxadiol (PPD)-type ginsenosides Rb1, Rb2, and Rc by intestinal microbial conversion. Although CK has been reported as an inducing effector for neuroprotection and improved cognition in hippocampus, its effect on adult neurogenesis has not been explored yet. Here, we investigated the effect of CK on hippocampal neurogenesis in both young (2 months) and elderly (24 months) mice. CK treatment increased the number of cells co-labeled with 5-ethynyl-2′-deoxyuridine (EdU) and proliferating cell nuclear antigen (PCNA); also, Ki67, specific markers for progenitor cells, was more expressed, thus enhancing the generation of new cells and progenitor cells in the dentate gyrus of both young and elderly mice. Moreover, CK treatment increased the number of cells co-labeled with EdU and NeuN, a specific marker for mature neuron in the dentate gyrus, suggesting that newly generated cells survived and differentiated into mature neurons at both ages. These findings demonstrate that CK increases adult hippocampal neurogenesis, which may be beneficial against neurodegenerative disorders such as AD.
Collapse
|
114
|
Ottoboni L, von Wunster B, Martino G. Therapeutic Plasticity of Neural Stem Cells. Front Neurol 2020; 11:148. [PMID: 32265815 PMCID: PMC7100551 DOI: 10.3389/fneur.2020.00148] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have garnered significant scientific and commercial interest in the last 15 years. Given their plasticity, defined as the ability to develop into different phenotypes inside and outside of the nervous system, with a capacity of almost unlimited self-renewal, of releasing trophic and immunomodulatory factors, and of exploiting temporal and spatial dynamics, NSCs have been proposed for (i) neurotoxicity testing; (ii) cellular therapies to treat CNS diseases; (iii) neural tissue engineering and repair; (iv) drug target validation and testing; (v) personalized medicine. Moreover, given the growing interest in developing cell-based therapies to target neurodegenerative diseases, recent progress in developing NSCs from human-induced pluripotent stem cells has produced an analog of endogenous NSCs. Herein, we will review the current understanding on emerging conceptual and technological topics in the neural stem cell field, such as deep characterization of the human compartment, single-cell spatial-temporal dynamics, reprogramming from somatic cells, and NSC manipulation and monitoring. Together, these aspects contribute to further disentangling NSC plasticity to better exploit the potential of those cells, which, in the future, might offer new strategies for brain therapies.
Collapse
Affiliation(s)
- Linda Ottoboni
- Neurology and Neuroimmunology Unit, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Gianvito Martino
- Neurology and Neuroimmunology Unit, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, School of Medicine, Milan, Italy
| |
Collapse
|
115
|
Anam MB, Ahmad SAI, Kudo M, Istiaq A, Felemban AAM, Ito N, Ohta K. Akhirin regulates the proliferation and differentiation of neural stem cells/progenitor cells at neurogenic niches in mouse brain. Dev Growth Differ 2020; 62:97-107. [DOI: 10.1111/dgd.12646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Mohammad Badrul Anam
- Department of Developmental Neurobiology Faculty of Life Sciences Kumamoto University Kumamoto Japan
- HIGO Program Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
| | - Shah Adil Ishtiyaq Ahmad
- Department of Developmental Neurobiology Faculty of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
- Department of Biotechnology and Genetic Engineering Mawlana Bhashani Science and Technology University Tangail Bangladesh
| | - Mikiko Kudo
- Department of Developmental Neurobiology Faculty of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
| | - Arif Istiaq
- Department of Developmental Neurobiology Faculty of Life Sciences Kumamoto University Kumamoto Japan
- HIGO Program Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
| | - Athary Abdulhaleem M. Felemban
- Department of Developmental Neurobiology Faculty of Life Sciences Kumamoto University Kumamoto Japan
- Department of Biology Faculty of Applied Science Umm Al‐Qura University Makkah Saudi Arabia
| | - Naofumi Ito
- Department of Developmental Neurobiology Faculty of Life Sciences Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
| | - Kunimasa Ohta
- Department of Developmental Neurobiology Faculty of Life Sciences Kumamoto University Kumamoto Japan
- HIGO Program Kumamoto University Kumamoto Japan
- Stem Cell‐Based Tissue Regeneration Research and Education Unit Kumamoto University Kumamoto Japan
- AMED Core Research for Evolutional Science and Technology (AMED‐CREST) Japan Agency for Medical Research and Development (AMED) Tokyo Japan
| |
Collapse
|
116
|
Excitation/inhibition imbalance and impaired neurogenesis in neurodevelopmental and neurodegenerative disorders. Rev Neurosci 2019; 30:807-820. [DOI: 10.1515/revneuro-2019-0014] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022]
Abstract
AbstractThe excitation/inhibition (E/I) balance controls the synaptic inputs to prevent the inappropriate responses of neurons to input strength, and is required to restore the initial pattern of network activity. Various neurotransmitters affect synaptic plasticity within neural networks via the modulation of neuronal E/I balance in the developing and adult brain. Less is known about the role of E/I balance in the control of the development of the neural stem and progenitor cells in the course of neurogenesis and gliogenesis. Recent findings suggest that neural stem and progenitor cells appear to be the target for the action of GABA within the neurogenic or oligovascular niches. The same might be true for the role of neuropeptides (i.e. oxytocin) in neurogenic niches. This review covers current understanding of the role of E/I balance in the regulation of neuroplasticity associated with social behavior in normal brain, and in neurodevelopmental and neurodegenerative diseases. Further studies are required to decipher the GABA-mediated regulation of postnatal neurogenesis and synaptic integration of newly-born neurons as a potential target for the treatment of brain diseases.
Collapse
|
117
|
Cheng X, Yeung PKK, Zhong K, Zilundu PLM, Zhou L, Chung SK. Astrocytic endothelin-1 overexpression promotes neural progenitor cells proliferation and differentiation into astrocytes via the Jak2/Stat3 pathway after stroke. J Neuroinflammation 2019; 16:227. [PMID: 31733648 PMCID: PMC6858703 DOI: 10.1186/s12974-019-1597-y] [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: 12/02/2018] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Background Endothelin-1 (ET-1) is synthesized and upregulated in astrocytes under stroke. We previously demonstrated that transgenic mice over-expressing astrocytic ET-1 (GET-1) displayed more severe neurological deficits characterized by a larger infarct after transient middle cerebral artery occlusion (tMCAO). ET-1 is a known vasoconstrictor, mitogenic, and a survival factor. However, it is unclear whether the observed severe brain damage in GET-1 mice post stroke is due to ET-1 dysregulation of neurogenesis by altering the stem cell niche. Methods Non-transgenic (Ntg) and GET-1 mice were subjected to tMCAO with 1 h occlusion followed by long-term reperfusion (from day 1 to day 28). Neurological function was assessed using a four-point scale method. Infarct area and volume were determined by 2,3,5-triphenyltetra-zolium chloride staining. Neural stem cell (NSC) proliferation and migration in subventricular zone (SVZ) were evaluated by immunofluorescence double labeling of bromodeoxyuridine (BrdU), Ki67 and Sox2, Nestin, and Doublecortin (DCX). NSC differentiation in SVZ was evaluated using the following immunofluorescence double immunostaining: BrdU and neuron-specific nuclear protein (NeuN), BrdU and glial fibrillary acidic protein (GFAP). Phospho-Stat3 (p-Stat3) expression detected by Western-blot and immunofluorescence staining. Results GET-1 mice displayed a more severe neurological deficit and larger infarct area after tMCAO injury. There was a significant increase of BrdU-labeled progenitor cell proliferation, which co-expressed with GFAP, at SVZ in the ipsilateral side of the GET-1 brain at 28 days after tMCAO. p-Stat3 expression was increased in both Ntg and GET-1 mice in the ischemia brain at 7 days after tMCAO. p-Stat3 expression was significantly upregulated in the ipsilateral side in the GET-1 brain than that in the Ntg brain at 7 days after tMCAO. Furthermore, GET-1 mice treated with AG490 (a JAK2/Stat3 inhibitor) sh owed a significant reduction in neurological deficit along with reduced infarct area and dwarfed astrocytic differentiation in the ipsilateral brain after tMCAO. Conclusions The data indicate that astrocytic endothelin-1 overexpression promotes progenitor stem cell proliferation and astr ocytic differentiation via the Jak2/Stat3 pathway.
Collapse
Affiliation(s)
- Xiao Cheng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine, 111 Dade Road, Guangzhou, 510120, China. .,School of Biomedical Sciences, The University of Hong Kong, HKSAR, China. .,The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou, 510405, China. .,Guangdong Provincial Chinese Emergency Key Laboratory, Guangzhou, 510120, China. .,State Key Laboratory of Dampness Syndrome of Traditional Chinese Medicine, Guangzhou, 510120, China.
| | - Patrick K K Yeung
- School of Biomedical Sciences, The University of Hong Kong, HKSAR, China
| | - Ke Zhong
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangdong Province, Guangzhou, China
| | - Prince L M Zilundu
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangdong Province, Guangzhou, China
| | - Lihua Zhou
- Department of Anatomy, Zhong Shan School of Medicine, Sun Yat-Sen University, Guangdong Province, Guangzhou, China
| | - Sookja K Chung
- Faculty of Medicine, Macau University of Science and Technology, Macau, China. .,School of Biomedical Sciences, The University of Hong Kong, HKSAR, China.
| |
Collapse
|
118
|
Gultyaeva VV, Zinchenko MI, Uryumtsev DY, Krivoschekov SG, Aftanas LI. [Exercise for depression treatment. Physiological mechanisms]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:112-119. [PMID: 31464298 DOI: 10.17116/jnevro2019119071112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This literature review considers meta-analyzes, systematic reviews and original research over the last decade addressing a comprehensive analysis of the antidepressant effect of targeted physical exercise and physical activity in general. Exercise is a promising non-pharmacological treatment for depression, showing effects that are comparable or may even exceed other first-line treatments of depression. The article introduces modern ideas about the mechanisms of depression and mechanisms of exercise effects on depression manifestations. The structures of the central nervous system, changing with the effective exercise-based treatment of depression, are indicated. Physical activity stimulates the secretion of growth factors, maintenance of angio-, synapto-, and neurogenesis. The regulation of antioxidant protection of neuronal mitochondria, a decrease in pro-inflammatory reactions and stress reactivity are also observed in response to regular exercise. Physical activity has a multimodal effect that stimulates biochemical pathways and restores neuronal structures disturbed in depression.
Collapse
Affiliation(s)
- V V Gultyaeva
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - M I Zinchenko
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - D Y Uryumtsev
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - S G Krivoschekov
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | - L I Aftanas
- Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| |
Collapse
|
119
|
Running-Activated Neural Stem Cells Enhance Subventricular Neurogenesis and Improve Olfactory Behavior in p21 Knockout Mice. Mol Neurobiol 2019; 56:7534-7556. [DOI: 10.1007/s12035-019-1590-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/27/2019] [Indexed: 01/17/2023]
|
120
|
Aguareles J, Paraíso-Luna J, Palomares B, Bajo-Grañeras R, Navarrete C, Ruiz-Calvo A, García-Rincón D, García-Taboada E, Guzmán M, Muñoz E, Galve-Roperh I. Oral administration of the cannabigerol derivative VCE-003.2 promotes subventricular zone neurogenesis and protects against mutant huntingtin-induced neurodegeneration. Transl Neurodegener 2019; 8:9. [PMID: 30899454 PMCID: PMC6407204 DOI: 10.1186/s40035-019-0148-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 02/15/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The administration of certain cannabinoids provides neuroprotection in models of neurodegenerative diseases by acting through various cellular and molecular mechanisms. Many cannabinoid actions in the nervous system are mediated by CB1 receptors, which can elicit psychotropic effects, but other targets devoid of psychotropic activity, including CB2 and nuclear PPARγ receptors, can also be the target of specific cannabinoids. METHODS We investigated the pro-neurogenic potential of the synthetic cannabigerol derivative, VCE-003.2, in striatal neurodegeneration by using adeno-associated viral expression of mutant huntingtin in vivo and mouse embryonic stem cell differentiation in vitro. RESULTS Oral administration of VCE-003.2 protected striatal medium spiny neurons from mutant huntingtin-induced damage, attenuated neuroinflammation and improved motor performance. VCE-003.2 bioavailability was characterized and the potential undesired side effects were evaluated by analyzing hepatotoxicity after chronic treatment. VCE-003.2 promoted subventricular zone progenitor mobilization, increased doublecortin-positive migrating neuroblasts towards the injured area, and enhanced effective neurogenesis. Moreover, we demonstrated the proneurogenic activity of VCE-003.2 in embryonic stem cells. VCE-003.2 was able to increase neuroblast formation and striatal-like CTIP2-mediated neurogenesis. CONCLUSIONS The cannabigerol derivative VCE-003.2 improves subventricular zone-derived neurogenesis in response to mutant huntingtin-induced neurodegeneration, and is neuroprotective by oral administration.
Collapse
Affiliation(s)
- José Aguareles
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Paraíso-Luna
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Belén Palomares
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Raquel Bajo-Grañeras
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Andrea Ruiz-Calvo
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Daniel García-Rincón
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Elena García-Taboada
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Ctra. Colmenar Viejo, km, 9100 Madrid, Spain
- Departamento de Bioquímica y Biología Molecular and Instituto Universitario de Investigación Neuroquímica, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
121
|
de Jesus Fonseca EG, Pedroso A, Neuls D, Barbosa D, Cidral-Filho FJ, Salgado ASI, Dubiela A, Carraro E, Kerppers II. Study of transcranial therapy 904 nm in experimental model of stroke. Lasers Med Sci 2019; 34:1619-1625. [PMID: 30826952 DOI: 10.1007/s10103-019-02758-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
Cerebrovascular accidents (CVAs), commonly known as strokes, can damage the brain through vascular injuries caused by either blood vessel blockages (ischemic stroke) or ruptures (hemorrhagic stroke) which disrupt regular brain blood supply and can cause severe damage to the individual. The objective of the present study was to evaluate the effects of photobiomodulation with a light-emitting diode (LED) device (904 nm, 110 mW, 7 J/cm2) on neurogenesis, muscle resistance, and motor behavior in animals submitted to an experimental model of hemiplegia. The sample consisted of 30 Wistar rats, divided into two groups: control group (GC) and 904-nm LED-treated group (TG). All animals underwent stereotactic surgery for electrode implant and subsequent electrolytic injury to induce an ischemic stroke. TG was subjected to daily LED irradiation (904 nm, 110 mW, 7 J/cm2) for 63 s. Suspension test results indicate an improvement of TG muscle resistance when compared with baseline evaluation (BLT); a reduction in open-field freezing time and the number of fecal bolus pellets suggest diminished anxiety induced by 904-nm LED treatment on treatment days 7 and 21 (TG7 and TG21) compared with the baseline results; and lastly, histological analysis showed important signs of neurogenesis in TG in comparison to CG, especially on treatment days 7 and 21 (TG7 and TG21). In conclusion, the present study suggests that 904-nm LED irradiation may beneficially affect neurogenesis, muscle resistance, and animal motor behavior following ischemic CVA.
Collapse
Affiliation(s)
| | - Ariele Pedroso
- Laboratory of Neuroanatomy and Neurophysiology, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil
| | - Débora Neuls
- Laboratory of Neuroanatomy and Neurophysiology, Universidade Estadual do Centro-Oeste, Guarapuava, PR, Brazil
| | - Danilo Barbosa
- Department of Physical Therapy, Functional Neurology Discipline, Universidade Estadual do Centro Oeste, Guarapuava, PR, Brazil
| | - Francisco José Cidral-Filho
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | | | - Angela Dubiela
- Laboratório de Neuroanatomia e Neurofisiologia, Universidade Estadual do Centro-Oeste, Simeão Camargo Varela de Sá, 03, Guarapuava, Paraná, 88040-080, Brazil
| | - Emerson Carraro
- Laboratório de Imunologia, virologia e Biologia Molecular, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná, Brazil
| | - Ivo Ilvan Kerppers
- Laboratório de Neuroanatomia e Neurofisiologia, Universidade Estadual do Centro-Oeste, Simeão Camargo Varela de Sá, 03, Guarapuava, Paraná, 88040-080, Brazil.
| |
Collapse
|
122
|
Shevela E, Davydova M, Starostina N, Yankovskaya A, Ostanin A, Chernykh E. Intranasal delivery of M2 macrophage-derived soluble products reduces neuropsychological deficit in patients with cerebrovascular disease: a pilot study. JOURNAL OF NEURORESTORATOLOGY 2019. [DOI: 10.26599/jnr.2019.9040010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: We assessed the safety and clinical effectiveness of intranasal therapy with M2 macrophage-derived soluble products (M2-SPs) for treating patients with cerebrovascular disease (CVD). Materials and methods: The protocol of the study was registered at www.ClinicalTrails.gov (NCT02957123). The study group comprised 30 patients with chronic CVD. Neurological status was examined before therapy and at 1- and 6-month follow–up. Concentrations of 32 cytokines in the blood serum were evaluated before and 1 month after therapy onset. Neurological assessment was conducted with the following scales: Subjective Assessment of Clinical (neurological) Symptoms (SACS), Hospital Anxiety and Depression Scale (HADS), Functional Mobility Assessment in Eldery Patients (FMA), and Montreal Cognitive Assessment (MoCa). Results: M2-SPs treatment (once daily for 28~30 days) was found to be safe and well tolerated. Neuropsychological improvements showed the amelioration of neurological symptoms, reduction in anxiety and depression levels, improvement in balance and gait ability as well as cognitive functions. Clinical effects could be detected at the end of treatment course and was stable during 6-month follow-up. Blood serum cytokine evaluation demonstrated diminished baseline levels of many cytokines including those with neurotrophic activity (brain-derived neurotrophic factor, BDNF; hepatocyte growth factor, HGF; migration inhibitory factor, MIF). Upon treatment, most pronounced clinical responses were observed in patients with most severe cytokine deficiency and post-therapy normalization of MIF and HGF levels. Conclusion: Intranasal therapy with M2-SPs is safe and according to preliminary data reduces neuropsychological deficit in patients with chronic CVD. The positive effect of M2-SPs treatment seems to be HGF- and MIF-dependent.
Collapse
|
123
|
Anderson SR, Vetter ML. Developmental roles of microglia: A window into mechanisms of disease. Dev Dyn 2019; 248:98-117. [PMID: 30444278 PMCID: PMC6328295 DOI: 10.1002/dvdy.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/21/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia are engineers of the central nervous system (CNS) both in health and disease. In addition to the canonical immunological roles of clearing damaging entities and limiting the spread of toxicity and death, microglia remodel the CNS throughout life. While they have been extensively studied in disease and injury, due to their highly variable functions, their precise role in these contexts still remains uncertain. Over the past decade, we have greatly expanded our understanding of microglial function, including their essential homeostatic roles during development. Here, we review these developmental roles, identify parallels in disease, and speculate whether developmental mechanisms re-emerge in disease and injury. Developmental Dynamics 248:98-117, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sarah R Anderson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, Utah
| | - Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| |
Collapse
|
124
|
Namestnikova DD, Tairova RT, Sukhinich KK, Cherkashova EA, Gubskiy IL, Gubskiy LV, Yarygin KN. [Cell therapy for ischemic stroke. Stem cell types and results of pre-clinical trials]. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:69-75. [PMID: 30499563 DOI: 10.17116/jnevro201811809269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The literature review addresses the use of stem cells (SC) in ischemic stroke (IS). Part 1 of the paper overviews the results of experimental animal studies. Characteristics of different SC types and results of their studies in experimental models of IS are presented in the first section, the second section considers pros and cons of the methods of SC injection.
Collapse
Affiliation(s)
- D D Namestnikova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - R T Tairova
- National Research Institute of Cerebrovascular Pathology and Stroke, Moscow, Russia
| | - K K Sukhinich
- Kol'tsov Institute of Development Biology, Moscow, Russia
| | - E A Cherkashova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - I L Gubskiy
- National Research Institute of Cerebrovascular Pathology and Stroke, Moscow, Russia
| | - L V Gubskiy
- National Research Institute of Cerebrovascular Pathology and Stroke, Moscow, Russia
| | - K N Yarygin
- Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
125
|
Alzoubi KH, Alibbini S, Khabour OF, El-Elimat T, Al-Zubi M, Alali FQ. Carob (Ceratonia siliqua L.) Prevents Short-Term Memory Deficit Induced by Chronic Stress in Rats. J Mol Neurosci 2018; 66:314-321. [PMID: 30218423 DOI: 10.1007/s12031-018-1161-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
Abstract
Long-term exposure to stressful conditions could impair the normal brain structure and function, specifically the hippocampus-dependent memory. This impairment could be attributed to a decrease in brain-derived neurotrophic factor (BDNF) levels during chronic stress. Knowing that carob [Ceratonia siliqua L. (Fabaceae)] is rich in a wide variety of polyphenols with a high antioxidant value, we hypothesized that the methanolic carob extract (C. siliqua) pods will prevent stress-induced memory impairment. Hence, the methanolic extract of carob pods was investigated for its ability to enhance learning and memory as well as to protect from memory impairment in normal stressed animals. Rats were chronically stressed for 7 weeks via the intruder stress model. Carob extract was administered to animals via intraperitoneal (i.p.) route at a daily dose of 50 mg/kg. Radial arm water maze (RAWM) was utilized to test for spatial learning and memory. In addition, brain tissues were dissected to determine BDNF levels. Chronic stress (CS) impaired short-term spatial memory (number of committed errors: P < 0.05, days to criterion (DTC): P < 0.001). Animal treatment with carob pod extract prevented the short-term memory impairment induced by CS (P < 0.05), while such treatment showed no effect on memory functions of unstressed rats. Moreover, carob pod extract prevented the reduction in the hippocampal BDNF (P < 0.05) induced by chronic stress exposure. In conclusion, CS impaired short-term memory function, while methanolic extract of carob pods prevented this impairment, probably as a result of preventing reduction in BDNF levels in the hippocampus.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Sanaa Alibbini
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Tamam El-Elimat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad Al-Zubi
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Feras Q Alali
- College of Pharmacy, Qatar University, Doha, 2713, Qatar.
| |
Collapse
|
126
|
Lin JP, Chen CQ, Huang LE, Li NN, Yang Y, Zhu SM, Yao YX. Dexmedetomidine Attenuates Neuropathic Pain by Inhibiting P2X7R Expression and ERK Phosphorylation in Rats. Exp Neurobiol 2018; 27:267-276. [PMID: 30181689 PMCID: PMC6120967 DOI: 10.5607/en.2018.27.4.267] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 01/24/2023] Open
Abstract
α2-Adrenoceptor agonists attenuate hypersensitivity under neuropathic conditions. However, the mechanisms underlying this attenuation remain largely unknown. In the present study, we explored the potential roles of purinergic receptor 7 (P2X7R)/extracellular signal-regulated kinase (ERK) signaling in the anti-nociceptive effect of dexmedetomidine in a rat model of neuropathic pain induced by chronic constriction injury (CCI) of the sciatic nerve. An animal model of CCI was adopted to mimic the clinical neuropathic pain state. Behavioral hypersensitivity to mechanical and thermal stimuli was determined by von Frey filament and Hargreaves' tests, and the spinal P2X7R expression level and ERK phosphorylation were analyzed using western blot analysis and immunohistochemistry. In parallel with the development of mechanical and thermal hyperalgesia, a significant increase in P2X7R expression was noted in the ipsilateral spinal cord on day 7 after CCI. Intrathecal administration of dexmedetomidine (2.5 µg) for 3 days not only attenuated neuropathic pain but also inhibited the CCI-induced P2X7R upregulation and ERK phosphorylation. Intrathecal dexmedetomidine administration did not produce obvious effects on locomotor function. The present study demonstrated that dexmedetomidine attenuates the neuropathic pain induced by CCI of the sciatic nerve in rats by inhibiting spinal P2X7R expression and ERK phosphorylation, indicating the potential therapeutic implications of dexmedetomidine administration for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Jia-Piao Lin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P.R. China
| | - Chao-Qin Chen
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P.R. China
| | - Ling-Er Huang
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P.R. China
| | - Na-Na Li
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P.R. China
| | - Yan Yang
- Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou 310016, P.R. China
| | - Sheng-Mei Zhu
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P.R. China
| | - Yong-Xing Yao
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P.R. China
| |
Collapse
|