Loading...

Table of Content

    15 June 2021, Volume 16 Issue 6 Previous Issue    Next Issue
    For Selected: Toggle Thumbnails
    Entacapone promotes hippocampal neurogenesis in mice
    Dae Young Yoo, Hyo Young Jung, Woosuk Kim, Kyu Ri Hahn, Hyun Jung Kwon, Sung Min Nam, Jin Young Chung, Yeo Sung Yoon, Dae Won Kim, In Koo Hwang
    2021, 16 (6):  1005-1010.  doi: 10.4103/1673-5374.300447
    Abstract ( 86 )   PDF (4070KB) ( 129 )   Save
    Entacapone, a catechol-O-methyltransferase inhibitor, can strengthen the therapeutic effects of levodopa on the treatment of Parkinson’s disease. However, few studies are reported on whether entacapone can affect hippocampal neurogenesis in mice. To investigate the effects of entacapone, a modulator of dopamine, on proliferating cells and immature neurons in the mouse hippocampal dentate gyrus, 60 mice (7 weeks old) were randomly divided into a vehicle-treated group and the groups treated with 10, 50, or 200 mg/kg entacapone. The results showed that 50 and 200 mg/kg entacapone increased the exploration time for novel object recognition. Immunohistochemical staining results revealed that after entacapone treatment, the numbers of Ki67-positive proliferating cells, doublecortin-positive immature neurons, and phosphorylated cAMP response element-binding protein (pCREB)-positive cells were significantly increased. Western blot analysis results revealed that treatment with tyrosine kinase receptor B (TrkB) receptor antagonist significantly decreased the exploration time for novel object recognition and inhibited the expression of phosphorylated TrkB and brain-derived neurotrophic factor (BDNF). Entacapone treatment antagonized the effects of TrkB receptor antagonist. These results suggest that entacapone treatment promoted hippocampal neurogenesis and improved memory function through activating the BDNF-TrkB-pCREB pathway. This study was approved by the Institutional Animal Care and Use Committee of Seoul National University (approval No. SNU-130730-1) on February 24, 2014.
    Related Articles | Metrics
    Electroacupuncture improves learning and memory functions in a rat cerebral ischemia/reperfusion injury model through PI3K/Akt signaling pathway activation
    Hui-Ling Wang, Fei-Lai Liu, Rui-Qing Li, Ming-Yue Wan, Jie-Ying Li, Jing Shi, Ming-Li Wu, Jun-Hua Chen, Wei-Juan Sun, Hong-Xia Feng, Wei Zhao, Jin Huang, Ren-Chao Liu, Wen-Xue Hao, Xiao-Dong Feng
    2021, 16 (6):  1011-1016.  doi: 10.4103/1673-5374.300454
    Abstract ( 147 )   PDF (965KB) ( 129 )   Save
    Electroacupuncture has been widely used to treat cognitive impairment after cerebral ischemia, but the underlying mechanism has not yet been fully elucidated. Studies have shown that autophagy plays an important role in the formation and development of cognitive impairment, and the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway plays an important role in autophagy regulation. To investigate the role played by the PI3K/Akt signaling pathway in the electroacupuncture treatment of cerebral ischemia/reperfusion rat models, we first established a rat model of cerebral ischemia/reperfusion through the occlusion of the middle cerebral artery using the suture method. Starting at 2 hours after modeling, electroacupuncture was delivered at the Shenting (GV24) and Baihui (GV20) acupoints, with a dilatational wave (1–20 Hz frequency, 2 mA intensity, 6 V peak voltage), for 30 minutes/day over 8 consecutive days. Our results showed that electroacupuncture reduced the infarct volume in a rat model of cerebral ischemia/reperfusion injury, increased the mRNA expression levels of the PI3K/Akt signaling pathway-related factors Beclin-1, mammalian target of rapamycin (mTOR), and PI3K, increased the protein expression levels of phosphorylated Akt, Beclin-1, PI3K, and mTOR in the ischemic cerebral cortex, and simultaneously reduced p53 mRNA and protein expression levels. In the Morris water maze test, the latency to find the hidden platform was significantly shortened among rats subjected to electroacupuncture stimulation compared with rats without electroacupuncture stimulation. In the spatial probe test, the number of times that a rat crossed the target quadrant was increased in rats subjected to electroacupuncture stimulation compared with rats without electroacupuncture stimulation. Electroacupuncture stimulation applied to the Shenting (GV24) and Baihui (GV20) acupoints activated the PI3K/Akt signaling pathway and improved rat learning and memory impairment. This study was approved by the Animal Ethics Committee of the First Affiliated Hospital of Henan University of Traditional Chinese Medicine, China (approval No. 8150150901) on March 10, 2016. 
    Related Articles | Metrics
    Normobaric oxygen therapy attenuates hyperglycolysis in ischemic stroke
    Zhe Cheng, Feng-Wu Li, Christopher R. Stone, Kenneth Elkin, Chang-Ya Peng, Redina Bardhi, Xiao-Kun Geng, Yu-Chuan Ding
    2021, 16 (6):  1017-1023.  doi: 10.4103/1673-5374.300452
    Abstract ( 144 )   PDF (1473KB) ( 107 )   Save
    Normobaric oxygen therapy has gained attention as a simple and convenient means of achieving neuroprotection against the pathogenic cascade initiated by acute ischemic stroke. The mechanisms underlying the neuroprotective efficacy of normobaric oxygen therapy, however, have not been fully elucidated. It is hypothesized that cerebral hyperglycolysis is involved in the neuroprotection of normobaric oxygen therapy against ischemic stroke. In this study, Sprague-Dawley rats were subjected to either 2-hour middle cerebral artery occlusion followed by 3- or 24-hour reperfusion or to a permanent middle cerebral artery occlusion event. At 2 hours after the onset of ischemia, all rats received either 95% oxygen normobaric oxygen therapy for 3 hours or room air. Compared with room air, normobaric oxygen therapy significantly reduced the infarct volume, neurological deficits, and reactive oxygen species and increased the production of adenosine triphosphate in ischemic rats. These changes were associated with reduced transcriptional and translational levels of the hyperglycolytic enzymes glucose transporter 1 and 3, phosphofructokinase 1, and lactate dehydrogenase. In addition, normobaric oxygen therapy significantly reduced adenosine monophosphate-activated protein kinase mRNA expression and phosphorylated adenosine monophosphate-activated protein kinase protein expression. These findings suggest that normobaric oxygen therapy can reduce hyperglycolysis through modulating the adenosine monophosphate-activated protein kinase signaling pathway and alleviating oxidative injury, thereby exhibiting neuroprotective effects in ischemic stroke. This study was approved by the Institutional Animal Investigation Committee of Capital Medical University (approval No. AEEI-2018-033) on August 13, 2018.  
    Related Articles | Metrics
    MicroRNA-670 aggravates cerebral ischemia/reperfusion injury via the Yap pathway 
    Shi-Jia Yu, Ming-Jun Yu, Zhong-Qi Bu, Ping-Ping He, Juan Feng
    2021, 16 (6):  1024-1030.  doi: 10.4103/1673-5374.300455
    Abstract ( 122 )   PDF (2130KB) ( 110 )   Save
    Apoptosis is an important programmed cell death process involved in ischemia/reperfusion injury. MicroRNAs are considered to play an important role in the molecular mechanism underlying the regulation of cerebral ischemia and reperfusion injury. However, whether miR-670 can regulate cell growth and death in cerebral ischemia/reperfusion and the underlying mechanism are poorly understood. In this study, we established mouse models of transient middle artery occlusion and Neuro 2a cell models of oxygen-glucose deprivation and reoxygenation to investigate the potential molecular mechanism by which miR-670 exhibits its effects during cerebral ischemia/reperfusion injury both in vitro and in vivo. Our results showed that after ischemia/reperfusion injury, miR-670 expression was obviously increased. After miR-670 expression was inhibited with an miR-670 antagomir, cerebral ischemia/reperfusion injury-induced neuronal death was obviously reduced. When miR-670 overexpression was induced by an miR-670 agomir, neuronal apoptosis was increased. In addition, we also found that miR-670 could promote Yap degradation via phosphorylation and worsen neuronal apoptosis and neurological deficits. Inhibition of miR-670 reduced neurological impairments after cerebral ischemia/reperfusion injury. These results suggest that microRNA-670 aggravates cerebral ischemia/reperfusion injury through the Yap pathway, which may be a potential target for treatment of cerebral ischemia/reperfusion injury. The present study was approved by the Institutional Animal Care and Use Committee of China Medical University on February 27, 2017 (IRB No. 2017PS035K).
    Related Articles | Metrics
    Corticospinal excitability during motor imagery is diminished by continuous repetition-induced fatigue
    Akira Nakashima, Takefumi Moriuchi, Daiki Matsuda, Takashi Hasegawa, Jirou Nakamura, Kimika Anan, Katsuya Satoh, Tomotaka Suzuki, Toshio Higashi, Kenichi Sugawara
    2021, 16 (6):  1031-1036.  doi: 10.4103/1673-5374.300448
    Abstract ( 130 )   PDF (471KB) ( 98 )   Save
    Application of continuous repetition of motor imagery can improve the performance of exercise tasks. However, there is a lack of more detailed neurophysiological evidence to support the formulation of clear standards for interventions using motor imagery. Moreover, identification of motor imagery intervention time is necessary because it exhibits possible central fatigue. Therefore, the purpose of this study was to elucidate the development of fatigue during continuous repetition of motor imagery through objective and subjective evaluation. The study involved two experiments. In experiment 1, 14 healthy young volunteers were required to imagine grasping and lifting a 1.5-L plastic bottle using the whole hand. Each participant performed the motor imagery task 100 times under each condition with 48 hours interval between two conditions: 500 mL or 1500 mL of water in the bottle during the demonstration phase. Mental fatigue and a decrease in pinch power appeared under the 1500-mL condition. There were changes in concentration ability or corticospinal excitability, as assessed by motor evoked potentials, between each set with continuous repetition of motor imagery also under the 1500-mL condition. Therefore, in experiment 2, 12 healthy volunteers were required to perform the motor imagery task 200 times under the 1500-mL condition. Both concentration ability and corticospinal excitability decreased. This is the first study to show that continuous repetition of motor imagery can decrease corticospinal excitability in addition to producing mental fatigue. This study was approved by the Institutional Ethics Committee at the Nagasaki University Graduate School of Biomedical and Health Sciences (approval No. 18121302) on January 30, 2019.
    Related Articles | Metrics
    TP53-induced glycolysis and apoptosis regulator alleviates hypoxia/ischemia-induced microglial pyroptosis and ischemic brain damage
    Lan-Lan Tan, Xiao-Lu Jiang, Li-Xiao Xu, Gen Li, Chen-Xi Feng, Xin Ding, Bin Sun, Zheng-Hong Qin, Zu-Bin Zhang, Xing Feng, Mei Li
    2021, 16 (6):  1037-1043.  doi: 10.4103/1673-5374.300453
    Abstract ( 264 )   PDF (3454KB) ( 236 )   Save
    Our previous studies have demonstrated that TP53-induced glycolysis and apoptosis regulator (TIGAR) can protect neurons after cerebral ischemia/reperfusion. However, the role of TIGAR in neonatal hypoxic-ischemic brain damage (HIBD) remains unknown. In the present study, 7-day-old Sprague-Dawley rat models of HIBD were established by permanent occlusion of the left common carotid artery followed by 2-hour hypoxia. At 6 days before induction of HIBD, a lentiviral vector containing short hairpin RNA of either TIGAR or gasdermin D (LV-sh_TIGAR or LV-sh_GSDMD) was injected into the left lateral ventricle and striatum. Highly aggressively proliferating immortalized (HAPI) microglial cell models of in vitro HIBD were established by 2-hour oxygen/glucose deprivation followed by 24-hour reoxygenation. Three days before in vitro HIBD induction, HAPI microglial cells were transfected with LV-sh_TIGAR or LV-sh_GSDMD. Our results showed that TIGAR expression was increased in the neonatal rat cortex after HIBD and in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Lentivirus-mediated TIGAR knockdown in rats markedly worsened pyroptosis and brain damage after hypoxia/ischemia in vivo and in vitro. Application of exogenous nicotinamide adenine dinucleotide phosphate (NADPH) increased the NADPH level and the glutathione/oxidized glutathione ratio and decreased reactive oxygen species levels in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Additionally, exogenous NADPH blocked the effects of TIGAR knockdown in neonatal HIBD in vivo and in vitro. These findings show that TIGAR can inhibit microglial pyroptosis and play a protective role in neonatal HIBD. The study was approved by the Animal Ethics Committee of Soochow University of China (approval No. 2017LW003) in 2017. 
    Related Articles | Metrics
    Apelin-13 inhibits apoptosis and excessive autophagy in cerebral ischemia/reperfusion injury
    Zi-Qi Shao, Shan-Shan Dou, Jun-Ge Zhu, Hui-Qing Wang, Chun-Mei Wang, Bao-Hua Cheng, Bo Bai
    2021, 16 (6):  1044-1051.  doi: 10.4103/1673-5374.300725
    Abstract ( 154 )   PDF (4557KB) ( 480 )   Save
    Apelin-13 is a novel endogenous ligand for an angiotensin-like orphan G-protein coupled receptor, and it may be neuroprotective against cerebral ischemia injury. However, the precise mechanisms of the effects of apelin-13 remain to be elucidated. To investigate the effects of apelin-13 on apoptosis and autophagy in models of cerebral ischemia/reperfusion injury, a rat model was established by middle cerebral artery occlusion. Apelin-13 (50 µg/kg) was injected into the right ventricle as a treatment. In addition, an SH-SY5Y cell model was established by oxygen-glucose deprivation/reperfusion, with cells first cultured in sugar-free medium with 95% N2 and 5% CO2 for 4 hours and then cultured in a normal environment with sugar-containing medium for 5 hours. This SH-SY5Y cell model was treated with 10–7 M apelin-13 for 5 hours. Results showed that apelin-13 protected against cerebral ischemia/reperfusion injury. Apelin-13 treatment alleviated neuronal apoptosis by increasing the ratio of Bcl-2/Bax and significantly decreasing cleaved caspase-3 expression. In addition, apelin-13 significantly inhibited excessive autophagy by regulating the expression of LC3B, p62, and Beclin1. Furthermore, the expression of Bcl-2 and the phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway was markedly increased. Both LY294002 
    (20 µM) and rapamycin (500 nM), which are inhibitors of the PI3K/Akt/mTOR pathway, significantly attenuated the inhibition of autophagy and apoptosis caused by apelin-13. In conclusion, the findings of the present study suggest that Bcl-2 upregulation and mTOR signaling pathway activation lead to the inhibition of apoptosis and excessive autophagy. These effects are involved in apelin-13-induced neuroprotection against cerebral ischemia/reperfusion injury, both in vivo and in vitro. The study was approved by the Animal Ethical and Welfare Committee of Jining Medical University, China (approval No. 2018-JS-001) in February 2018.  
    Related Articles | Metrics
    Brief inhalation of sevoflurane can reduce glial scar formation after hypoxic-ischemic brain injury in neonatal rats
    Qiu-Shi Gao, Ya-Han Zhang, Hang Xue, Zi-Yi Wu, Chang Li, Ping Zhao
    2021, 16 (6):  1052-1061.  doi: 10.4103/1673-5374.300456
    Abstract ( 110 )   PDF (6993KB) ( 24 )   Save
    Previous studies have demonstrated that sevoflurane postconditioning can provide neuroprotection after hypoxic-ischemic injury and improve learning and memory function in developing rodent brains. The classical Rice-Vannucci model was used to induce hypoxic-ischemic injury, and newborn (postnatal day 7) rats were treated with 2.4% sevoflurane for 30 minutes after hypoxic-ischemic injury. Our results showed that sevoflurane postconditioning significantly improved the learning and memory function of rats, decreased astrogliosis and glial scar formation, increased numbers of dendritic spines, and protected the histomorphology of the hippocampus. Mechanistically, sevoflurane postconditioning decreased expression of von Hippel-Lindau of hypoxia-inducible factor-1α and increased expression of DJ-1. Injection of 1.52 μg of the hypoxia-inducible factor-1α inhibitor YC-1 (Lificiguat) into the left lateral ventricle 30 minutes before hypoxic-ischemic injury reversed the neuroprotection induced by sevoflurane. This finding suggests that sevoflurane can effectively alleviate astrogliosis in the hippocampus and reduce learning and memory impairments caused by glial scar formation after hypoxic-ischemic injury. The underlying mechanism may be related to upregulated DJ-1 expression, reduced ubiquitination of hypoxia-inducible factor-1α, and stabilized hypoxia-inducible factor-1α expression. This study was approved by the Laboratory Animal Care Committee of China Medical University, China (approval No. 2016PS337K) on November 9, 2016.
    Related Articles | Metrics
    Astaxanthin alleviates pathological brain aging through the upregulation of hippocampal synaptic proteins
    Ning Liu, Liang Zeng, Yi-Ming Zhang, Wang Pan, Hong Lai
    2021, 16 (6):  1062-1067.  doi: 10.4103/1673-5374.300460
    Abstract ( 134 )   PDF (6687KB) ( 203 )   Save
    Oxidative stress is currently considered to be the main cause of brain aging. Astaxanthin can improve oxidative stress under multiple pathological conditions. It is therefore hypothesized that astaxanthin might have therapeutic effects on brain aging. To validate this hypothesis and investigate the underlying mechanisms, a mouse model of brain aging was established by injecting amyloid beta (Aβ)25–35 (5 μM, 
    3 μL/injection, six injections given every other day) into the right lateral ventricle. After 3 days of Aβ25–35 injections, the mouse models were intragastrically administered astaxanthin (0.1 mL/d, 10 mg/kg) for 30 successive days. Astaxanthin greatly reduced the latency to find the platform in the Morris water maze, increased the number of crossings of the target platform, and increased the expression of brain-derived neurotrophic factor, synaptophysin, sirtuin 1, and peroxisome proliferator-activated receptor-γ coactivator 1α. Intraperitoneal injection of the sirtuin 1 inhibitor nicotinamide (500 μM/d) for 7 successive days after astaxanthin intervention inhibited these phenomena. These findings suggest that astaxanthin can regulate the expression of synaptic proteins in mouse hippocampus through the sirtuin 1/peroxisome proliferator-activated receptor-γ coactivator 1α signaling pathway, which leads to improvements in the learning, cognitive, and memory abilities of mice. The study was approved by the Animal Ethics Committee, China Medical University, China (approval No. CMU2019294) on January 15, 2019.  
    Related Articles | Metrics
    Collagen/heparan sulfate porous scaffolds loaded with neural stem cells improve neurological function in a rat model of traumatic brain injury
    Jian Zhang, Ren-Jie Wang, Miao Chen, Xiao-Yin Liu, Ke Ma, Hui-You Xu, Wu-Sheng Deng, Yi-Chao Ye, Wei-Xin Li, Xu-Yi Chen, Hong-Tao Sun
    2021, 16 (6):  1068-1077.  doi: 10.4103/1673-5374.300458
    Abstract ( 130 )   PDF (5177KB) ( 160 )   Save
    One reason for the poor therapeutic effects of stem cell transplantation in traumatic brain injury is that exogenous neural stem cells cannot effectively migrate to the local injury site, resulting in poor adhesion and proliferation of neural stem cells at the injured area. To enhance the targeted delivery of exogenous stem cells to the injury site, cell therapy combined with neural tissue engineering technology is expected to become a new strategy for treating traumatic brain injury. Collagen/heparan sulfate porous scaffolds, prepared using a freeze-drying method, have stable physical and chemical properties. These scaffolds also have good cell biocompatibility because of their high porosity, which is suitable for the proliferation and migration of neural stem cells. In the present study, collagen/heparan sulfate porous scaffolds loaded with neural stem cells were used to treat a rat model of traumatic brain injury, which was established using the controlled cortical impact method. At 2 months after the implantation of collagen/heparan sulfate porous scaffolds loaded with neural stem cells, there was significantly improved regeneration of neurons, nerve fibers, synapses, and myelin sheaths in the injured brain tissue. Furthermore, brain edema and cell apoptosis were significantly reduced, and rat motor and cognitive functions were markedly recovered. These findings suggest that the novel collagen/heparan sulfate porous scaffold loaded with neural stem cells can improve neurological function in a rat model of traumatic brain injury. This study was approved by the Institutional Ethics Committee of Characteristic Medical Center of Chinese People’s Armed Police Force, China (approval No. 2017-0007.2) on February 10, 2019.
    Related Articles | Metrics
    Ascorbic acid accelerates Wallerian degeneration after peripheral nerve injury
    Lixia Li, Yizhou Xu, Xianghai Wang, Jingmin Liu, Xiaofang Hu, Dandan Tan, Zhenlin Li, Jiasong Guo
    2021, 16 (6):  1078-1085.  doi: 10.4103/1673-5374.300459
    Abstract ( 131 )   PDF (5311KB) ( 296 )   Save
    Wallerian degeneration occurs after peripheral nerve injury and provides a beneficial microenvironment for nerve regeneration. Our previous study demonstrated that ascorbic acid promotes peripheral nerve regeneration, possibly through promoting Schwann cell proliferation and phagocytosis and enhancing macrophage proliferation, migration, and phagocytosis. Because Schwann cells and macrophages are the main cells involved in Wallerian degeneration, we speculated that ascorbic acid may accelerate this degenerative process. To test this hypothesis, 400 mg/kg ascorbic acid was administered intragastrically immediately after sciatic nerve transection, and 200 mg/kg ascorbic acid was then administered intragastrically every day. In addition, rat sciatic nerve explants were treated with 200 μM ascorbic acid. Ascorbic acid significantly accelerated the degradation of myelin basic protein-positive myelin and neurofilament 200-positive axons in both the transected nerves and nerve explants. Furthermore, ascorbic acid inhibited myelin-associated glycoprotein expression, increased c-Jun expression in Schwann cells, and increased both the number of macrophages and the amount of myelin fragments in the macrophages. These findings suggest that ascorbic acid accelerates Wallerian degeneration by accelerating the degeneration of axons and myelin in the injured nerve, promoting the dedifferentiation of Schwann cells, and enhancing macrophage recruitment and phagocytosis. The study was approved by the Southern Medical University Animal Care and Use Committee (approval No. SMU-L2015081) on October 15, 2015. 
    Related Articles | Metrics
    Decellularized peripheral nerve grafts by a modified protocol for repair of rat sciatic nerve injury
    Arash Zaminy, Sara Sayad-Fathi, Farshad Moharrami Kasmaie, Zohreh Jahromi, Adib Zendedel
    2021, 16 (6):  1086-1092.  doi: 10.4103/1673-5374.300449
    Abstract ( 106 )   PDF (3830KB) ( 119 )   Save
    Studies have shown that acellular nerve xenografts do not require immunosuppression and use of acellular nerve xenografts for repair of peripheral nerve injury is safe and effective. However, there is currently no widely accepted standard chemical decellularization method. The purpose of this study is to investigate the efficiency of bovine-derived nerves decellularized by the modified Hudson’s protocol in the repair of rat sciatic nerve injury. In the modified Hudson’s protocol, Triton X-200 was replaced by Triton X-100, and DNase and RNase were used to prepare accelular nerve xenografts. The efficiency of bovine-derived nerves decellularized by the modified Hudson’s protocol was tested in vitro by hematoxylin & eosin, Alcian blue, Masson’s trichrome, and Luxol fast blue staining, immunohistochemistry, and biochemical assays. The decellularization approach excluded cells, myelin, and axons of nerve xenografts, without affecting the organization of nerve xenografts. The decellularized nerve xenograft was used to bridge a 7 mm-long sciatic nerve defect to evaluate its efficiency in the repair of peripheral nerve injury. At 8 weeks after transplantation, sciatic function index in rats subjected to transplantation of acellular nerve xenograft was similar to that in rats undergoing transplantation of nerve allograft. Morphological analysis revealed that there were a large amount of regenerated myelinated axons in acellular nerve xenograft; the number of Schwann cells in the acellular nerve xenograft was similar to that in the nerve allograft. These findings suggest that acellular nerve xenografts prepared by the modified Hudson’s protocol can be used for repair of peripheral nerve injury. This study was approved by the Research Ethics Committee, Research and Technology Chancellor of Guilan  University of Medical Sciences, Iran (approval No. IR.GUMS.REC.1395.332) on February 11, 2017.
    Related Articles | Metrics
    Production of chitosan scaffolds by lyophilization or electrospinning: which is better for peripheral nerve regeneration?
    Yu-Xuan Wu, Hao Ma, Jian-Lan Wang, Wei Qu
    2021, 16 (6):  1093-1098.  doi: 10.4103/1673-5374.300463
    Abstract ( 147 )   PDF (1524KB) ( 199 )   Save
    Both lyophilization and electrospinning are commonly used to make chitosan scaffolds. However, it remains unknown which method is better for cell growth. In this study, we established the following groups: (1) lyophilization group—chitosan scaffolds were prepared by lyophilization method and seeded with Schwann cells from Sprague-Dawley rats aged 3–5 days; (2) electrospinning group—chitosan scaffolds were prepared by electrospinning method and seeded with Schwann cells; (3) control group—Schwann cells were cultured on culture dishes. The growth of Schwann cells was evaluated by immunofluorescence and scanning electron microscopy. Western blot assay was performed to explore the mechanism of Schwann cell growth. Both materials were non-toxic and suitable for the growth of Schwann cells. The pores produced by electrospinning were much smaller than those produced by lyophilization. The proliferation rate and adhesion rate of Schwann cells in the electrospinning group were higher than those in the lyophilization group. Schwann cells cultured on electrospinning scaffolds formed a Bungner band-like structure, and a much greater amount of brain-derived neurotrophic factor was secreted, which can promote the growth of neurons. Our findings show that the chitosan scaffold prepared by the electrospinning method has a nanofiber structure that provides an extracellular matrix that is more favorable for cell-cell interactions. The electrospinning method is more suitable for nerve regeneration than the lyophilization method. This research was approved by the Medical Ethical Committee of Dalian Medical University (approval No. AEE1-2016-045) on March 3, 2016.
    Related Articles | Metrics
    Neuroprotective and anti-inflammatory effects of a therapy combining agonists of nicotinic α7 and σ1 receptors in a rat model of Parkinson’s disease
    Steven Vetel, Laura Foucault-Fruchard, Claire Tronel, Frédéric Buron, Jackie Vergote, Sylvie Bodard, Sylvain Routier, Sophie Sérrière, Sylvie Chalon
    2021, 16 (6):  1099-1104.  doi: 10.4103/1673-5374.300451
    Abstract ( 289 )   PDF (1263KB) ( 136 )   Save
    To date there is no treatment able to stop or slow down the loss of dopaminergic neurons that characterizes Parkinson’s disease. It was recently observed in a rodent model of Alzheimer’s disease that the interaction between the α7 subtype of nicotinic acetylcholine receptor (α7-nAChR) and sigma-1 receptor (σ1-R) could exert neuroprotective effects through the modulation of neuroinflammation which is one of the key components of the pathophysiology of Parkinson’s disease. In this context, the aim of the present study was to assess the effects of the concomitant administration of N-(3R)-1-azabicyclo[2.2.2]oct-3-yl-furo[2,3-c]pyridine-5-carboxamide (PHA) 543613 as an α7-nAChR agonist and 2-(4-morpholinethyl) 1-phenylcyclohexanecarboxylate (PRE)-084 as a σ1-R agonist in a well-characterized 6-hydroxydopamine rat model of Parkinson’s disease. The animals received either vehicle separately or the dual therapy PHA/PRE once a day until day 14 post-lesion. Although no effect was noticed in the amphetamine-induced rotation test, our data has shown that the PHA/PRE treatment induced partial protection of the dopaminergic neurons (15–20%), assessed by the dopamine transporter density in the striatum and immunoreactive tyrosine hydroxylase in the substantia nigra. Furthermore, this dual therapy reduced the degree of glial activation consecutive to the 6-hydroxydopamine lesion, i.e, the 18 kDa translocation protein density and glial fibrillary acidic protein staining in the striatum, and the CD11b and glial fibrillary acidic protein staining in the substantia nigra. Hence, this study reports for the first time that concomitant activation of α7-nAChR and σ1-R can provide a partial recovery of the nigro-striatal dopaminergic neurons through the modulation of microglial activation. The study was approved by the Regional Ethics Committee (CEEA Val de Loire n°19) validated this protocol (Authorization N°00434.02) on May 15, 2014.
    Related Articles | Metrics
    Myricetin reduces cytotoxicity by suppressing hepcidin expression in MES23.5 cells
    Han Deng, Shang Liu, Dong Pan, Yi Jia, Ze-Gang Ma
    2021, 16 (6):  1105-1110.  doi: 10.4103/1673-5374.300461
    Abstract ( 114 )   PDF (1485KB) ( 111 )   Save
    Multiple studies implicate iron accumulation in the substantia nigra in the degeneration of dopaminergic neurons in Parkinson’s disease. Indeed, slowing of iron accumulation in cells has been identified as the key point for delaying and treating Parkinson’s disease. Myricetin reportedly plays an important role in anti-oxidation, anti-apoptosis, anti-inflammation, and iron chelation. However, the mechanism underlying its neuroprotection remains unclear. In the present study, MES23.5 cells were treated with 1 × 10–6 M myricetin for 1 hour, followed by co-treatment with 400 nM rotenone for 24 hours to establish an in vitro cell model of Parkinson’s disease. Our results revealed that myricetin alleviated rotenone-induced decreases in cell viability, suppressed the production of intracellular reactive oxygen species, and restored mitochondrial transmembrane potential. In addition, myricetin significantly suppressed rotenone-induced hepcidin gene transcription and partly relieved rotenone-induced inhibition of ferroportin 1 mRNA and protein levels. Furthermore, myricetin inhibited rotenone-induced phosphorylation of STAT3 and SMAD1 in MES23.5 cells. These findings suggest that myricetin protected rotenone-treated MES23.5 cells by potently inhibiting hepcidin expression to prevent iron accumulation, and this effect was mediated by alteration of STAT3 and SMAD1 signaling pathways. 
    Related Articles | Metrics
    A network-based cognitive training induces cognitive improvements and neuroplastic changes in patients with relapsing-remitting multiple sclerosis: #br# an exploratory case-control study#br#
    Riccardo Manca, Micaela Mitolo, Iain D. Wilkinson, David Paling, Basil Sharrack, Annalena Venneri
    2021, 16 (6):  1111-1120.  doi: 10.4103/1673-5374.300450
    Abstract ( 101 )   PDF (1952KB) ( 112 )   Save
    Cognitive impairments are commonly observed in patients with multiple sclerosis and are associated with lower levels of quality of life. No consensus has been reached on how to tackle effectively cognitive decline in this clinical population non-pharmacologically. This exploratory case-control study aims to investigate the effectiveness of a hypothesis-based cognitive training designed to target multiple domains by promoting the synchronous co-activation of different brain areas and thereby improve cognition and induce changes in functional connectivity in patients with relapsing-remitting multiple sclerosis. Forty-five patients (36 females and 9 males, mean age 44.62 ± 8.80 years) with clinically stable relapsing-remitting multiple sclerosis were assigned to either a standard cognitive training or to control groups (sham training and non-active control). The standard training included twenty sessions of computerized exercises involving various cognitive functions supported by distinct brain networks. The sham training was a modified version of the standard training that comprised the same exercises and number of sessions but with increased processing speed load. The non-active control group received no cognitive training. All patients underwent comprehensive neuropsychological and magnetic resonance imaging assessments at baseline and after 5 weeks. Cognitive and resting-state magnetic resonance imaging data were analyzed using repeated measures models. At reassessment, the standard training group showed significant cognitive improvements compared to both control groups in memory tasks not specifically targeted by the training: the Buschke Selective Reminding Test and the Semantic Fluency test. The standard training group showed reductions in functional connectivity of the salience network, in the anterior cingulate cortex, associated with improvements on the Buschke Selective Reminding Test. No changes were observed in the sham training group. These findings suggest that multi-domain training that stimulates multiple brain areas synchronously may improve cognition in people with relapsing-remitting multiple sclerosis if sufficient time to process training material is allowed. The associated reduction in functional connectivity of the salience network suggests that training-induced neuroplastic functional reorganization may be the mechanism supporting performance gains. This study was approved by the Regional Ethics Committee of Yorkshire and Humber (approval No. 12/YH/0474) on November 20, 2013.
    Related Articles | Metrics
    Peritoneal macrophages attenuate retinal ganglion cell survival and neurite outgrowth
    Jia-Jian Liang, Yu-Fen Liu, Tsz Kin Ng, Ci-Yan Xu, Mingzhi Zhang, Chi Pui Pang, Ling-Ping Cen
    2021, 16 (6):  1121-1126.  doi: 10.4103/1673-5374.300462
    Abstract ( 129 )   PDF (2127KB) ( 99 )   Save
    Inflammation is a critical pathophysiological process that modulates neuronal survival in the central nervous system after disease or injury. However, the effects and mechanisms of macrophage activation on neuronal survival remain unclear. In the present study, we co-cultured adult Fischer rat retinas with primary peritoneal macrophages or zymosan-treated peritoneal macrophages for 7 days. Immunofluorescence analysis revealed that peritoneal macrophages reduced retinal ganglion cell survival and neurite outgrowth in the retinal explant compared with the control group. The addition of zymosan to peritoneal macrophages attenuated the survival and neurite outgrowth of retinal ganglion cells. Conditioned media from peritoneal macrophages also reduced retinal ganglion cell survival and neurite outgrowth. This result suggests that secretions from peritoneal macrophages mediate the inhibitory effects of these macrophages. In addition, increased inflammation- and oxidation-related gene expression may be related to the enhanced retinal ganglion cell degeneration caused by zymosan activation. In summary, this study revealed that primary rat peritoneal macrophages attenuated retinal ganglion cell survival and neurite outgrowth, and that macrophage activation further aggravated retinal ganglion cell degeneration. This study was approved by the Animal Ethics Committee of the Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou, Guangdong Province, China, on March 11, 2014 (approval no. EC20140311(2)-P01).
    Related Articles | Metrics
    Islet amyloid polypeptide & amyloid beta peptide roles in Alzheimer’s disease: two triggers, one disease
    Sofia Ferreira, Ana F. Raimundo, Regina Menezes, Ivo C. Martins
    2021, 16 (6):  1127-1130.  doi: 10.4103/1673-5374.300323
    Abstract ( 84 )   PDF (922KB) ( 55 )   Save
    Alzheimer’s disease (AD) is a neurodegenerative disorder that affects millions worldwide. Due to population ageing, the incidence of AD is increasing. AD patients develop cognitive decline and dementia, features for which is known, requiring permanent care. This poses a major socio-economic burden on healthcare systems as AD patients’ relatives and healthcare workers are forced to cope with rising numbers of affected people. Despite recent advances, AD pathological mechanisms are not fully understood. Nevertheless, it is clear that the amyloid beta (Aβ) peptide, which forms amyloid plaques in AD patients’ brains, plays a key role. Type 2 diabetes, the most common form of diabetes, affects hundreds of million people globally. Islet amyloid polypeptide (IAPP) is a hormone co-produced and secreted with insulin in pancreatic β-cells, with a key role in diabetes, as it helps regulate glucose levels and control adiposity and satiation. Similarly to Aβ, IAPP is very amyloidogenic, generating intracellular amyloid deposits that cause β-cell dysfunction and death. It is now clear that IAPP can also have a pathological role in AD, decreasing cognitive function. IAPP harms the blood-brain barrier, directly interacts and co-deposits with Aβ, promoting diabetes-associated dementia. IAPP can cause a metabolic dysfunction in the brain, leading to other diabetes-related forms of AD. Thus, here we discuss IAPP association with diabetes, Aβ and dementia, in the context of what we designate a “diabetes brain phenotype” AD hypothesis. Such approach helps to set a conceptual framework for future IAPP-based drugs against AD. 
    Related Articles | Metrics
    The Yin-Yang of osteopontin in nervous system diseases: damage versus repair
    Giuseppe Cappellano, Domizia Vecchio, Luca Magistrelli, Nausicaa Clemente, Davide Raineri, Camilla Barbero Mazzucca, Eleonora Virgilio, Umberto Dianzani, Annalisa Chiocchetti, Cristoforo Comi
    2021, 16 (6):  1131-1137.  doi: 10.4103/1673-5374.300328
    Abstract ( 203 )   PDF (778KB) ( 174 )   Save
    Osteopontin is a broadly expressed pleiotropic protein, and is attracting increased attention because of its role in the pathophysiology of several inflammatory, degenerative, autoimmune, and oncologic diseases. In fact, in the last decade, several studies have shown that osteopontin contributes to tissue damage not only by recruiting harmful inflammatory cells to the site of lesion, but also increasing their survival. The detrimental role of osteopontin has been indeed well documented in the context of different neurological conditions (i.e., multiple sclerosis, Parkinson’s, and Alzheimer’s diseases). Intriguingly, recent findings show that osteopontin is involved not only in promoting tissue damage (the Yin), but also in repair/regenerative mechanisms (the Yang), mostly triggered by the inflammatory response. These two apparently discordant roles are partly related to the presence of different functional domains in the osteopontin molecule, which are exposed after thrombin or metalloproteases cleavages. Such functional domains may in turn activate intracellular signaling pathways and mediate cell-cell and cell-matrix interactions. This review describes the current knowledge on the Yin and Yang features of osteopontin in nervous system diseases. Understanding the mechanisms behind the Yin/Yang would be relevant to develop highly specific tools targeting this multifunctional protein.
    Related Articles | Metrics
    Cell transplantation to replace retinal ganglion cells faces challenges – the Switchboard Dilemma
    Yuan Liu, Richard K. Lee
    2021, 16 (6):  1138-1141.  doi: 10.4103/1673-5374.300329
    Abstract ( 83 )   PDF (564KB) ( 111 )   Save
    The mammalian retina displays incomplete intrinsic regenerative capacities; therefore, retina degeneration is a major cause of irreversible blindness such as glaucoma, age-related macular degeneration and diabetic retinopathy. These diseases lead to the loss of retinal cells and serious vision loss in the late stage. Stem cell transplantation is a great promising novel treatment for these incurable retinal degenerative diseases and represents an exciting area of regenerative neurotherapy. Several suitable stem cell sources for transplantation including human embryonic stem cells, induced pluripotent stem cells and adult stem cells have been identified as promising target populations. However, the retina is an elegant neuronal complex composed of various types of cells with different functions. The replacement of these different types of cells by transplantation should be addressed separately. So far, retinal pigment epithelium transplantation has achieved the most advanced stage of clinical trials, while transplantation of retinal neurons such as retinal ganglion cells and photoreceptors has been mostly studied in pre-clinical animal models. In this review, we opine on the key problems that need to be addressed before stem cells transplantation, especially for replacing injured retinal ganglion cells, may be used practically for treatment. A key problem we have called the Switchboard Dilemma is a major block to have functional retinal ganglion cell replacement. We use the public switchboard telephone network as an example to illustrate different difficulties for replacing damaged components in the retina that allow for visual signaling. Retinal ganglion cell transplantation is confronted by significant hurdles, because retinal ganglion cells receive signals from different interneurons, integrate and send signals to the correct targets of the visual system, which functions similar to the switchboard in a telephone network – therefore the Switchboard Dilemma. 
    Related Articles | Metrics
    Motor tract reorganization after acute central nervous system injury: a translational perspective
    Hajime Takase, Robert W. Regenhardt
    2021, 16 (6):  1144-1149.  doi: 10.4103/1673-5374.300330
    Abstract ( 93 )   PDF (662KB) ( 136 )   Save
    Acute central nervous system injuries are among the most common causes of disability worldwide, with widespread social and economic implications. Motor tract injury accounts for the majority of this disability; therefore, there is impetus to understand mechanisms underlying the pathophysiology of injury and subsequent reorganization of the motor tract that may lead to recovery. After acute central nervous system injury, there are changes in the microenvironment and structure of the motor tract. For example, ischemic stroke involves decreased local blood flow and tissue death from lack of oxygen and nutrients. Traumatic injury, in contrast, causes stretching and shearing injury to microstructures, including myelinated axons and their surrounding vessels. Both involve blood-brain barrier dysfunction, which is an important initial event. After acute central nervous system injury, motor tract reorganization occurs in the form of cortical remapping in the gray matter and axonal regeneration and rewiring in the white matter. Cortical remapping involves one cortical region taking on the role of another. cAMP-response-element binding protein is a key transcription factor that can enhance plasticity in the peri-infarct cortex. Axonal regeneration and rewiring depend on complex cell-cell interactions between axons, oligodendrocytes, and other cells. The RhoA/Rho-associated coiled-coil containing kinase signaling pathway plays a central role in axon growth/regeneration through interactions with myelin-derived axonal growth inhibitors and regulation of actin cytoskeletal dynamics. Oligodendrocytes and their precursors play a role in myelination, and neurons are involved through their voltage-gated calcium channels. Understanding the pathophysiology of injury and the biology of motor tract reorganization may allow the development of therapies to enhance recovery after acute central nervous system injury. These include targeted rehabilitation, novel pharmacotherapies, such as growth factors and axonal growth inhibitor blockade, and the implementation of neurotechnologies, such as central nervous system stimulators and robotics. The translation of these advances depends on careful alignment of preclinical studies and human clinical trials. As experimental data mount, the future is one of optimism.
    Related Articles | Metrics
    Functional repertoire of protein kinases and phosphatases in synaptic plasticity and associated neurological disorders
    Raheel Khan, Don Kulasiri, Sandhya Samarasinghe
    2021, 16 (6):  1150-1157.  doi: 10.4103/1673-5374.300331
    Abstract ( 119 )   PDF (1388KB) ( 107 )   Save
    Protein phosphorylation and dephosphorylation are two essential and vital cellular mechanisms that regulate many receptors and enzymes through kinases and phosphatases. Ca2+- dependent kinases and phosphatases are responsible for controlling neuronal processing; balance is achieved through opposition. During molecular mechanisms of learning and memory, kinases generally modulate positively while phosphatases modulate negatively. This review outlines some of the critical physiological and structural aspects of kinases and phosphatases involved in maintaining postsynaptic structural plasticity. It also explores the link between neuronal disorders and the deregulation of phosphatases and kinases.
    Related Articles | Metrics
    Ultrasonic vocalizations in mice: relevance for ethologic and neurodevelopmental disorders studies 
    Marika Premoli, Maurizio Memo, Sara Anna Bonini
    2021, 16 (6):  1158-1167.  doi: 10.4103/1673-5374.300340
    Abstract ( 324 )   PDF (1027KB) ( 160 )   Save
    Mice use ultrasonic vocalizations (USVs) to communicate each other and to convey their emotional state. USVs have been greatly characterized in specific life phases and contexts, such as mother isolation-induced USVs for pups or female-induced USVs for male mice during courtship. USVs can be acquired by means of specific tools and later analyzed on the base of both quantitative and qualitative parameters. Indeed, different ultrasonic call categories exist and have already been defined. The understanding of different calls meaning is still missing, and it will represent an essential step forward in the field of USVs. They have long been studied in the ethological context, but recently they emerged as a precious instrument to study pathologies characterized by deficits in communication, in particular neurodevelopmental disorders (NDDs), such as autism spectrum disorders. This review covers the topics of USVs characteristics in mice, contexts for USVs emission and factors that modulate their expression. A particular focus will be devoted to mouse USVs in the context of NDDs. Indeed, several NDDs murine models exist and an intense study of USVs is currently in progress, with the aim of both performing an early diagnosis and to find a pharmacological/behavioral intervention to improve patients’ quality of life. 
    Related Articles | Metrics
    Repetitive transcranial magnetic stimulation for lower extremity motor function in patients with stroke: a systematic review and network meta-analysis
    Yun-Juan Xie, Yi Chen, Hui-Xin Tan, Qi-Fan Guo, Benson Wui-Man Lau, Qiang Gao
    2021, 16 (6):  1168-1176.  doi: 10.4103/1673-5374.300341
    Abstract ( 139 )   PDF (659KB) ( 166 )   Save
    Transcranial magnetic stimulation, a type of noninvasive brain stimulation, has become an ancillary therapy for motor function rehabilitation. Most previous studies have focused on the effects of repetitive transcranial magnetic stimulation (rTMS) on motor function in stroke patients. There have been relatively few studies on the effects of different modalities of rTMS on lower extremity motor function and corticospinal excitability in patients with stroke. The MEDLINE, Embase, Cochrane Library, ISI Science Citation Index, Physiotherapy Evidence Database, China National Knowledge Infrastructure Library, and ClinicalTrials.gov databases were searched. Parallel or crossover randomized controlled trials that addressed the effectiveness of rTMS in patients with stroke, published from inception to November 28, 2019, were included. Standard pairwise meta-analysis was conducted using R version 3.6.1 with the “meta” package. Bayesian network analysis using the Markov chain Monte Carlo algorithm was conducted to investigate the effectiveness of different rTMS protocol interventions. Network meta-analysis results of 18 randomized controlled trials regarding lower extremity motor function recovery revealed that low-frequency rTMS had better efficacy in promoting lower extremity motor function recovery than sham stimulation. Network meta-analysis results of five randomized controlled trials demonstrated that high-frequency rTMS led to higher amplitudes of motor evoked potentials than low-frequency rTMS or sham stimulation. These findings suggest that rTMS can improve motor function in patients with stroke, and that low-frequency rTMS mainly affects motor function, whereas high-frequency rTMS increases the amplitudes of motor evoked potentials. More high-quality randomized controlled trials are needed to validate this conclusion. The work was registered in PROSPERO (registration No. CRD42020147055) on April 28, 2020. 
    Related Articles | Metrics
    Current application and future directions of photobiomodulation in central nervous diseases
    Muyue Yang, Zhen Yang, Pu Wang, Zhihui Sun
    2021, 16 (6):  1177-1185.  doi: 10.4103/1673-5374.300486
    Abstract ( 275 )   PDF (339KB) ( 288 )   Save
    Photobiomodulation using light in the red or near-infrared region is an innovative treatment strategy for a wide range of neurological and psychological conditions. Photobiomodulation can promote neurogenesis and elicit anti-apoptotic, anti-inflammatory and antioxidative responses. Its therapeutic effects have been demonstrated in studies on neurological diseases, peripheral nerve injuries, pain relief and wound healing. We conducted a comprehensive literature review of the application of photobiomodulation in patients with central nervous system diseases in February 2019. The NCBI PubMed database, EMBASE database, Cochrane Library and ScienceDirect database were searched. We reviewed 95 papers and analyzed. Photobiomodulation has wide applicability in the treatment of stroke, traumatic brain injury, Parkinson’s disease, Alzheimer’s disease, major depressive disorder, and other diseases. Our analysis provides preliminary evidence that PBM is an effective therapeutic tool for the treatment of central nervous system diseases. However, additional studies with adequate sample size are needed to optimize treatment parameters.
    Related Articles | Metrics
    Polyglutamine diseases: looking beyond the neurodegenerative universe
    Michal Mielcarek, Mark Isalan
    2021, 16 (6):  1186-1187.  doi: 10.4103/1673-5374.300434
    Abstract ( 98 )   Save
    Multisystem disorders are often manifested by affecting more than one bodily system or tissue. We have recently reported a significantly higher prevalence of co-existing conditions in the cohorts of both pre- and symptomatic Huntington’s disease (HD) gene carriers. We reported that even pre-symptomatic HD patients had a significantly higher number of comorbid conditions, while the symptomatic group of HD patients was characterized by a significantly lower percentage of subjects without any comorbidity (7%) in comparison to the control group (50%). This led us to conclude that HD patients have more comorbidities than controls and the number of comorbidities increases in number as the disease progresses. For the first time, we identified 8 clusters of comorbid conditions, with musculoskeletal, psychiatric and cardiovascular clusters being significantly more frequent in both pre- and symptomatic HD patients, while neurological and gastrointestinal clusters showed significantly higher occurrences in the HD symptomatic group (Zielonka et al., 2020).
    Related Articles | Metrics
    Implanted devices: the importance of both electrochemical performance and biological acceptance 
    Ashley N. Dalrymple
    2021, 16 (6):  1188-1189.  doi: 10.4103/1673-5374.300342
    Abstract ( 95 )   PDF (687KB) ( 94 )   Save
    Neural interfaces can be implanted throughout the body to restore function, including cochlear implants for severe hearing loss, deep brain stimulation for tremor, and spinal cord stimulation for pain. These devices are intended to remain implanted and effective for the lifetime of the user, which could be several decades. Device performance and longevity can be impacted by the state of the electrode-tissue interface. Electrochemical performance and tissue reaction to implanted electrodes are important factors to consider when testing novel electrodes and materials, and can facilitate understanding of the reactions at the interface. The works summarized in this perspective highlight the significance of evaluating the electrochemical properties and bioreactivity of implanted electrodes in concert through chronic in vivo studies. Cochlear implants are used as a case study; however, the results are relevant to all neural interfaces. Electrochemical performance and tissue reactivity must be considered in future studies evaluating electrodes and materials prior to testing in people. 
    Related Articles | Metrics
    Strategies on the application of stem cells based therapies for the treatment of optic neuropathies
    Sanaz Behtaj, Maksym Rybachuk
    2021, 16 (6):  1190-1191.  doi: 10.4103/1673-5374.300343
    Abstract ( 119 )   PDF (550KB) ( 61 )   Save
    The retinal ganglion cells (RGCs) are not able to regenerate following optic nerve injury resulting in an irreversible vision loss in patients with optic neuropathies including glaucoma. Recent findings in ocular regeneration have opened promising avenues to apply stem cell-based modalities to restore vision in progressive optic neuropathies. Stem cell-based therapies can help to improve retinal regeneration by solving two major problems: (1) by preventing secondary degeneration of RGCs and preserving the remaining vision, and (2) by replacing degenerated RGCs and promoting RGC axon regeneration in the damaged area. The first approach, known as neuroprotective therapy, uses stem cells incorporated into the degenerating retina with an aim to offer a nourishing environment for damaged RGCs resulting in anatomic and functional improvement. The second approach, known as RGC replacement therapy, ultimately aims at replacing the damaged RGCs with healthy RGCs or RGC precursors (Gao et al., 2012; Fu et al., 2019) in order to restore the visual function. Both approaches are graphically represented in Figure 1. The implementation of cell replacement therapeutic approaches requires successful generation of clinically safe and functional RGCs in an environment where the transplants survive, appropriately integrate and engraft, as well as establish neurites within the hosts’ retina and direct consequent axons towards the relevant regions in the brain (Behtaj et al., 2020). In this work, we discuss the challenges that are required to be addressed prior to the implementation of stem cell-based therapies in clinical practice and, suggest potential solutions to overcome the current limitations.
    Related Articles | Metrics
    Astrocytic role of Thy-1 induced inhibition of axonal sprouting
    Sara T. Whiteman, Jason M. Askvig
    2021, 16 (6):  1192-1193.  doi: 10.4103/1673-5374.300429
    Abstract ( 62 )   PDF (215KB) ( 102 )   Save
    Since the identification of Thy-1 (CD-90) in 1964 by Reif and Allen, the precise functions of this glycosyl phosphatidylinositol-anchored surface protein within the central nervous system have been difficult to characterize. Thy-1 is a 110-amino acid cellular adhesion molecule that plays a role in cell-cell communication by interacting with other  cellular adhesion molecules, including integrins (Leyton et al., 2001). While found on a diverse array of cells within the body, Thy-1 is present on the surface of almost every neuron in the brain (Morris and Grosveld, 1989). In Reif and Allen’s 1964 study, they found that Thy-1 levels increased in the brain nearly 100-fold during post-natal development (Reif and Allen, 1964). Later, Xue et al. (1990) elaborated on this discovery and found that Thy-1 protein levels increased following the cessation of axonal and dendritic growth in the olfactory system. It was subsequently shown that neuronal Thy-1 inhibited neurite outgrowth on astrocytes in vitro, but not Schwann cells or embryonic glia, and anti-Thy-1 antibody is able to counteract the inhibition (Tiveron et al., 1992). Moreover, Thy-1 is localized to the ends of axon terminals, which may account for the ability of Thy-1 to block axon growth (Herrera-Molina et al., 2012).

    Related Articles | Metrics
    Microglia-oligodendrocyte intercellular communication: role of extracellular vesicle lipids in functional signalling
    Alice Gualerzi, Marta Lombardi, Claudia Verderio
    2021, 16 (6):  1194-1195.  doi: 10.4103/1673-5374.300430
    Abstract ( 69 )   PDF (572KB) ( 127 )   Save
    The term microglia refers to the group of resident brain immune-cells that are responsible, mainly, for the immune response and the homeostasis of the brain. Unlike monocyte-derived macrophages that infiltrate the brain, microglia are long-lived cells which arise exclusively from the embryonic yolk sac (Stratoulias et al., 2019). Impairment in microglia functions is at the basis for the development of multiple brain diseases, including multiple sclerosis and neurodegenerative diseases. In the last decade, the number of research articles and reviews dealing with the role of microglia in the pathogenesis of brain disorders has exponentially increased. Indeed, microglia cells play a major role in both the brain homeostasis and the onset and maintenance of the inflammatory processes within the central nervous system (CNS) that often accompany the diseases. The complex mechanisms by which microglia exert their action in pathological conditions have not been completely clarified, yet, but their ability to mediate the intercellular communication with all of the other cell populations in the CNS tissue has clearly emerged in both physiological and pathological conditions (Paolicelli et al., 2018).
    Related Articles | Metrics
    Fibrinogen triggered signaling pathways modify stem cell behavior in central nervous system disease
    Jia-Di Lin, Yu-Hsuan Chu, Suvra Nath, Christian Schachtrup
    2021, 16 (6):  1196-1197.  doi: 10.4103/1673-5374.300436
    Abstract ( 102 )   PDF (660KB) ( 85 )   Save
    Neural stem/precursor cells (NSPCs) hold great promise in improving central nervous system (CNS) repair, either by triggering endogenous NSPC sources of the CNS or by transplantation of NSPCs. The molecular mechanisms of NSPC survival and integration as well as their cell fate determination are still insufficiently understood, yet will be instrumental for harnessing these cells for brain repair. In our recent Nature Communications manuscript entitled “Fibrinogen induces neural stem cell differentiation into astrocytes in the subventricular zone via BMP signaling” (Pous et al., 2020), we advanced towards understanding how CNS disease alters the brain subventricular zone (SVZ) stem cell niche environment by the disease-triggered deposition of blood-derived factors and how these factors regulate NSPC fate and brain repair. Here, we summarize the relevance of our original findings for NSPC biology in CNS disease, its possible implications for other CNS stem cell niches and other CNS diseases. Also, we discuss how current knowledge can be applied to control NSPC fate and functions tailored to promote CNS repair.
    Related Articles | Metrics
    Chaperone proteins as ameliorators of α-synuclein-induced synaptic pathologies: insights into Parkinson’s disease
    Susan M.L. Banks, Audrey T. Medeiros, Rui Sousa, Eileen M. Lafer, Jennifer R. Morgan
    2021, 16 (6):  1198-1199.  doi: 10.4103/1673-5374.300431
    Abstract ( 201 )   PDF (1012KB) ( 105 )   Save
    Neurodegenerative disorders, such as Parkinson’s disease (PD) and other synucleinopathies, impact the lives of millions of patients and their caregivers. Synucleinopathies include PD, dementia with Lewy Bodies (DLB), multiple system atrophy, and several Alzheimer’s Disease variants. They are clinically characterized by intracellular inclusions called Lewy Bodies, which are rich in atypical aggregates of the protein α-synuclein. While dopaminergic neurons in the substantia nigra are particularly susceptible to α-synuclein-induced aggregation and neurodegeneration, glutamatergic neurons in other brain regions (e.g. cortex) are also frequently affected in PD and other synucleinopathies (Schulz-Schaeffer 2010). Several point mutations in the α-synuclein gene (SNCA), as well as duplication/triplication of SNCA, are linked to familial Parkinson’s disease. In animal models, these genetic alterations lead to overexpression and aberrant accumulation of α-synuclein within neurons, and eventually to neurodegeneration. Interestingly, in both animal models and human patients, α-synuclein aggregation often occurs at neuronal synapses and within axons prior to the appearance of larger aggregates (i.e. Lewy bodies) and other signs of neurodegeneration (Schulz-Schaeffer 2010; Volpicelli-Daley et al., 2011). The level of synaptic aggregation of α-synuclein is highly correlated with greater cognitive deficits in PD and DLB patients (Schulz-Schaeffer 2010). Thus, it is essential to understand how excess α-synuclein impacts synapses, as this may represent an early stage in the neurodegenerative disease progression and thus a viable target for therapeutic intervention, particularly with respect to cognitive impairment.
    Related Articles | Metrics
    The role of viruses in the pathogenesis of Parkinson’s disease
    José Fidel Baizabal-Carvallo, Marlene Alonso-Juarez
    2021, 16 (6):  1200-1201.  doi: 10.4103/1673-5374.300437
    Abstract ( 76 )   PDF (223KB) ( 188 )   Save
    Parkinson’s disease (PD) is a neurological degenerative disorder characterized by loss of dopaminergic neurons in the substantia nigra (SN) and intracellular inclusions called Lewy bodies and Lewy tangles, composed mainly by aggregates of α-synuclein. Braak et al. (2003) proposed that the olfactory epithelium and intestines are the anatomical sites where PD initiates; as pathological aggregates of α-synuclein are detected in these tissues in very early or prodromal PD. In this scenario, α-synuclein seems to reach the central nervous system (CNS) by axonal transport through the sympathetic nervous system, the glossopharyngeal and vagus nerves as well as the olfactory pathways (Braak et al., 2003).
    Related Articles | Metrics
    The role of microglia versus peripheral macrophages in maladaptive plasticity after nerve injury
    Thomas A. Szabo-Pardi, Nilesh M. Agalave, Michael D. Burton
    2021, 16 (6):  1202-1203.  doi: 10.4103/1673-5374.300438
    Abstract ( 95 )   PDF (865KB) ( 104 )   Save
    Microglia and macrophages encompass the innate immune response to injury in the central and peripheral nervous systems, respectively, and are intimately involved in the pathogenesis of maladaptive changes (Tsuda, 2019). These dynamic cells can influence neuronal activity in active and quiescent states. Conflicting findings argue that peripheral macrophages facilitate the development of nerve injury-induced neuropathic pain, as opposed to central microglia (Lopes et al., 2017; Yu et al., 2020). It is imperative to discern their spatiotemporal contributions to the development and maintenance of maladaptive conditions, such as neuropathic pain (Inoue and Tsuda, 2018). The individual role of these cell types is difficult to parse out because both microglia and macrophages exhibit a keen ability to react quickly to injury and remain reactive after injury-induced changes. Appropriate methods to isolate and characterize these cells in downstream applications is necessary to uncover key findings (Agalave et al., 2020).
    Related Articles | Metrics
    Microglial depletion and repopulation: a new era of regenerative medicine?
    Alexandra M. Barnett, Fulton T. Crews, Leon G. Coleman
    2021, 16 (6):  1204-1205.  doi: 10.4103/1673-5374.300439
    Abstract ( 115 )   PDF (374KB) ( 127 )   Save
    Microglia have multiple functions and phenotypes that can prevent or worsen neuropathology.  Microglial depletion and repopulation methods provide a promising technique for understanding microglial biology. Their utility as therapeutic modalities is now under consideration. As resident immune cells in the central nervous system (CNS), microglia maintain the local environment and promote neuronal vitality. However, persistent proinflammatory signaling due to aberrant microglial activation can be detrimental. This is seen in settings such as traumatic brain injury (TBI) (Henry et al., 2020), stroke (Li et al., 2017), and alcohol use disorder (AUD) (Coleman et al., 2020), when the loss of homeostatic control results in persistent proinflammatory signaling that contributes to ongoing neuropathology (Figure 1). Thus, selective replacement of chronically proinflammatory-activated microglia could improve functional outcomes. Depletion of microglia from the CNS microenvironment has served as a helpful tool to understand the contribution of microglia to brain disease. In this perspective, we discuss our recent findings regarding microglial depletion and repopulation in AUD as well as the benefits and detriments of microglial depletion and repopulation and their potential therapeutic applications in other neuropathological disease models. We recently reported that microglial depletion and repopulation protected against long-term proinflammatory activation in a model of AUD (Coleman et al., 2020). In a primary ex vivo brain slice culture model, binge ethanol caused a persistent induction of proinflammatory cytokines. Microglial depletion and repopulation after ethanol treatment, using methods described below, resulted in a normalization of proinflammatory cytokines to baseline levels, and an increase in protective trophic factors such as brain-derived neurotrophic factor. Thus, repopulation of microglia could potentially reverse neuropathology associated with chronic proinflammatory signaling in AUD and other proinflammatory neurological diseases (Figure 1).  
    Related Articles | Metrics
    Functional imaging of astrocyte activity
    Hiroki Kato, Tatsusada Okuno
    2021, 16 (6):  1206-1207.  doi: 10.4103/1673-5374.300432
    Abstract ( 66 )   PDF (572KB) ( 97 )   Save
    Astrocytes have become known to play a central role in various neuroinflammatory diseases. The evaluation of astrocyte activity using functional imaging is becoming more important. Glucose metabolism or oxygen metabolism in the brain can be assessed using the established clinical imaging methods of F-18 fluorodeoxyglucose positron emission tomography (PET) and O-15 PET, respectively. However, until recently, the highly specific evaluation of metabolic activity in astrocytes has never been applied clinically. Since acetate is selectively taken up and metabolized by astrocytes, its usefulness as a tracer for measuring astrocyte activity has been proposed in basic research. In a human study, the activation of astrocytes associated with neuronal activation has been evaluated in vivo using 1-C-11 acetate PET (Wyss et al., 2009). Astrocytes supply lactate as an energy source to neurons through monocarboxylate transporters (MCTs) and receive and metabolize neurotransmitter glutamate from neurons. The tricarboxylic acid (TCA) cycle in astrocytes provides energy to convert glutamate released from neurons into glutamine as well as newly generating glutamine for neurons. 1-C-11 acetate is selectively taken up by astrocytes mainly by MCT, especially MCT1 and MCT2, and is metabolized by the TCA cycle via acetyl-CoA. Half of the label derived from 1-C-11 acetate is washed out as CO2 in the second round of the TCA cycle in astrocytes, and most of the remaining label is metabolized to glutamate. Considering the short half-life of C-11, metabolites in neurons derived from labeled glutamate, which had been transformed from glutamine passed from the astrocytes, is negligible (Wyss et al., 2009). Therefore, the CO2 washout rate is an index that quantitatively represents the metabolic activity of astrocytes, and the index can be imaged using quantitative PET as the efflux rate of the tracer. Moreover, the tracer accumulation is considered to reflect mainly the labeled glutamine/glutamate pool derived from 1-C-11 acetate. Therefore, 1-C-11 acetate PET can be used to evaluate the central part of astrocyte energy metabolism (Figure 1). 
    Related Articles | Metrics
    Degenerative and regenerative processes in amyotrophic lateral sclerosis: motor reserve, adaptation and putative compensatory changes
    Peter Bede, Ulrich Bogdahn, Jasmin Lope, Kai Ming Chang, Sophia Xirou, Foteini Christidi
    2021, 16 (6):  1208-1209.  doi: 10.4103/1673-5374.300440
    Abstract ( 80 )   PDF (473KB) ( 113 )   Save
    Research in ALS has gained unprecedented momentum in recent years fueled by important conceptual developments, establishment of international consortia, breakthrough genetic discoveries and relentless technological advances. The first genotype-specific pharmaceutical trials signal the paradigm shift from the notion of ‘one-drug-for-all’ to precision, individualized therapies. The once arcane presymptomatic phase of the disease is gradually unraveled by seminal studies of asymptomatic mutation carriers (Geevasinga et al., 2015; Querin et al., 2019). The meticulous analysis of data from large population-based registries has contributed to the identification of etiological factors, genetic risk profiles, epigenetic and environmental modifiers. Progression patterns have been characterized in vivo by robust clinical, neurophysiology and neuroimaging studies and led to the development of clinical staging systems and biomarkers with practical utility in clinical trials (Chipika et al., 2019). While ALS was once considered a ‘pure’ motor system disorder, it is now widely regarded as multisystem condition with frontotemporal, cerebellar, and subcortical grey matter involvement and a range of extrapyramidal, cognitive, and behavioral manifestations (Elamin et al., 2017). Disease-specific functional rating scales are now routinely used and screening instruments have been developed to assess the most commonly affected cognitive and behavioral domains in ALS. Advances in genetics paved the way for the first large presymptomatic studies which confirmed considerable cerebral and spinal cord alterations decades before symptom manifestation (Vucic et al., 2008; Querin et al., 2019). The characterization of genotype-associated molecular cascades, pathological signatures and clinical features were important milestones for the development of novel therapies, and the first antisense oligonucleotide trials are now underway. The datasets generated by multicenter initiatives offer unprecedented data mining opportunities; clustering patterns, prognostic determinants, and reliable diagnostic indicators were identified using machine-learning approaches that could not have previously been applied to smaller datasets. Technological advances in electrophysiology and the emergence of magnetoencephalography generated important functional insights (Bede et al., 2018). Novel imaging modalities, such as multi-voxel spectroscopy, spinal cord imaging, diffusion kurtosis imaging captured pathological changes that were previously impossible to ascertain in vivo (Bede et al., 2017; Huang et al., 2020). Advanced neurophysiology techniques, such as transcranial magnetic stimulation or motor unit number estimation are now widely used in both clinical and academic settings and contribute to diagnostic clarification and the monitoring of individual patients. In response to the inevitable sample size limitations of single-center studies (Schuster et al., 2016), ambitious international initiatives such as Project MinE established large biobanks to conduct genetic studies with sufficient statistical power. Societies such as NISALS provide pioneering frameworks to conduct large multicenter neuroimaging studies.   
    Related Articles | Metrics
    Complement: a global immunometabolic regulator in amyotrophic lateral sclerosis
    Martin W. Lo, John D. Lee
    2021, 16 (6):  1210-1211.  doi: 10.4103/1673-5374.300441
    Abstract ( 113 )   PDF (626KB) ( 127 )   Save
    Recently, McDonald et al. (2020) proposed that complement causes a metabolic switch to lipid consumption in amyotrophic lateral sclerosis (ALS), which raises the possibility of complement as a global immunometabolic regulator. In ALS, neuromuscular degeneration occurs concurrently with complement-mediated inflammation, glucose intolerance, and elevated lipid consumption. Moreover, complement is being increasingly recognized as a metabolic regulator that can induce insulin resistance and alter fuel source selection. Thus, the authors propose that chronic complement activation may be driving metabolic reprogramming in ALS, which has broad implications for the field of immunometabolism and would be the first mechanistic explanation of the “lipid switch” in ALS. Specifically, it suggests that complement can drive a global shift in resources during inflammation, in which glucose is redirected from peripheral tissues to immune cells to support their inflammatory actions. To illustrate this perspective, here we introduce immunometabolism and make a case for complement as a global immunometabolic regulator in ALS.
    Related Articles | Metrics
    ULK1 as a novel therapeutic target in neurodegeneration
    Björn Friedhelm Vahsen, Paul Lingor
    2021, 16 (6):  1212-1213.  doi: 10.4103/1673-5374.300442
    Abstract ( 77 )   PDF (545KB) ( 83 )   Save
    Axonal degeneration is an early and key pathophysiological feature of many traumatic and neurodegenerative disorders of the central nervous system (CNS), such as spinal cord injury (SCI), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). As the regenerative capacity of injured axons is severely restricted in the CNS, axonal degeneration frequently results in the irreversible loss of neuronal connections causing progressive neurological deficits and clinical disability. A better understanding of the mechanisms of axon degeneration is therefore hoped to unravel new therapeutic avenues to combat neurodegeneration (Lingor et al., 2012).
    Related Articles | Metrics
    Cell-based therapies for age-related macular degeneration: cell replacement versus paracrine effects#br#
    Xiaoyan Peng, Ling Gao, Yongqing Liu
    2021, 16 (6):  1214-1215.  doi: 10.4103/1673-5374.300443
    Abstract ( 81 )   PDF (428KB) ( 104 )   Save
    Age-related macular degeneration (AMD) is a leading cause for severe visual loss and legal blindness in seniors worldwide. The molecular basis for the disease remains poorly understood, likely involving genetic and environment-related ocular defects. Its pathogenesis proceeds slowly, started with deposits of fatty proteins (drusen) in the Bruch’s membrane, followed by gradual impairments of the posterior choriocapillaris and the anterior retinal pigment epithelium (RPE), and lead to irreversible degeneration of the light receiving neurons (photoreceptor) and vision decline. Clinically, AMD is divided into two subgroups: dry or atrophy form and wet or exudative form. Twenty percent of AMD patients have the wet form. The wet AMD is linked to choroidal neovascularization  located in the subretinal macular region, with subsequent bleeding, and a possible sudden loss of central vision. AMD is an incurable devastating disease though the wet form is treatable by anti-vascular endothelial growth factor (VEGF) drugs to inhibit choroidal neovascularization so as to improve or maintain patients’ visual function. For the dry form of AMD unfortunately, there is no effective treatment available in the clinic though the light at the end of tunnel is emerging: cell-based therapy is a potential solution for treating AMD (Chichagova et al., 2018).
    Related Articles | Metrics
    Prion protein in myelin maintenance:  what does the goat say?
    Fredrik S. Skedsmo, Arild Espenes, Michael A. Tranulis
    2021, 16 (6):  1216-1217.  doi: 10.4103/1673-5374.300444
    Abstract ( 84 )   PDF (219KB) ( 336 )   Save
    The cellular prion protein PrPC has been extensively studied because it can adopt a pathogenic three-dimensional conformation that causes rare, but invariably fatal, neurodegenerative prion diseases in humans and other mammals. The disease-causing conformer of the protein is called PrPSc, of which oligomeric aggregates constitute prion agents that can bind to, and convert further, PrPC molecules into PrPSc (Prusiner, 1998). Thus, in the poorly understood process of prion propagation, there is transfer of biological information encoded solely by protein conformation. Prions can spread within a tissue secondary to a spontaneous conversion of PrPC to PrPSc or upon transmission of prion agents between individuals.
    Related Articles | Metrics
    Schwann-like adipose-derived stem cells as a promising therapeutic tool for peripheral nerve regeneration: effects of cholinergic stimulation
    Roberta Piovesana, Alessandro Faroni, Ada Maria Tata, Adam J. Reid
    2021, 16 (6):  1218-1220.  doi: 10.4103/1673-5374.300433
    Abstract ( 125 )   PDF (896KB) ( 89 )   Save
    Peripheral nerve injuries (PNIs) are a common clinical problem usually as a consequence of trauma. Despite optimal surgical management, PNI has a lifelong impact on function and wellbeing of the patient.
    Related Articles | Metrics
    The inducible nitric oxide synthase-inhibitor 1400W as a potential treatment for retinal diseases
    Sven Schnichels, Stephanie C. Joachim
    2021, 16 (6):  1221-1222.  doi: 10.4103/1673-5374.300445
    Abstract ( 73 )   PDF (762KB) ( 87 )   Save
    The overproduction of reactive oxygen species is defined as oxidative stress. While deprived oxygen supply in tissues is known as hypoxia. These mechanisms contribute to the pathogenesis of several retinal diseases, like glaucoma, age-related macular degeneration (AMD), and retinal ischemia. Glaucoma is a neurodegenerative disease defined by a progressive loss of retinal ganglion cells (RGCs) and their axons causing visual field defects, which ultimately leads to blindness. While AMD pathogenesis is further characterized by soft drusen, it involves the retinal pigment epithelium and the Bruch’s membrane - choroid complex. AMD is a disease finally leading to death of the photoreceptors, especially in the macula region, resulting in central vision loss. The pathomechanisms of both diseases are not yet fully understood, but oxidative as well as hypoxic stresses seem to play a crucial. Moreover, hypoxia is also an inducer of oxidative stress by causing cellular stress, followed by mitochondrial stress, in neurodegenerative retinal diseases. In experimental models of retinal degeneration numerous triggers of cell death have been identified, including genetic mutations, intracellularly elevated calcium levels, endoplasmic reticulum stress, as well as hypoxia, and oxidative stress (Grossniklaus et al., 2010; Pang and Clark, 2020). 
    Related Articles | Metrics
    Thrombin in peripheral nerves: friend or foe?
    Elena Pompili, Cinzia Fabrizi
    2021, 16 (6):  1223-1224.  doi: 10.4103/1673-5374.300446
    Abstract ( 79 )   PDF (458KB) ( 72 )   Save
    Differently from the central nervous system (CNS), the peripheral nervous system (PNS) exhibits a high regenerative capacity. This ability is  related to the remarkable plasticity of Schwann cells (SCs) which after nerve injury convert to a repair-promoting phenotype to a large extent. Nerve injury is accompanied by a rapid rise of thrombin levels (Bushi et al., 2016; Gera et al., 2016). Thrombin is the key effector protease of the coagulation cascade which elicits hormone-like actions by the activation of G-protein coupled receptors known as protease-activated receptors (PARs). Inflammation and coagulation are two complex and interconnected pathways whose mutual interactions have been only partially elucidated.
    Related Articles | Metrics