Loading...

Table of Content

    15 July 2021, Volume 16 Issue 7 Previous Issue    Next Issue
    For Selected: Toggle Thumbnails
    Transcranial pulse current stimulation improves the locomotor function in a rat model of stroke
    Wen-Jing Wang, Yan-Biao Zhong, Jing-Jun Zhao, Meng Ren, Si-Cong Zhang, Ming-Shu Xu, Shu-Tian Xu, Ying-Jie Zhang, Chun-Lei Shan
    2021, 16 (7):  1229-1234.  doi: 10.4103/1673-5374.301018
    Abstract ( 166 )   PDF (1065KB) ( 145 )   Save
    Previous studies have shown that transcranial pulse current stimulation (tPCS) can increase cerebral neural plasticity and improve patients’ locomotor function. However, the precise mechanisms underlying this effect remain unclear. In the present study, rat models of stroke established by occlusion of the right cerebral middle artery were subjected to tPCS, 20 minutes per day for 7 successive days. tPCS significantly reduced the Bederson score, increased the foot print area of the affected limbs, and reduced the standing time of affected limbs of rats with stroke compared with that before intervention. Immunofluorescence staining and western blot assay revealed that tPCS significantly increased the expression of microtubule-associated protein-2 and growth-associated protein-43 around the ischemic penumbra. This finding suggests that tPCS can improve the locomotor function of rats with stroke by regulating the expression of microtubule-associated protein-2 and growth-associated protein-43 around the ischemic penumbra. These findings may provide a new method for the clinical treatment of poststroke motor dysfunction and a theoretical basis for clinical application of tPCS. The study was approved by the Animal Use and Management Committee of Shanghai University of Traditional Chinese Medicine of China (approval No. PZSHUTCM190315003) on February 22, 2019. 
    Related Articles | Metrics
    Comparative transcriptomic analysis of rat versus mouse cerebral cortex after traumatic brain injury
    Meng-Shi Yang, Xiao-Jian Xu, Bin Zhang, Fei Niu, Bai-Yun Liu
    2021, 16 (7):  1235-1243.  doi: 10.4103/1673-5374.301028
    Abstract ( 168 )   PDF (3916KB) ( 171 )   Save
    The heterogeneity of traumatic brain injury (TBI)-induced secondary injury has greatly hampered the development of effective treatments for TBI patients. Targeting common processes across species may be an innovative strategy to combat debilitating TBI. In the present study, a cross-species transcriptome comparison was performed for the first time to determine the fundamental processes of secondary brain injury in Sprague-Dawley rat and C57/BL6 mouse models of TBI, caused by acute controlled cortical impact. The RNA sequencing data from the mouse model of TBI were downloaded from the Gene Expression Omnibus (ID: GSE79441) at the National Center for Biotechnology Information. For the rat data, peri-injury cerebral cortex samples were collected for transcriptomic analysis 24 hours after TBI. Differentially expressed gene-based functional analysis revealed that common features between the two species were mainly involved in the regulation and activation of the innate immune response, including complement cascades as well as Toll-like and nucleotide oligomerization domain-like receptor pathways. These findings were further corroborated by gene set enrichment analysis. Moreover, transcription factor analysis revealed that the families of signal transducers and activators of transcription (STAT), basic leucine zipper (BZIP), Rel homology domain (RHD), and interferon regulatory factor (IRF) transcription factors play vital regulatory roles in the pathophysiological processes of TBI, and are also largely associated with inflammation. These findings suggest that targeting the common innate immune response might be a promising therapeutic approach for TBI. The animal experimental procedures were approved by the Beijing Neurosurgical Institute Animal Care and Use Committee (approval No. 201802001) on June 6, 2018.
    Related Articles | Metrics
    Delayed atomoxetine or fluoxetine treatment coupled with limited voluntary running promotes motor recovery in mice after ischemic stroke
    Faisal F. Alamri, Abdullah Al Shoyaib, Nausheen Syeara, Anisha Paul, Srinidhi Jayaraman, Serob T. Karamyan, Thiruma V. Arumugam, Vardan T. Karamyan
    2021, 16 (7):  1244-1251.  doi: 10.4103/1673-5374.301031
    Abstract ( 141 )   PDF (2116KB) ( 151 )   Save
    Currently, there is an unmet need for treatments promoting post-stroke functional recovery. The aim of this study was to evaluate and compare the dose-dependent effect of delayed atomoxetine or fluoxetine therapy (starting on post-stroke day 5), coupled with limited physical exercise (2 hours daily voluntary wheel running; post-stroke days 9 to 42), on motor recovery of adult male mice after photothrombotic stroke. These drugs are selective norepinephrine or serotonin reuptake inhibitors indicated for disorders unrelated to stroke. The predetermined primary end-point for this study was motor function measured in two tasks of spontaneous motor behaviors in grid-walking and cylinder tests. Additionally, we quantified the running distance and speed throughout the study, the number of parvalbumin-positive neurons in the medial agranular cortex and infarct volumes. Both sensorimotor tests revealed that neither limited physical exercise nor a drug treatment alone significantly facilitated motor recovery in mice after stroke. However, combination of physical exercise with either of the drugs promoted restoration of motor function by day 42 post-stroke, with atomoxetine being a more potent drug. This was accompanied by a significant decrease in parvalbumin-positive inhibitory interneurons in the ipsilateral medial agranular cortex of mice with recovering motor function, while infarct volumes were comparable among experimental groups. If further validated in larger studies, our observations suggest that add-on atomoxetine or fluoxetine therapy coupled with limited, structured physical rehabilitation could offer therapeutic modality for stroke survivors who have difficulty to engage in early, high-intensity physiotherapy. Furthermore, in light of the recently completed Assessment oF FluoxetINe In sTroke recoverY (AFFINITY) and Efficacy oF Fluoxetine-a randomisEd Controlled Trial in Stroke (EFFECTS) trials, our observations call for newly designed studies where fluoxetine or atomoxetine pharmacotherapy is evaluated in combination with structured physical rehabilitation rather than alone. This study was approved by the Texas Tech University Health Sciences Center Institutional Animal Care and Use Committee (protocol # 16019).
    Related Articles | Metrics
    Extremely low frequency electromagnetic fields promote cognitive function and hippocampal neurogenesis of rats with cerebral ischemia
    Qiang Gao, Aaron Leung, Yong-Hong Yang, Benson Wui-Man Lau, Qian Wang, Ling-Yi Liao, Yun-Juan Xie, Cheng-Qi He
    2021, 16 (7):  1252-1257.  doi: 10.4103/1673-5374.301020
    Abstract ( 132 )   PDF (1107KB) ( 243 )   Save
    Extremely low frequency electromagnetic fields (ELF-EMF) can improve the learning and memory impairment of rats with Alzheimer’s disease, however, its effect on cerebral ischemia remains poorly understood. In this study, we established rat models of middle cerebral artery occlusion/reperfusion. One day after modeling, a group of rats were treated with ELF-EMF (50 Hz, 1 mT) for 2 hours daily on 28 successive days. Our results showed that rats treated with ELF-EMF required shorter swimming distances and latencies in the Morris water maze test than those of untreated rats. The number of times the platform was crossed and the time spent in the target quadrant were greater than those of untreated rats. The number of BrdU+/NeuN+ cells, representing newly born neurons, in the hippocampal subgranular zone increased more in the treated than in untreated rats. Up-regulation in the expressions of Notch1, Hes1, and Hes5 proteins, which are the key factors of the Notch signaling pathway, was greatest in the treated rats. These findings suggest that ELF-EMF can enhance hippocampal neurogenesis of rats with cerebral ischemia, possibly by affecting the Notch signaling pathway. The study was approved by the Institutional Ethics Committee of Sichuan University, China (approval No. 2019255A) on March 5, 2019. 
    Related Articles | Metrics
    Cortical transcriptome analysis after spinal cord injury reveals the regenerative mechanism of central nervous system in CRMP2 knock-in mice
    Ayaka Sugeno, Wenhui Piao, Miki Yamazaki, Kiyofumi Takahashi, Koji Arikawa, Hiroko Matsunaga, Masahito Hosokawa, Daisuke Tominaga, Yoshio Goshima, Haruko Takeyama, Toshio Ohshima
    2021, 16 (7):  1258-1265.  doi: 10.4103/1673-5374.301035
    Abstract ( 86 )   PDF (2722KB) ( 335 )   Save
    Recent studies have shown that mutation at Ser522 causes inhibition of collapsin response mediator protein 2 (CRMP2) phosphorylation and induces axon elongation and partial recovery of the lost sensorimotor function after spinal cord injury (SCI). We aimed to reveal the intracellular mechanism in axotomized neurons in the CRMP2 knock-in (CRMP2KI) mouse model by performing transcriptome analysis in mouse sensorimotor cortex using micro-dissection punching system. Prior to that, we analyzed the structural pathophysiology in axotomized or neighboring neurons after SCI and found that somatic atrophy and dendritic spine reduction in sensorimotor cortex were suppressed in CRMP2KI mice. Further analysis of the transcriptome has aided in the identification of four hemoglobin genes Hba-a1, Hba-a2, Hbb-bs, and Hbb-bt that are significantly upregulated in wild-type mice with concomitant upregulation of genes involved in the oxidative phosphorylation and ribosomal pathways after SCI. However, we observed substantial upregulation in channel activity genes and downregulation of genes regulating vesicles, synaptic function, glial cell differentiation in CRMP2KI mice. Moreover, the transcriptome profile of CRMP2KI mice has been discussed wherein energy metabolism and neuronal pathways were found to be differentially regulated. Our results showed that CRMP2KI mice displayed improved SCI pathophysiology not only via microtubule stabilization in neurons, but also possibly via the whole metabolic system in the central nervous system, response changes in glial cells, and synapses. Taken together, we reveal new insights on SCI pathophysiology and the regenerative mechanism of central nervous system by the inhibition of CRMP2 phosphorylation at Ser522. All these experiments were performed in accordance with the guidelines of the Institutional Animal Care and Use Committee at Waseda University, Japan (2017-A027 approved on March 21, 2017; 2018-A003 approved on March 25, 2018; 2019-A026 approved on March 25, 2019).
    Related Articles | Metrics
    Positive effects of music therapist’s selected auditory stimulation on the autonomic nervous system of patients with disorder of consciousness: a randomized controlled trial
    Xiao-Ying Zhang, Jian-Jun Li, Hai-Tao Lu, Wen-Jia Teng, Song-Huai Liu
    2021, 16 (7):  1266-1272.  doi: 10.4103/1673-5374.301021
    Abstract ( 130 )   PDF (1381KB) ( 396 )   Save
    The current randomized controlled trial was performed at the China Rehabilitation Science Institute, China to test the hypothesis that musical auditory stimulation has positive effects on the autonomic nervous system of patients with disorder of consciousness. Although past studies have recommended that patients with disorder of consciousness listen to patient-preferred music, this practice is not universally accepted by researchers. Twenty patients with severe disorder of consciousness listened to either therapist-selected (n = 10, 6 males and 4 females; 43.33 ± 18.76 years old) or patient-preferred (n = 10, 5 males and 5 females, 48.83 ± 18.79 years old) musical therapy, 30 minutes/day, 5 times/week for 6 weeks. The results showed no obvious differences in heart rate variability-related parameters including heart rate, standard deviation of normal-to-normal R-R intervals, and the root-mean-square of successive heartbeat interval differences of successive heartbeat intervals between the two groups of patients. However, percentage of differences exceeding 50 ms between adjacent normal number of intervals, low-frequency power/high-frequency power, high-frequency power norm, low-frequency power norm, and total power were higher in patients receiving therapist-selected music than in patients receiving their own preferred music. In contrast, this relationship was reversed for the high-frequency power and very-low-frequency band. These results suggest that compared with preferred musical stimulation, therapist-selected musical stimulation resulted in higher interactive activity of the autonomic nervous system. Therefore, therapist-selected musical stimulation should be used to arouse the autonomic nervous system of patients with disorder of consciousness. This study was approved by the Institutional Ethics Committee of China Rehabilitation Research Center, China (approval No. 2018-022-1) on March 12, 2018 and registered with the Chinese Clinical Trial Registry (registration number ChiCTR1800017809) on August 15, 2018. 
    Related Articles | Metrics
    Neuroprotective effect of immunomodulatory peptides in rats with traumatic spinal cord injury
    Dulce Parra-Villamar, Liliana Blancas-Espinoza, Elisa Garcia-Vences, Juan Herrera-García, Adrian Flores-Romero, Alberto Toscano-Zapien, Jonathan Vilchis Villa, Rodríguez Barrera-Roxana, Soria Zavala Karla, Antonio Ibarra, Raúl Silva-García
    2021, 16 (7):  1273-1280.  doi: 10.4103/1673-5374.301485
    Abstract ( 143 )   PDF (4901KB) ( 339 )   Save
    Several therapies have shown obvious effects on structural conservation contributing to motor functional recovery after spinal cord injury (SCI). Nevertheless, neither strategy has achieved a convincing effect. We purposed a combined therapy of immunomodulatory peptides that individually have shown significant effects on motor functional recovery in rats with SCI. The objective of this study was to investigate the effects of the combined therapy of monocyte locomotion inhibitor factor (MLIF), A91 peptide, and glutathione monoethyl ester (GSH-MEE) on chronic-stage spinal cord injury. Female Sprague-Dawley rats underwent a laminectomy of the T9 vertebra and a moderate contusion. Six groups were included: sham, PBS, MLIF + A91, MLIF + GSH-MEE, A91 + GSH-MEE, and MLIF + A91 + GSH-MEE. Two months after injury, motor functional recovery was evaluated using the open field test. Parenchyma and white matter preservation was evaluated using hematoxylin & eosin staining and Luxol Fast Blue staining, respectively. The number of motoneurons in the ventral horn and the number of axonal fibers were determined using hematoxylin & eosin staining and immunohistochemistry, respectively. Collagen deposition was evaluated using Masson’s trichrome staining. The combined therapy of MLIF, A91, and GSH-MEE greatly contributed to motor functional recovery and preservation of the medullary parenchyma, white matter, motoneurons, and axonal fibres, and reduced the deposition of collagen in the lesioned area. The combined therapy of MLIF, A91, and GSH-MEE preserved spinal cord tissue integrity and promoted motor functional recovery of rats after SCI. This study was approved by the National Commission for Scientific Research on Bioethics and Biosafety of the Instituto Mexicano del Seguro Social under registration number R-2015-785-116 (approval date November 30, 2015) and R-2017-3603-33 (approval date June 5, 2017). 
    Related Articles | Metrics
    Suicide transport blockade of motor neuron survival generates a focal graded injury and functional deficit
    Allison S. Liang, Joanna E. Pagano, Christopher A. Chrzan, Randall D. McKinnon
    2021, 16 (7):  1281-1287.  doi: 10.4103/1673-5374.301032
    Abstract ( 117 )   PDF (5964KB) ( 300 )   Save
    We describe a pre-clinical spinal cord motor neuron injury model that is minimal invasive, reproducible, focal and easily applied to small rodents. Retrograde axonal transport of a pro-apoptotic phosphatidylinosotol 3’-kinase inhibitor, wortmannin, via the sciatic nerve results in loss of ipsilateral lumbar motor neurons proportional to the level of drug administered. Motor neuron loss was detected by choline acetyltransferase (ChAT) immunostaining and with a transgenic thy1-eGFP marker. The short half-life of wortmannin generates minimal wound spread, and wortmannin does not affect axon transport, as determined by co-injection of a pseudorabies virus tracer. Using quantitative transcript analysis, we found that ChAT transcripts significantly decreased at 14 days post-delivery of 1 μg wortmannin, relative to sham controls, and remained low after 90 days. Smaller effects were observed with 200 ng and 100 ng wortmannin. Wortmannin also generated a transient and significant increase in astrocyte Gfap transcripts after 14 days with a return to control levels at 90 days. Treated mice had hind limb spasticity and a forced motor function defect that was quantified using a water exit test. Controls rapidly exit a shallow water tray, and wortmannin treated animals were up to 12-fold slower, a phenotype that persisted for at least 3 months. Thus the focal delivery of wortmannin to motor neurons generates a reproducible and scalable injury that can facilitate quantitative studies on neural regeneration and repair. The efficacy of sciatic nerve suicide transport can also explain neurotoxin-mediated selective loss of motor neurons in diseases such as amyotrophic lateral sclerosis. All procedures were performed at Rutgers under established Institutional Animal Care and Use protocols (eIACUC_TR201800022, approved on March 20, 2018). 
    Related Articles | Metrics
    Reducing LncRNA-5657 expression inhibits the brain inflammatory reaction in septic rats
    Yi-An Zhan, Xin-Liang Qiu, Xu-Zhen Wang, Ning Zhao, Ke-Jian Qian
    2021, 16 (7):  1288-1293.  doi: 10.4103/1673-5374.301022
    Abstract ( 145 )   PDF (1820KB) ( 146 )   Save
    Our preliminary study found that the long noncoding RNA (LncRNA)-5657 can reduce the expression of inflammatory factors during inflammatory reactions in rat glial cells. However, the role played by LncRNA-5657 during septic brain injury remains unclear. In the present study, rat models of septic encephalopathy were established by cecal ligation and puncture, and then the rats were treated with a hippocampal injection small hairpin RNA (shRNA) against LncRNA-5657 (sh-LnCRNA-5657). The sh-LncRNA-5657 treatment reduced the level of neuronal degeneration and necrosis in the rat hippocampus, reduced the immunoreactivities of aquaporin 4, heparanase, and metallopeptidase-9, and lowered the level of tumor necrosis factor-alpha. Glial cells were pre-treated with sh-LncRNA-5657 and then treated with 1 µg/mL lipopolysaccharide. Sh-LncRNA-5657 transfection decreased the expression of LncRNA-5657 in lipopolysaccharide-treated glial cells and decreased the mRNA and protein levels of tumor necrosis factor-alpha, interleukin-1β, and interleukin-6. These findings suggested that LncRNA-5657 expression can significantly reduce the inflammatory reaction during septic encephalopathy and induce protective effects against this disease. This study was approved by the Institutional Ethics Committee at the First Affiliated Hospital of Nanchang University of China (approval No. 2017-004) in 2017. 
    Related Articles | Metrics
    Identification of potential oxidative stress biomarkers for spinal cord injury in erythrocytes using mass spectrometry
    Li-Jian Zhang, Yao Chen, Lu-Xuan Wang, Xiao-Qing Zhuang He-Chun Xia
    2021, 16 (7):  1294-1301.  doi: 10.4103/1673-5374.301487
    Abstract ( 129 )   PDF (2103KB) ( 149 )   Save
    Oxidative stress is a hallmark of secondary injury associated with spinal cord injury. Identifying stable and specific oxidative biomarkers is of important significance for studying spinal cord injury-associated secondary injury. Mature erythrocytes do not contain nuclei and mitochondria and cannot be transcribed and translated. Therefore, mature erythrocytes are highly sensitive to oxidative stress and may become a valuable biomarker. In the present study, we revealed the proteome dynamics of protein expression in erythrocytes of beagle dogs in the acute and subacute phases of spinal cord injury using mass spectrometry-based approaches. We found 26 proteins that were differentially expressed in the acute (0–3 days) and subacute (7–21 days) phases of spinal cord injury. Bioinformatics analysis revealed that these differentially expressed proteins were involved in glutathione metabolism, lipid metabolism, and pentose phosphate and other oxidative stress pathways. Western blot assays validated the differential expression of glutathione synthetase, transaldolase, and myeloperoxidase. This result was consistent with mass spectrometry results, suggesting that erythrocytes can be used as a novel sample source of biological markers of oxidative stress in spinal cord injury. Glutathione synthetase, transaldolase, and myeloperoxidase sourced from erythrocytes are potential biomarkers of oxidative stress after spinal cord injury. This study was approved by the Experimental Animal Centre of Ningxia Medical University, China (approval No. 2017-073) on February 13, 2017.
    Related Articles | Metrics
    Genetic polymorphisms in pri-let-7a-2 are associated with ischemic stroke risk in a Chinese Han population from Liaoning, China: a case-control study
    Yu-Ye Wang, He-Yu Zhang, Wen-Juan Jiang, Fang Liu, Lei Li, Shu-Min Deng, Zhi-Yi He, Yan-Zhe Wang
    2021, 16 (7):  1302-1307.  doi: 10.4103/1673-5374.301019
    Abstract ( 103 )   PDF (573KB) ( 116 )   Save
    Ischemic stroke is a complicated disease, and its pathogenesis has been attributed to the occurrence of genetic polymorphisms. Evidence has suggested that the microRNA let-7a is involved in the pathogenesis of ischemic stroke. Pri-miRNA is the primary transcript, which undergoes several processing steps to generate pre-miRNA and, later, mature miRNAs. In this case-control study, we analyzed the distribution of pri-let-7a-2 variants in patients at a high risk for ischemic stroke and the interactions of pri-let-7a-2 variants and environmental factors. Blood samples and clinical information were collected from 1086 patients with ischemic stroke and 836 healthy controls between December 2013 and December 2015 at the First Affiliated Hospital of China Medical University. We found that the rs1143770 CC genotype and the C allele were associated with a decreased risk of ischemic stroke, whereas the rs629367 CC genotype was associated with an increased risk for ischemic stroke. Moreover, these two single-nucleotide polymorphisms were in linkage disequilibrium in this study sample. We analyzed gene-environment interactions and found that rs1143770 exerted a combined effect on the pathogenesis of ischemic stroke, together with alcohol use, smoking, and a history of hypertension. Therefore, the detection of pri-let-7a-2 polymorphisms may increase the awareness of ischemic stroke risk. This study was approved by the Institutional Ethics Committee of the First Affiliated Hospital of China Medical University, China (approval No. 2012-38-1) on February 20, 2012, and was registered with the Chinese Clinical Trial Registry (registration number: ChiCTR-COC-17013559) on December 27, 2017.
    Related Articles | Metrics
    Transferrin receptor 1 plays an important role in muscle development and denervation-induced muscular atrophy
    Ying Li, Juan-Xian Cheng, Hai-Hong Yang, Li-Ping Chen, Feng-Jiao Liu, Yan Wu, Ming Fan, Hai-Tao Wu
    2021, 16 (7):  1308-1316.  doi: 10.4103/1673-5374.301024
    Abstract ( 181 )   PDF (3226KB) ( 144 )   Save
    Previous studies demonstrate an accumulation of transferrin and transferrin receptor 1 (TfR1) in regenerating peripheral nerves. However, the expression and function of transferrin and TfR1 in the denervated skeletal muscle remain poorly understood. In this study, a mouse model of denervation was produced by complete tear of the left brachial plexus nerve. RNA-sequencing revealed that transferrin expression in the denervated skeletal muscle was upregulated, while TfR1 expression was downregulated. We also investigated the function of TfR1 during development and in adult skeletal muscles in mice with inducible deletion or loss of TfR1. The ablation of TfR1 in skeletal muscle in early development caused severe muscular atrophy and early death. In comparison, deletion of TfR1 in adult skeletal muscles did not affect survival or glucose metabolism, but caused skeletal muscle atrophy and motor functional impairment, similar to the muscular atrophy phenotype observed after denervation. These findings suggest that TfR1 plays an important role in muscle development and denervation-induced muscular atrophy. This study was approved by the Institutional Animal Care and Use Committee of Beijing Institute of Basic Medical Sciences, China (approval No. SYXK 2017-C023) on June 1, 2018.
    Related Articles | Metrics
    Absence of ephrin-A2/A3 increases retinal regenerative potential for Müller cells in Rhodopsin knockout mice
    Rui-Lin Zhu, Yuan Fang, Hong-Hua Yu, Dong F. Chen, Liu Yang, Kin-Sang Cho
    2021, 16 (7):  1317-1322.  doi: 10.4103/1673-5374.301034
    Abstract ( 92 )   PDF (1818KB) ( 142 )   Save
    Müller cells (MC) are considered dormant retinal progenitor cells in mammals. Previous studies demonstrated ephrin-As act as negative regulators of neural progenitor cells in the retina and brain. It remains unclear whether the lack of ephrin-A2/A3 is sufficient to promote the neurogenic potential of MC. Here we investigated whether the MC is the primary retinal cell type expressing ephrin-A2/A3 and their role on the neurogenic potential of Müller cells. In this study, we showed that ephrin-A2/A3 and their receptor EphA4 were expressed in retina and especially enriched in MC. The level of ephrinAs/EphA4 expression increased as the retina matured that is correlated with the reduced proliferative and progenitor cell potential of MC. Next, we investigated the proliferation in primary MC cultures isolated from wild-type and A2–/– A3–/– mice by 5-ethynyl-2′-deoxyuridine (EdU) incorporation. We detected a significant increase of EdU+ cells in MC derived from A2–/– A3–/– mice. Next, we investigated the role of ephrin-A2/A3 in mice undergoing photoreceptor degeneration such as Rhodopsin knockout (Rho–/–) mice. To further evaluate the role of ephrin-A2/A3 in MC proliferation in vivo, EdU was injected intraperitoneally to adult wild-type, A2–/– A3–/– , Rho–/– and Rho–/– A2–/– A3–/–  mice and the numbers of EdU+ cells distributed among different layers of the retina. EphrinAs/EphA4 expression was upregulated in the retina of Rho–/– mice compared to the wild-type mice. In addition, cultured MC derived from ephrin-A2–/– A3–/– mice also expressed higher levels of progenitor cell markers and exhibited higher proliferation potential than those from wild-type mice. Interestingly, we detected a significant increase of EdU+ cells in the retinas of adult ephrin-A2–/– A3–/– mice mainly in the inner nuclear layer; and these EdU+ cells were co-localized with MC marker, cellular retinaldehyde-binding protein, suggesting some proliferating cells are from MC. In Rhodopsin knockout mice (Rho–/– A2–/– A3–/– mice), a significantly greater amount of EdU+ cells were located in the ciliary body, retina and RPE than that of Rho–/–  mice. Comparing between 6 and 12 weeks old Rho–/– A2–/– A3–/– mice, we recorded more EdU+ cells in the outer nuclear layer in the 12-week-old mice undergoing severe retinal degeneration. Taken together, Ephrin-A2/A3 are negative regulators of the proliferative and neurogenic potentials of MC. Absence of ephrin-A2/A3 promotes the migration of proliferating cells into the outer nuclear layer and may lead to retinal cell regeneration. All experimental procedures were approved by the Animal Care and Use Committee at Schepens Eye Research Institute, USA (approval No. S-353-0715) on October 24, 2012.
    Related Articles | Metrics
    Utility of somatosensory and motor-evoked potentials in reflecting gross and fine motor functions after unilateral cervical spinal cord contusion injury
    Rong Li, Zu-Cheng Huang, Hong-Yan Cui, Zhi-Ping Huang, Jun-Hao Liu, Qing-An Zhu, Yong Hu
    2021, 16 (7):  1323-1330.  doi: 10.4103/1673-5374.301486
    Abstract ( 198 )   PDF (2055KB) ( 146 )   Save
    Fine motor skills are thought to rely on the integrity of ascending sensory pathways in the spinal dorsal column as well as descending motor pathways that have a neocortical origin. However, the neurophysiological processes underlying communication between the somatosensory and motor pathways that regulate fine motor skills during spontaneous recovery after spinal cord contusion injury remain unclear. Here, we established a rat model of cervical hemicontusive injury using C5 laminectomy followed by contusional displacement of 1.2 mm (mild injury) or 2.0 mm (severe injury) to the C5 spinal cord. Electrophysiological recordings were performed on the brachial muscles up to 12 weeks after injury to investigate the mechanisms by which spinal cord pathways participate in motor function. After spinal cord contusion injury, the amplitudes of somatosensory and motor-evoked potentials were reduced, and the latencies were increased. The forelimb open field locomotion test, grooming test, rearing test and Montoya staircase test revealed improvement in functions. With increasing time after injury, the amplitudes of somatosensory and motor-evoked potentials in rats with mild spinal cord injury increased gradually, and the latencies gradually shortened. In comparison, the recovery times of somatosensory and motor-evoked potential amplitudes and latencies were longer, and the recovery of motor function was delayed in rats with severe spinal cord injury. Correlation analysis revealed that somatosensory-evoked potential and motor-evoked potential parameters were correlated with gross and fine motor function in rats with mild spinal cord contusion injury. In contrast, only somatosensory-evoked potential amplitude was correlated with fine motor skills in rats with severe spinal cord injury. Our results show that changes in both somatosensory and motor-evoked potentials can reflect the changes in gross and fine motor functions after mild spinal cord contusion injury, and that the change in somatosensory-evoked potential amplitude can also reflect the change in fine motor function after severe spinal cord contusion injury. This study was approved by the Animal Ethics Committee of Nanfang Hospital, Southern Medical University, China (approval No. NFYY-2017-67) on June 11, 2017.
    Related Articles | Metrics
    Ninjurin-1: a biomarker for reflecting the process of neuroinflammation after spinal cord injury
    Poornima D. E. Weerasinghe-Mudiyanselage, Jeongtae Kim, Yuna Choi, Changjong Moon, Taekyun Shin, Meejung Ahn
    2021, 16 (7):  1331-1335.  doi: 10.4103/1673-5374.301033
    Abstract ( 93 )   PDF (2843KB) ( 262 )   Save
    Previous studies have shown that Ninjurin-1 participates in cell trafficking and axonal growth following central and peripheral nervous system neuroinflammation. But its precise roles in these processes and involvement in spinal cord injury pathophysiology remain unclear. Western blot assay revealed that Ninjurin-1 levels in rats with spinal cord injury exhibited an upregulation until day 4 post-injury and slightly decreased thereafter compared with sham controls. Immunohistochemistry analysis revealed that Ninjurin-1 immunoreactivity in rats with spinal cord injury sharply increased on days 1 and 4 post-injury and slightly decreased on days 7 and 21 post-injury compared with sham controls. Ninjurin-1 immunostaining was weak in vascular endothelial cells, ependymal cells, and some glial cells in sham controls while it was relatively strong in macrophages, microglia, and reactive astrocytes. These findings suggest that a variety of cells, including vascular endothelial cells, macrophages, and microglia, secrete Ninjurin-1 and they participate in the pathophysiology of compression-induced spinal cord injury. All experimental procedures were approved by the Care and Use of Laboratory Animals of Jeju National University (approval No. 2018-0029) on July 6, 2018.
    Related Articles | Metrics
    Efficacy of short-term multidisciplinary intensive rehabilitation in patients with different Parkinson’s disease motor subtypes: a prospective pilot study with 3-month follow-up 
    Ke-Ke Chen, Zhao-Hui Jin, Lei Gao, Lin Qi, Qiao-Xia Zhen, Cui Liu, Ping Wang, Yong-Hong Liu, Rui-Dan Wang, Yan-Jun Liu, Jin-Ping Fang, Yuan Su, Xiao-Yan Yan, Ai-Xian Liu, Bo-Yan Fang
    2021, 16 (7):  1336-1343.  doi: 10.4103/1673-5374.301029
    Abstract ( 122 )   PDF (856KB) ( 260 )   Save
    Parkinson’s disease (PD) can be classified into three motor-based subtypes: postural instability/gait difficulty (PIGD), tremor dominant (TD), and indeterminate. The neuropathophysiological mechanisms of the three motor subtypes are different, which may lead to different responses to therapy. Sixty-nine patients with idiopathic Parkinson’s disease (Hoehn–Yahr stage ≤ 3) were screened from 436 patients with Parkinsonism recruited through outpatient services and the internet. According to the Movement Disorder Society Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) TD/PIGD ratio, the patients were divided into PIGD (TD/PIGD ≤ 0.09; n = 36), TD (TD/PIGD ≥1.15; n = 19), and indeterminate (TD/PIGD = 0.90–1.15; n = 14) groups. All patients received 2 weeks of multidisciplinary intensive rehabilitation treatment (MIRT) during hospitalization, as well as a remote home rehabilitation health education class. Compared with the scores at admission, all patients showed significant improvements in their MDS-UPDRS III score, walking ability, balance, and posture control at discharge. Moreover, the MDS-UPDRS III score improvement was greater in the PIGD group than in the TD group. The follow-up data, collected for 3 months after discharge, showed that overall symptom improvement in each group was maintained for 1–3 months. Furthermore, there were no significant differences in the duration or grade effects of symptom improvement among the three groups. These findings suggest that 2 weeks of MIRT is effective for improving motor performance in all three motor subtypes. Patients in the PIGD group had a better response after hospitalization than those in the TD group. This study was approved by the Institutional Ethics Committee of Beijing Rehabilitation Hospital of Capital Medical University of China (approval No. 2018bkky022) on May 7, 2018 and registered with the Chinese Clinical Trial Registry (registration No. ChiCTR1900020771) on January 19, 2019.
    Related Articles | Metrics
    Quercetin protects against diabetic retinopathy in rats by inducing heme oxygenase-1 expression
    Guang-Rui Chai, Shu Liu, Hong-Wei Yang, Xiao-Long Chen
    2021, 16 (7):  1344-1350.  doi: 10.4103/1673-5374.301027
    Abstract ( 198 )   PDF (3120KB) ( 114 )   Save
    Quercetin is a widely-occurring flavonoid that protects against cancer, and improves memory and cardiovascular functions. However, whether quercetin exhibits therapeutic effects in diabetic retinopathy remains unclear. In this study, we established a rat model of streptozocin-induced diabetic retinopathy. Seventy-two hours later, the rats were intraperitoneally administered 150 mg/kg quercetin for 16 successive weeks. Quercetin markedly increased the thickness of the retinal cell layer, increased the number of ganglion cells, and decreased the overexpression of the pro-inflammatory factors interleukin-1β, interleukin-18, interleukin-6 and tumor necrosis factor-α in the retinal tissue as well as the overexpression of high mobility group box-1 and the overactivation of the NLRP3 inflammasome. Furthermore, quercetin inhibited the overexpression of TLR4 and NF-κBp65, reduced the expression of the pro-angiogenic vascular endothelial growth factor and soluble intercellular adhesion molecule-1, and upregulated the neurotrophins brain-derived neurotrophic factor and nerve growth factor. Intraperitoneal injection of the heme oxygenase-1 inhibitor zinc protoporphyrin blocked the protective effect of quercetin. These findings suggest that quercetin exerts therapeutic effects in diabetic retinopathy possibly by inducing heme oxygenase-1 expression. This study was approved by the Animal Ethics Committee of China Medical University, China (approval No. 2016PS229K) on April 8, 2016.
    Related Articles | Metrics
    Central post-stroke pain due to injury of the medial lemniscus in a patient with medullary infarction
    Sung Ho Jang, Hyeok Gyu Kwon
    2021, 16 (7):  1351-1352.  doi: 10.4103/1673-5374.301036
    Abstract ( 100 )   PDF (701KB) ( 103 )   Save
    Central post-stroke pain (CPSP), which is neuropathic pain associated with a brain lesion following a stroke, was first described by Dejerine and Roussy in 1906 in a thalamic infarct patient (Dejerine and Roussy, 1906; Watson and Sandroni, 2016). Since then, many studies have suggested that the pathophysiological mechanism underlying CPSP is activated by injury to the somatosensory nervous system (Watson and Sandroni, 2016). There are two major pathways within the somatosensory nervous system: the spinothalamic tract (STT), which is chiefly responsible for pain and touch sensations, and the medial lemniscus (ML) pathway, which is mainly involved in proprioception and tactile discrimination (Naidich et al., 2009). Many studies have suggested that STT injury is a major pathogenetic mechanism of CPSP, but a few studies have suggested that ML injury is related to the development of CPSP (Kim and Choi-Kwon, 1999; Seghier et al., 2005; Kim, 2007; Hong et al., 2010; Jang et al., 2017).  
    Related Articles | Metrics
    Hydrogen sulfide enhances adult neurogenesis in a mouse model of Parkinson’s disease
    Min Wang, Juan-Juan Tang, Lin-Xiao Wang, Jun Yu, Li Zhang, Chen Qiao
    2021, 16 (7):  1353-1358.  doi: 10.4103/1673-5374.301026
    Abstract ( 150 )   PDF (1947KB) ( 119 )   Save
    Hydrogen sulfide (H2S) is regarded to be a protectant against diseases of the central nervous system and cardiovascular system. However, the mechanism by which H2S elicits neuroprotective effects in the progression of Parkinson’s disease (PD) remains unclear. To investigate the role of H2S in delaying the pathological process of PD, we used the most common sodium hydrosulfide (NaHS) as an H2S donor and established a mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/p) in the present study. Our results show that H2S reduced neuronal loss during the progression of PD. Notably, we found that H2S exhibited protective effects on dopaminergic neurons. Excitingly, H2S also increased the proliferation of neural stem cells in the subventricular zone. Next, we evaluated whether the neuroprotective effects of H2S on dopaminergic neurons in PD are dependent on adult nerve regeneration by treating primary adult neural stem cells cultured ex vivo with 1-methyl-4-phenylpyridine. Our results show that H2S could prevent nerve injury induced by 1-methyl-4-phenylpyridine, promote the growth of neurospheres, and promote neurogenesis by regulating Akt/glycogen synthase kinase-3β/β-catenin pathways in adult neural stem cells. These findings confirm that H2S can increase neurogenesis in an adult mouse model of PD by regulating the Akt/glycogen synthase kinase-3β/β-catenin signaling pathway. This study was approved by the Animal Care and Use Committee of Nanjing Medical University, China (IACUC Approval No. 1601153-3). 
    Related Articles | Metrics
    Dynamic glial response and crosstalk in demyelination-remyelination and neurodegeneration processes
    Tianci Chu, Lisa B.E. Shields, Wenxin Zeng, Yi Ping Zhang, Yuanyi Wang, Gregory N. Barnes, Christopher B. Shields, Jun Cai
    2021, 16 (7):  1359-1368.  doi: 10.4103/1673-5374.300975
    Abstract ( 94 )   PDF (352KB) ( 158 )   Save
    Multiple sclerosis is an autoimmune disease in which the immune system attacks the myelin sheath in the central nervous system. It is characterized by blood-brain barrier dysfunction throughout the course of multiple sclerosis, followed by the entry of immune cells and activation of local microglia and astrocytes. Glial cells (microglia, astrocytes, and oligodendrocyte lineage cells) are known as the important mediators of neuroinflammation, all of which play major roles in the pathogenesis of multiple sclerosis. Network communications between glial cells affect the activities of oligodendrocyte lineage cells and influence the demyelination-remyelination process. A finely balanced glial response may create a favorable lesion environment for efficient remyelination and neuroregeneration. This review focuses on glial response and neurodegeneration based on the findings from multiple sclerosis and major rodent demyelination models. In particular, glial interaction and molecular crosstalk are discussed to provide insights into the potential cell- and molecule-specific therapeutic targets to improve remyelination and neuroregeneration. 
    Related Articles | Metrics
    Pro- and anti-epileptic roles of microglia
    Shinichi Kinoshita, Ryuta Koyama
    2021, 16 (7):  1369-1371.  doi: 10.4103/1673-5374.300976
    Abstract ( 201 )   PDF (696KB) ( 168 )   Save
    Microglia are brain-resident immune cells that contribute to the maintenance of brain homeostasis. In the epileptic brain, microglia show various activation phenotypes depending on the stage of epileptogenesis. Therefore, it remains unclear whether microglial activation acts in a pro-epileptic or anti-epileptic manner. In mesial temporal lobe epilepsy, one of the most common form of epilepsies, microglia exhibit at least two distinct morphologies, amoeboid shape and ramified shape. Amoeboid microglia are often found in sclerotic area, whereas ramified microglia are mainly found in non-sclerotic area; however, it remains unclear whether these structurally distinct microglia share separate roles in the epileptic brain. Here, we review the roles of the two distinct microglial phenotypes, focusing on their pro- and anti-epileptic roles in terms of inflammatory response, regulation of neurogenesis and microglia-neuron interaction. 
    Related Articles | Metrics
    Hydroxyethylstarch revisited for acute brain injury treatment
    Martin A. Schick, Malgorzata Burek, Carola Y. Förster, Michiaki Nagai, Christian Wunder, Winfried Neuhaus
    2021, 16 (7):  1372-1376.  doi: 10.4103/1673-5374.300978
    Abstract ( 87 )   PDF (1212KB) ( 128 )   Save
    Infusion of the colloid hydroxyethylstarch has been used for volume substitution to maintain hemodynamics and microcirculation after e.g., severe blood loss. In the last decade it was revealed that hydroxyethylstarch can aggravate acute kidney injury, especially in septic patients. Because of the serious risk for critically ill patients, the administration of hydroxyethylstarch was restricted for clinical use. Animal studies and recently published in vitro experiments showed that hydroxyethylstarch might exert protective effects on the blood-brain barrier. Since the prevention of blood-brain barrier disruption was shown to go along with the reduction of brain damage after several kinds of insults, we revisit the topic hydroxyethylstarch and discuss a possible niche for the application of hydroxyethylstarch in acute brain injury treatment. 
    Related Articles | Metrics
    Progress in clinical trials of stem cell therapy for cerebral palsy
    Zhong-Yue Lv, Ying Li, Jing Liu
    2021, 16 (7):  1377-1382.  doi: 10.4103/1673-5374.300979
    Abstract ( 131 )   PDF (871KB) ( 155 )   Save
    Cerebral palsy is the most common disease in children associated with lifelong disability in many countries. Clinical research has demonstrated that traditional physiotherapy and rehabilitation therapies cannot alone cure cerebral palsy. Stem cell transplantation is an emerging therapy that has been applied in clinical trials for a variety of neurological diseases because of the regenerative and unlimited proliferative capacity of stem cells. In this review, we summarize the design schemes and results of these clinical trials. Our findings reveal great differences in population characteristics, stem cell types and doses, administration methods, and evaluation methods among the included clinical trials. Furthermore, we also assess the safety and efficacy of these clinical trials. We anticipate that our findings will advance the rational development of clinical trials of stem cell therapy for cerebral palsy and contribute to the clinical application of stem cells.
    Related Articles | Metrics
    Oxidative stress factors in Parkinson’s disease
    Jolanta Dorszewska, Marta Kowalska, Michał Prendecki, Thomas Piekut, Joanna Kozłowska, Wojciech Kozubski
    2021, 16 (7):  1383-1391.  doi: 10.4103/1673-5374.300980
    Abstract ( 146 )   PDF (417KB) ( 182 )   Save
    Parkinson’s disease (PD) is the second most common cause of neurodegeneration. Over the last two decades, various hypotheses have been proposed to explain the etiology of PD. Among these is the oxidant-antioxidant theory, which asserts that local and systemic oxidative damage triggered by reactive oxygen species and other free radicals may promote dopaminergic neuron degeneration. Excessive reactive oxygen species formation, one of the underlying causes of pathology in the course of PD has been evidenced by various studies showing that oxidized macromolecules including lipids, proteins, and nucleic acids accumulate in brain tissues of PD patients. DNA oxidation may produce various lesions in the course of PD. Mutations incurred as a result of DNA oxidation may further enhance reactive oxygen species production in the brains of PD patients, exacerbating neuronal loss due to defects in the mitochondrial electron transport chain, antioxidant depletion, and exposure to toxic oxidized dopamine. The protein products of SNCA, PRKN, PINK1, DJ1, and LRRK2 genes are associated with disrupted oxidoreductive homeostasis in PD. SNCA is the first gene linked with familial PD and is currently known to be affected by six mutations correlated with the disorder: A53T, A30P, E46K, G51D, H50Q and A53E. PRKN encodes Parkin, an E3 ubiquitin ligase which mediates the proteasome degradation of redundant and disordered proteins such as glycosylated α-synuclein. Over 100 mutations have been found among the 12 exons of PRKN. PINK1, a mitochondrial kinase highly expressed in the brain, may undergo loss of function mutations which constitute approximately 1–8% of early onset PD cases. More than 50 PD-promoting mutations have been found in PINK1. Mutations in DJ-1, a neuroprotective protein, are a rare cause of early onset PD and constitute only 1% of cases. Around 20 mutations have been found in DJ1 among PD patients thus far. Mutations in the LRRK2 gene are the most common known cause of familial autosomal dominant PD and sporadic PD. Treatment of PD patients, especially in the advanced stages of the disease, is very difficult. The first step in managing progressive PD is to optimize dopaminergic therapy by increasing the doses of dopamine agonists and L-dopa. The next step is the introduction of advanced therapies, such as deep brain stimulation. Genetic factors may influence the response to L-dopa and deep brain stimulation therapy and the regulation of oxidative stress. Consequently, research into minimally invasive surgical interventions, as well as therapies that target the underlying etiology of PD is warranted.
    Related Articles | Metrics
    Genes associated with Alzheimer’s disease affecting ischemic neurodegeneration of the hippocampal CA3 region
    Ryszard Pluta, Marzena Ułamek-Kozioł
    2021, 16 (7):  1392-1393.  doi: 10.4103/1673-5374.300982
    Abstract ( 81 )   PDF (595KB) ( 88 )   Save
    Neurodegeneration in the brain after ischemia with reperfusion mimicking the neuropathology of Alzheimer’s disease: Brain ischemia with reperfusion, which is one of the main causes of morbidity and mortality in the world, triggers various neuropathological changes characteristic for Alzheimer’s disease (AD) such as increased blood-brain barrier permeability, excitotoxicity, necrosis, autophagy, mitophagy, apoptosis, neuroinflammation, amyloid plaques, neurofibrillary tangles, cerebral vessel pathology, and brain atrophy that lead to the death of neurons, deteriorating motor, sensory and cognitive functions (Figure 1) (Kato et al., 1988; Wisniewski et al., 1995; Van Groen et al., 2005; Kocki et al., 2015; Ułamek-Kozioł et al., 2016, 2017, 2019). Brain ischemia is recognized as a major contributor to the dysfunction of an aging brain and the development of neurodegenerative diseases, including AD (Pluta, 2019). The explanation of the final mechanisms of post-ischemic neurodegeneration progress and etiology of AD as well as the development of causal treatment for both diseases seems to involve complicated and complex procedures which require endless revision (Kametani and Hasegawa, 2018; Pluta, 2019). This is partly due to the fact that neurodegenerative processes leading to dementia after both ischemia and AD are not well understood yet. Therefore, there is no causal treatment or adequate criterion for early diagnosis of dementia. Recently, an etiological link has been proposed between dementia following ischemia and dementia due to Alzheimer’s disease (Figure 1) (Salminen et al., 2017; Pluta, 2019). Our article presents facts supporting the idea that neuropathological mechanisms after cerebral ischemia contribute to the development of the genotype and phenotype of AD. The main goal is to broaden knowledge about the overall ischemia processes underlying neuronal death and their impact on regeneration and functional recovery during neurodegeneration of the AD type, and what follows, their relationship with neuronal processes involved in the possible development of AD (Figure 1). Therefore, the main challenge of this new research strategy is to identify genomic and proteomic changes that are common for both ischemia and AD and can be cured. In particular, we are looking for evidence linking the expression of genes associated with AD after ischemia with their role in the regulation of AD proteins in the ischemic brain. Furthermore, we try to understand the role of expression of AD genes along with their proteins during the clinical progress and maturation of post-ischemic brain neurodegeneration with the association of the genotype and phenotype of AD. In this perspective, we combine the importance of cerebral ischemia-induced AD genes expression with a presentation of the final relevant ischemic, genetic mechanism of protein placement in the brain and its reference to the progression of AD. These findings are likely to lead to the development of much-needed new causal therapies and early dementia diagnostic methods in both incurable diseases.
    Related Articles | Metrics
    Potential contributions of trace amines in Alzheimer’s disease and therapeutic prospects
    Sudip Dhakal, Ian Macreadie
    2021, 16 (7):  1394-1396.  doi: 10.4103/1673-5374.300985
    Abstract ( 138 )   PDF (842KB) ( 137 )   Save
    Trace amines and their biochemistry: Trace amines represent endogenous monoamines present in very low concentration (usually nM) in the brain in comparison to classical monoamine neurotransmitters. These trace amines can regulate the amount of the classical monoamine neurotransmitters in the synaptic cleft. Trace amines that have crucial roles in neuromodulation and neurotransmission include p-tyramine, β-phenylethylamine, tryptamine, 3-methoxytyramine and their derivatives including octopamine, synephrine, N-methyltyramine, N-methyltryptamine and N-methylphenethylamine. The decarboxylation of aromatic L-amino acids by aromatic amino acid decarboxylase (AAAD) (EC 4.1.1.28) is the primary source of these trace amines in cell (Gainetdinov et al., 2018). The distribution of AAAD throughout the body varies between tissues. The decarboxylation of tyrosine, phenylalanine and tryptophan leads to formation of tyramine, β-phenylethylamine and tryptamine, respectively (Figure 1A and B). Although initially thought to have no pathological and physiological relevance, trace amines proved to be more important with the discovery of trace amine associated receptors (TAARs). These trace amines have been reported to play a crucial role in regulating dopamine and serotonin levels in the synaptic cleft by signaling through TAARs. TAARs are Type A G-protein coupled receptors, which become selectively activated by p-tyramine and β-phenylethylamine but not by the classical monoamine neurotransmitters. In the meantime, other trace amines including tryptamine and octopamine also interact with TAARs as agonists (Berry, 2016). 
    Related Articles | Metrics
    Leukocyte telomere length and plasma interleukin-1β and interleukin-18 levels in mild cognitive impairment and Alzheimer’s disease: new biomarkers for diagnosis and disease progression?
    Rosa Maria Corbo, Rita Businaro, Daniela Scarabino
    2021, 16 (7):  1397-1398.  doi: 10.4103/1673-5374.300986
    Abstract ( 99 )   PDF (370KB) ( 100 )   Save
    Alzheimer’s disease and mild cognitive impairment biomarkers: Alzheimer’s disease (AD) is a progressive neurodegenerative disease of advancing age. It affects around 47 million people in the world and the number is estimated to increase to 152 million by 2050. Someone develops dementia every three seconds and the current annual cost of dementia is estimated at US $1 trillion, a figure set to double by 2030 (Alzheimer’s Disease International, 2019).
    Related Articles | Metrics
    The effects of amyloid-beta on hippocampal glutamatergic receptor and transporter expression
    Andrea Kwakowsky, Henry J. Waldvogel, Richard LM Faull
    2021, 16 (7):  1399-1401.  doi: 10.4103/1673-5374.301009
    Abstract ( 82 )   PDF (479KB) ( 100 )   Save
    The leading form of dementia worldwide, Alzheimer’s disease (AD) is a common neurodegenerative disorder. The underlying causes of AD are not well understood, and no current treatments are preventing the onset or delay progression of the disease. Currently, most investigation is directed towards the amyloid-beta (Aβ) and tau pathologies, yet there are many other underlying processes that have been implicated to contribute directly to AD progression. One such phenomenon is glutamatergic excitotoxicity, a loss of neuromodulatory balance inducing a hyper-excitable neuronal state, leading to cell death across several brain regions (Zhang et al., 2016; Bukke et al., 2020). Glutamate is the primary excitatory neurotransmitter in the brain and is involved in many critical signaling and metabolic functions but control of the glutamatergic system requires constant moderation to avoid excitotoxicity occurring (Bukke et al., 2020). As yet, glutamatergic signaling changes that contribute to this process, or result due to this process, have not been thoroughly investigated. 
    Related Articles | Metrics
    TDP-43 and amyloid precursor protein processing: implications for Alzheimer’s disease
    David A. Hicks
    2021, 16 (7):  1402-1403.  doi: 10.4103/1673-5374.300983
    Abstract ( 64 )   PDF (633KB) ( 100 )   Save
    In this perspective article, I will discuss our recent publication (Hicks et al., 2020), specifically its major findings and integration with the published literature. Alzheimer’s disease (AD) is a progressive neurodegenerative disease of predominantly unknown aetiology. Its neuropathological hallmarks are extracellular plaques formed of the amyloid beta (Aβ) peptide and neurofibrillary tangles of tau protein. Aβ peptides (Aβ1–40 and Aβ1–42 are considered the most important) are generated by sequential proteolysis of the amyloid precursor protein (APP). This proteolysis can take one of two pathways: the nonamyloidogenic or amyloidogenic (Figure 1). The former is mediated by α-secretase, predominantly a disintegrin and metalloprotease domain-containing 10 (ADAM10) and the γ-secretase complex. This pathway releases the APP ectodomain as sAPPα, but crucially, results in cleavage of APP within the Aβ region, abrogating its generation. However, the amyloidogenic pathway is mediated by β-site APP cleaving enzyme 1 (BACE1, also known as β-secretase) and the γ-secretase complex, which results in the generation of Aβ species and the amyloid precursor protein intracellular domain. The amyloid precursor protein intracellular domain (AICD) has been shown to traffic to the nucleus where it can act as a transcriptional modifier. Although there are other minor APP secretases, those outlined herein represent the most significant. Alterations in APP processing have been linked to AD, insofar as the Aβ1–42: Aβ1–40 ratio is increased in the disease (Andrew et al., 2016). 
    Related Articles | Metrics
    Heparan sulfate proteoglycans as possible diagnostic molecular tools with therapeutic potential in Alzheimer´s disease
    Iván Fernández-Vega, Laura Lorente-Gea, Carla Martín, Luis M. Quirós
    2021, 16 (7):  1404-1405.  doi: 10.4103/1673-5374.301056
    Abstract ( 92 )   PDF (725KB) ( 81 )   Save
    Importance of dementia and Alzheimer’s disease (AD): On Earth, there is a “country” that we call Dementia with around 50 million inhabitants and an estimated global economic cost of US $1 trillion (Wimo et al., 2017). AD may contribute to 60–70% of cases, with other major forms of dementia being vascular dementia, dementia with Lewy bodies and a group of diseases that contribute to frontotemporal dementia. However, the boundaries between different forms of dementia are indistinct and mixed forms often co-exist, which are only confirmed after postmortem neuropathological studies (Fernandez-Vega et al., 2015). 
    Related Articles | Metrics
    Dopaminergic-cholinergic imbalance in movement disorders: a role for the novel striatal dopamine D2- muscarinic acetylcholine M1 receptor heteromer
    René A. J. Crans, Francisco Ciruela
    2021, 16 (7):  1406-1408.  doi: 10.4103/1673-5374.300988
    Abstract ( 92 )   PDF (1028KB) ( 144 )   Save
    The striatum is the primary input structure of the basal ganglia, which participates in motivational and goal-directed behaviors (Pisani et al., 2007). In physiological conditions, local cholinergic interneurons (ChIs) and dopaminergic afferents modulate basal ganglia output through striatal projection neurons, also called medium spiny neurons (MSNs). In general, the release of the neurotransmitters dopamine (DA) and acetylcholine (ACh) elicits contradictory effects on MSNs, which express their corresponding DA receptors (DARs) and muscarinic acetylcholine receptors (mAChRs), respectively (Ztaou and Amalric, 2019). Recently, we discovered a novel receptor-receptor interaction (i.e., heteromerization) between the dopamine D2 receptor (D2R) and the muscarinic acetylcholine M1 receptor (M1R), both expressed at striatopallidal MSNs (Crans et al., 2020). The putative striatal D2R-M1R complex coordinates a sophisticated interplay between the dopaminergic and cholinergic neurotransmission systems. Fuxe et al. (2012) foresaw that the existence of this heteromer within the striatum would mechanistically justify the use of anticholinergics in Parkinson’s disease (PD) treatment, thus opening up the development of novel pharmacotherapeutic strategies for PD management. As a proof of concept, we demonstrated that an M1R-selective antagonist (i.e., VU0255035, 10 mg/kg, i.p.) potentiated the antiparkinsonian-like efficacy of an ineffective D2R-selective agonist dose (i.e., sumanirole, 3 mg/kg, i.p.) in a rodent model of experimental Parkinsonism (Crans et al., 2020). Overall, the novel D2R-M1R heteromer could serve as a specific drug target to alleviate motor deficits in PD, whereas it may avoid major adverse effects associated with traditional pharmacotherapies. 
    Related Articles | Metrics
    Application of flavonoids for the protection of nigral dopaminergic neurons from oxidative stress
    Sang Ryong Kim
    2021, 16 (7):  1409-1410.  doi: 10.4103/1673-5374.301010
    Abstract ( 80 )   PDF (451KB) ( 83 )   Save
    Oxidative stress in Parkinson’s disease (PD): Oxidative stress is the imbalance between oxidants, which generate reactive oxygen species (ROS; free radicals), and antioxidants, which remove free radicals. Under healthy conditions, the levels of oxidants and antioxidants are well-balanced. However, excessive production of ROS and a deficiency of antioxidants leads to oxidative stress, which may be the cause of accelerating the development of neurodegenerative diseases (Hwang, 2013), suggesting that the prevention of ROS production and reduction of oxidative stress is critical for both the prevention and treatment of neurodegenerative diseases, including PD.
    Related Articles | Metrics
    Novel perspectives for neurodegeneration prevention: effects of bioactive polyphenols
    Stefania Crispi, Stefania Filosa
    2021, 16 (7):  1411-1412.  doi: 10.4103/1673-5374.300989
    Abstract ( 80 )   PDF (359KB) ( 126 )   Save
    Neurodegenerative diseases are becoming a big challenge for modern society. Neurodegenerative disorders strongly impact on patient and their caregivers. Moreover, since the population is becoming older, these pathologies will deeply influence medical and socio-economic conditions in the next years. Therefore, efforts are needed to find new strategies devoted to define new protocols and identify novel substances able to prevent neurodegeneration or to improve the quality of life of people affected by neurodegenerative diseases (Alzheimer’s Disease International, 2019).
    Related Articles | Metrics
    Why use pre-differentiated cells to address complex multi-factorial neurodegenerative diseases?
    Alex Kopyov, Toni L. Uhlendorf, Randy W. Cohen
    2021, 16 (7):  1413-1414.  doi: 10.4103/1673-5374.30099
    Abstract ( 87 )   PDF (747KB) ( 104 )   Save
    Past four decades have seen a concerted push to develop regenerative treatments for incurable neurodegenerative diseases such as Parkinson’s disease (PD). PD’s pathophysiology is primarily characterized by dopamine deficiency caused by the progressive depletion of neurons in substantia nigra (Bernheimer et al., 1973). So, it is not very surprising that most attempts at regenerative treatment of PD focus on using pre-differentiated, dopaminergic neurons. However, PD’s pathogenesis spans far beyond strictly dopamine deficiency and is still not completely known (Chaudhuri et al., 2006). Thus, attempts at implanting pre-differentiated dopaminergic neurons that are locked into a single, inflexible function, predictably ran into the same problems as dopamine replacement therapy: 1) These cells are not curative; 2) They cannot address all PD deficits, and 3) They tend to cause side effects (Kordower et al., 2017). In order to address the multifactorial nature of this disease, we suggest the use of non-tumorigenic undifferentiated stem cells. Unlike adult cells, undifferentiated cells, due to their inherent plasticity, have the potential to respond to microenvironmental cues (Martínez-Cerdeño et al., 2017) from the Parkinsonian brain, target multiple systems and pathways, and eventually restore both structure and function (Figure 1). 

    Related Articles | Metrics
    A13 dopamine cell group in the zona incerta is a key neuronal nucleus in nociceptive processing
    Shunpei Moriya, Tomoyuki Kuwaki
    2021, 16 (7):  1415-1416.  doi: 10.4103/1673-5374.300991
    Abstract ( 132 )   PDF (805KB) ( 95 )   Save
    In stressful modern society, pain caused by physical and mental disorders is an increasing social health problem all over the world. Physical and mental activity depends on the state of various neurotransmitters in the central nervous system (CNS). Monoaminergic systems play important roles in regulating physiological functions including nociception. It is well known that in the CNS, there are descending pathways related to nociceptive processing known as the descending antinociceptive system (DAS). Noradrenalin and serotonin are major components of the DAS that form the locus coeruleus (LC)-spinal dorsal horn noradrenergic circuit and periaqueductal gray (PAG)-rostro ventricular medulla (RVM)-spinal dorsal horn serotonergic circuit (Jordan et al., 2008). Dopaminergic pathways also play roles in regulating nociceptive processing in the CNS.
    Related Articles | Metrics
    Glial derived neurotrophic factor: a sufficient essential molecular regulator of mammalian blood-nerve barrier tight junction formation
    Chaoling Dong, Aarti Choudhary, Eroboghene E. Ubogu
    2021, 16 (7):  1417-1418.  doi: 10.4103/1673-5374.300992
    Abstract ( 67 )   PDF (1474KB) ( 87 )   Save
    Peripheral nerves coordinate signal transduction from the periphery to the central nervous system for processing and transmission back as required for normal mammalian function. Peripheral nerves and nerve roots are structurally divided into three compartments: the outermost epineurium, inner perineurium that surrounds nerve fascicles and the innermost endoneurium (Mizisin and Weerasuriya, 2011). As with all organs, peripheral nerve vascularization occurs during development and is maintained in health. Adaptations are expected dependent on tissue-specific functions and physiologic state. The peripheral nerve internal microenvironment is tightly controlled to facilitate coordinated and regulated axonal transmission. Peripheral nerves and nerve roots are perfused by extrinsic blood vessels called the vasa nervorum. These macrovessels penetrate the outer epineurium to give rise to epineurial arteries and arterioles and receive blood from epineurial venules and veins. Macrovessels subsequently transverse the inner perineurium, formed by multiple concentric layers of specialized epithelioid myofibroblasts, to perfuse the innermost endoneurium, in which reside myelinated and unmyelinated axons in a loose array of collagen fibers. Non-fenestrated tight junction-forming capillary-like microvessels exist within the endoneurium which are in direct contact with circulating blood. Thus, these microvessels are considered to form the blood-nerve barrier (BNB) (Ubogu, 2020). Tight junction-forming perineurial myofibroblasts provide a critical interface between the endoneurial and epineurial interstitial fluid compartments that further regulate the endoneurial microenvironment; however, these cells are not in direct contact with circulating blood. Peripheral neuropathies affect over 100 million people worldwide, and a common consequence of peripheral nerve disease is chronic pain, which may affect as many as 1% of individuals during their lifetimes. Understanding peripheral nerve molecular and biophysical microvascular adaptations and BNB function during development and in normal physiological states, as well as in diseases, including peripheral nerve injury, provides an avenue to understand how peripheral nerve regeneration and homeostatic restoration may occur. These processes could be harnessed for therapeutic development in peripheral neuropathies and chronic neuropathic pain to support neural regeneration and restore normal biological function. 
    Related Articles | Metrics
    The role of dietary nitrate supplementation in neurovascular function
    Masahiro Horiuchi, Gabriella M.K. Rossetti, Samuel J. Oliver
    2021, 16 (7):  1419-1420.  doi: 10.4103/1673-5374.300993
    Abstract ( 82 )   PDF (728KB) ( 103 )   Save
    A healthy diet combined with increased physical activity levels may protect against cerebrovascular dysfunction and cognitive impairments (Gorelick et al., 2011). Compared with the number of studies on the effects of physical activity, studies on how diet affects cerebral blood flow (CBF) regulation and cognitive function are relatively limited. Here, we focus on the potential role of dietary nitrate supplementation in CBF regulation and cognitive function under normoxic and hypoxic conditions.
    Related Articles | Metrics
    Astrocyte-derived extracellular vesicles mediate intercellular communications of the neurogliovascular unit
    Augustas Pivoriūnas, Alexei Verkhratsky
    2021, 16 (7):  1421-1422.  doi: 10.4103/1673-5374.300994
    Abstract ( 74 )   PDF (621KB) ( 114 )   Save
    The cortical grey matter of mammals has a specific cyto-architecture defined by the process of “tiling” in which protoplasmic astrocytes parcellate the nervous tissue into spatially segregated territorial domains. Within these domains astrocytes integrate neuronal structures, microglial cells and neighboring capillaries into the neuro-gliovascular unit or neurogliovascular unit (NGVU) (Verkhratsky and Nedergaard, 2018; Sweeney et al., 2019). Signaling within NGVU is of fundamental importance for sustaining brain function, while disruptions of these signaling pathways define many aspects of neuropathology. 
    Related Articles | Metrics
    Structural integrity and remodeling underlying functional recovery after stroke
    Frederique Wieters, Markus Aswendt
    2021, 16 (7):  1423-1424.  doi: 10.4103/1673-5374.301004
    Abstract ( 75 )   PDF (216KB) ( 85 )   Save
    Stroke is the second leading cause of death worldwide with about 50% of survivors being chronically disabled (Donkor, 2018). The behavioral improvement seen in stroke patients in the first weeks after a stroke is contributed by behavioral compensation, reorganization in somatotopic maps and activity in peri-infarct but also distant regions which are connected to the stroke area as supported by animal studies. Spontaneous recovery is related to region-specific changes in recovery-related genes (Ito et al., 2018), growth factor expression, axonal sprouting and dendritic spine turnover (Murphy and Corbett, 2009). These brain plasticity processes follow an intrinsic time-line with a limited period of heightened neuroplasticity for which the behavioral experience is a key modulator. However, experimental studies correlating structural and functional brain network changes during recovery are scarce and it remains controversial which mechanisms, regions and time points are most relevant and thus best suited for translational interventions. In that context, longitudinal in vivo imaging will be essential. MRI is particularly suited for repetitive, non-invasive imaging with high spatial resolution. Clinical stroke studies use routinely MRI in a standardized acquisition and post-processing regime for measuring the stroke size and location. In addition, structural connectivity analyses using diffusion MRI (dMRI) in the corticospinal and corticocortical fiber tracts effectively predict motor impairment and improvement, respectively (Koch et al., 2016). Stroke disrupts brain connectivity by primary mechanisms such as cell death and injury in white matter tracts, but also secondary mechanisms such as axonal degeneration which spreads to structurally connected cells (Cao et al., 2020). On the macroscopic level, dMRI, sensitive to tissue-specific water diffusion properties, offers the unique possibility to quantify microstructural changes, e.g. related to stroke-related processes of cell swelling, cell lysis and demyelination longitudinally on the whole brain level expressed in the dMRI measures of axial, radial, mean diffusivity, and fractional anisotropy. Furthermore, fiber tracking, a more complex post-processing and mathematical modeling of dMRI exploits the preferential diffusivity of protons along the myelin sheets to generate a quantitative measure of fiber tracts between brain regions. Experimental studies in rats and mice only recently evolved due to technical challenges such as the required signal-to-noise and susceptibility-induced image distortions (Hoehn and Aswendt, 2013). Based on pioneering work more than 20 years ago in a mouse model of reversible focal ischemia measuring the acute temporal dynamics of diffusion, so far, only ex vivo Diffusion Tensor Imaging (DTI) fiber tracking was applied in stroke mice to compare selected ipsi- vs. contralesional tracts (Granziera et al., 2007). 
    Related Articles | Metrics
    Phase-specific manipulation of neuronal activity: a promising stroke therapy approach
    Dennis A. Turner, Wei Yang
    2021, 16 (7):  1425-1426.  doi: 10.4103/1673-5374.301005
    Abstract ( 93 )   PDF (510KB) ( 85 )   Save
    Introduction: Ischemic stroke accounts for ~87% of all stroke cases (Virani et al., 2020). It is a leading cause of death and long-term disability worldwide, and constitutes a major burden for families and healthcare systems alike. Although medical treatment can help prevent stroke, post-stroke treatment is limited to either pharmacologic (e.g., tissue plasminogen activator - tPA) or mechanical (e.g., thrombectomy) reperfusion. During the past few decades, many neuroprotective and neurorestorative strategies have been tested in hopes of discovering improved treatment options for stroke patients, particularly patients who are not eligible for reperfusion therapy. Sadly, such hopes have not yet been fulfilled, and thus, patients are still in dire need of new stroke therapies as well as increased vigilance for amelioration of risk factors.  
    Related Articles | Metrics
    Role of N4-acetylcytidine for continuously activating NLRP3 inflammosome by HMGB1 pathway in microglia  
    Hua Bai, Qifang Zhang
    2021, 16 (7):  1427-1428.  doi: 10.4103/1673-5374.301006
    Abstract ( 79 )   PDF (270KB) ( 112 )   Save
    N4-acetylcytidine (N4A) is an organic compound and a metabolite of transferrable ribonucleic acid. Its molecular formula is C11H15N3O6. Preliminary studies suggest that N4A was mainly found on tRNA and 18S rRNA, while recent studies have shown that there is also a large amount of N4A on mRNA, whose abundance is not even lower than the m7G cap modification carried by mRNA (Arango et al., 2018). The generation of N4A is catalyzed by N-acetyltransferase 10 (NAT10) or its homologous enzyme. N4A is produced by acetylation in eukaryotic RNA and is the only human enzyme with both acetyltransferase and RNA binding activity (Arango et al., 2018). The full transcriptome mapping of N4A shows abundant discrete acetylation regions in the coding sequence. The ablation of NAT10 reduces the detection of N4A at mRNA localization sites and is globally correlated with the down-regulation of tmRNA. N4A is widely distributed in the human transcriptome, and most sites occur in the coding sequence. Compared with unmodified cytosine, N4A increases the thermal stability of Watson-Crick base pair guanosine, thus affecting the interaction with homologous tRNAs during translation. After the release of N4A from tRNA metabolism, it participates in the systematic immune response (Ito et al., 2014).
    Related Articles | Metrics
    Sustained neuronal viability by paracrine factors: new opportunities for endothelial progenitor cell secretome
    Stefano Di Santo, Hans Rudolf Widmer
    2021, 16 (7):  1429-1430.  doi: 10.4103/1673-5374.301007
    Abstract ( 126 )   PDF (425KB) ( 76 )   Save
    Despite the big progresses in the field of regenerative medicine, the loss of neurons remains essentially an unresolved challenge. Among the different approaches under investigation, there are great expectations on stem and progenitor cells-based strategies. Due to their capacity to home to the site of injury and theoretically generate all kinds of neuronal cells, stem cells seem the ideal candidates for targeted therapeutic interventions also in light of the lack of spontaneous regeneration of  the central nervous system. Unfortunately, the substantial failure to replace dead or dysfunctional cells has limited the development of stem cells-based therapies. This aspect is especially evident for the central nervous system due to the complex architecture of the tissue, the inherent fragility of mature neurons and the lack of methods to control and guide the proliferation and differentiation of neuronal precursors. However, there is compelling evidence that stem and progenitor cells are able to support different healing processes by means of paracrine actions. Taking advantage of these characteristics, researchers have investigated the cyto-protective and restorative activities of the factors released by in vitro cultured stem cells in the form of conditioned medium (CM).
    Related Articles | Metrics
    Disordered structure and flexible roles: using the prion protein N1 fragment for neuroprotective and regenerative therapy
    Behnam Mohammadi, Markus Glatzel, Hermann Clemens Altmeppen
    2021, 16 (7):  1431-1432.  doi: 10.4103/1673-5374.301008
    Abstract ( 112 )   PDF (547KB) ( 95 )   Save
    The cellular prion protein (PrPC) is a truly remarkable cell surface glycoprotein. With (i) its broad expression pattern and (ii) particularly high levels in the nervous system, (iii) its critical involvement in fatal neurodegenerative diseases affecting different mammalian species, (iv) its structurally diverging bipartite buildup, (v) its high degree of evolutionary conservation and (vi) a variety of –at least suggested– functions despite (vii) a surprising lack of major phenotypic deficits when absent (as in respective knock-out animals), PrPC has raised considerable research interest over the last four decades. While most of these aspects have been reviewed extensively in the past (Linsenmeier et al., 2017), this perspective will focus exclusively on a soluble peptide, termed N1, which is constitutively generated by the main proteolytic cleavage event occurring on PrPC (Figure 1B). In fact, considering that particular fragments of PrPC account for intrinsic functions, may help to explain the multitude of physiological roles so far mostly –and maybe in part mistakenly– attributed to full-length PrPC as the ‘precursor’. The N1 fragment basically consists of the flexible N-terminal half of PrPC (after removal of the signal peptide) ranging from residue 23 to ~110, contains several sites for coordinative binding of divalent cations and interaction with other binding partners, and represents a prime example of an intrinsically disordered peptide (Gonsberg et al., 2017). Physiologically it results from the α-cleavage of PrPC which may take place at or en route to the cell surface or after re-internalization in endosomal compartments. It is eventually released into the extracellular space and tissue/body fluids where it is expected to exert its functions. Of note, while candidates have been suggested and controversially discussed, the responsible protease has not been convincingly identified yet, thus precluding any pharmacological manipulation at present. It would not even be surprising if different proteases could orchestrate and ensure this important cleavage in a redundant fashion (Linsenmeier et al., 2017).
    Related Articles | Metrics
    A closer look at cannabimimetic terpenes, polyphenols, and flavonoids: a promising road forward#br#
    Juliana Cavalli, Rafael Cypriano Dutra
    2021, 16 (7):  1433-1435.  doi: 10.4103/1673-5374.301011
    Abstract ( 118 )   PDF (1215KB) ( 104 )   Save
    Despite the current interest in the potential medical use of Cannabis, it is worth remembering that cannabis is not a new drug, with both a nonmedical and medical history supporting its effectiveness. This history advanced in scientific strength and importance worldwide when Professor Raphael Mechoulam and colleagues initiated their pioneering discoveries in 1971. At first, Mechoulam et al. (1972) achieved the complete synthesis of the pure compounds from hashish (including ∆1-tetrahydrocannabinol and other neutral cannabinoids such as cannabigerol, cannabichromene, and cannabicyclol), and established their molecular structures. This set a strong pace for the study of their structure-activity relationship and started to pave a promising road of discovery! Later on, the most important compounds were isolated and identified, namely ∆9-tetrahydrocannabinol (∆9-THC), cannabidiol (CBD), and cannabinol, which represent some of the main tools used in preclinical and clinical research in the cannabinoid field. The endocannabinoid system (ECS) was proposed with the discovery of the endocannabinoids anandamide and 2-arachidonoylglycerol. With further progress in research, it became clear that the functions of the endocannabinoid signaling system are not limited to the brain, but are exerted throughout the organism. It has been proposed that endocannabinoids are released from cells as soon as their biosynthesis ends, avoiding release via secretory vesicles. Their actions on receptors are locally restricted, possibly due to their high lipophilicity and rapid inactivation under physiological conditions. In this scenario, the former would determine the promiscuity of cannabinoids in terms of their actions on different molecular targets, while the latter would affect the levels of cannabinoids since they are under the influence of biosynthesis and degradative enzymes. Both endocannabinoid compounds have been shown to mimic some effects of synthetic cannabinoids on their G-protein coupled receptors (CB1R and CB2R) and metabolizing enzymes. The G-protein coupled receptors CB1R (cloned in 1990) and CB2R (cloned in 1993) exhibit 48% similarity in their amino acid sequences. These receptors have been shown to exhibit particular differences in signaling mechanisms, tissue distribution and sensitivity to agonists and antagonists that depict marked binding selectivity between both receptors. When CBR is activated, adenylate cyclase is inhibited provoking the conversion inhibition of ATP to cyclic AMP. CB1R and CB2R located at peripheral, spinal, or supraspinal sites are important targets, mediating the effects of cannabinoids via the inhibition of presynaptic neurotransmitter and neuropeptide release, modulation of postsynaptic neuronal excitability, activation of the descending inhibitory pain pathway, and reductions in neuroinflammatory signaling (Starowicz and Finn, 2017). Other receptors have been reported to be activated by cannabinoid drugs and related molecules, including GPR55, GPR18, and GPR119. However, the CB1R has been considered a key component of the ECS since it is the most abundant metabotropic receptor in the brain and interacts with endogenous and exogenous cannabinoids, including ∆9-THC. Furthermore, there is a large body of evidence to demonstrate that CB1R and CB2R, as well as their ligands, play a significant role in physiological and pathological processes. Therefore, changes in endocannabinoids anandamide and 2-arachidonoylglycerol concentration, as well as activation or deactivation of both CBRs in the tissues have been widely studied, as they can be relevant for the modulation of neurological and neurodegenerative diseases, neuroinflammation, cancer, immune-mediated inflammatory diseases, and autoimmunity (Di Marzo, 2008). 
    Related Articles | Metrics
    Neuritogenic function of microglia in maternal immune activation and autism spectrum disorders
    Srinidhi Venkatesan Kalavai, Seiko Ikezu
    2021, 16 (7):  1436-1437.  doi: 10.4103/1673-5374.301012
    Abstract ( 75 )   PDF (589KB) ( 159 )   Save
    Autism spectrum disorder and maternal immune activation: Environmental factors during pregnancy, such as infections, maternal stress or autoimmune disorders, are closely associated with the prevalence of neurodevelopmental disorders including autism spectrum disorder (ASD), bipolar disorder and schizophrenia. It has been shown that severe infections during pregnancy cause maternal immune activation (MIA) and significantly increase the risk of ASD in the offspring although the mechanisms are poorly understood. Many rodent MIA studies support this causal link by showing that offspring of dams administered with polyinosinic:polycytidylic acid (polyI:C), a viral mimetic Toll-like receptor 3 agonist, exhibit long-lasting ASD-like behavioral abnormalities such as increased repetitive behavior, impaired social interaction and communication. Interestingly, MIA alters inflammatory cytokine expressions persisting through development and adulthood in the brain of the offspring, suggesting that chronic neuroimmune dysfunction plays a role in mediating the deleterious effects of MIA on neurodevelopment (Garay et al., 2013). 
    Related Articles | Metrics
    Clusterin: a multifaceted protein in the brain
    Hee-Jung Moon, Sarah K. Herring, Liqin Zhao
    2021, 16 (7):  1438-1439.  doi: 10.4103/1673-5374.301013
    Abstract ( 105 )   PDF (403KB) ( 112 )   Save
    Late-onset Alzheimer’s disease (LOAD), the most common cause of dementia, currently affects 5.6 million Americans ages 65 and older. LOAD is a neurodegenerative disorder characterized by progressive loss in synaptic function, notable bioenergetic decline, increased neuronal death and brain atrophy, and significant cognitive impairment. Because the etiology of LOAD remains unknown, a treatment for LOAD has not yet been formulated, a fact that is clearly demonstrated by the more than 200 failed clinical trials. These failures underscore the significance of identifying the LOAD risk mechanisms that would allow early intervention in the preclinical stage of LOAD. Genome-wide association studies have identified more than a dozen genetic risk variants that are associated with the development of LOAD. Clusterin (CLU), also known as apolipoprotein J (APOJ), has been established as the third most prominent genetic risk factor for LOAD after apolipoprotein E (APOE) and bridging integrator 1 (BIN1) (Harold et al., 2009; Lambert et al., 2009). A number of single nucleotide polymorphisms (SNPs) within the CLU locus, with the majority being intronic, have been linked to significantly altered LOAD risk, independent of APOE status (Figure 1A; (Medical Genetics and Human Variation, 2019)); however, it is unclear how these SNPs affect CLU mRNA, protein isoform expression and function. 
    Related Articles | Metrics
    The molecular profile of nerve repair: humans mirror rodents
    Matthew B. Wilcox, Kristjan R. Jessen, Tom J. Quick, James B. Phillips
    2021, 16 (7):  1440-1441.  doi: 10.4103/1673-5374.301014
    Abstract ( 81 )   PDF (468KB) ( 88 )   Save
    Peripheral nerve injuries (PNI) are common following blunt or penetrating trauma and can lead to disability and chronic pain in affected individuals, with limited options available to promote regeneration and functional recovery. From animal models, it is known that the regenerative capacity of the peripheral nervous system (PNS) is heavily dependent upon the remarkable ability of Schwann cells to undergo a phenotypic shift from a supportive/myelinating/maintaining phenotype to one that encourages neural regeneration. In rodents, a great deal is known about the molecular signals that control this process or mark the cells and cellular changes involved (Boerboom et al., 2017; Jessen and Mirsky, 2019). Effective translation of the wealth of animal model data into a human paradigm of nerve injury would be of great benefit in the development of improved clinical treatments. However, progress has been limited by ethical and practical challenges associated with studying human nerve injury (Hewitt et al., 2008; Wilcox et al., 2019). Moreover, the intricate anatomy and diverse range of injuries make PNI a heterogeneous pathology to study. To address this issue, in our recent article entitled Characterizing cellular and molecular features of human peripheral nerve degeneration, we analyzed nerve tissue retrieved from patients undergoing reconstructive nerve procedures (Wilcox et al., 2020). Since the patients had a range of differing time intervals between injury and surgery, it was possible to construct an impression of the phenotypic changes of Schwann cells within a population over acute and chronic time points of denervation. The findings reveal novel information about the cellular and molecular features that underpin human nerve degeneration. The patterns of changes seen in the human nerve samples were similar to those previously reported in rodent models of neural degeneration. Schwann cells adopted a repair phenotype in acutely injured nerve samples which faded over time with chronic denervation. This finding may assist clinicians to optimize the timing of surgical nerve repair, to understand how pharmacological interventions might be used to improve clinical outcomes following surgery, or perhaps to ensure that surgery is not necessary.
    Related Articles | Metrics
    A standardized crush tool to produce consistent retinal ganglion cell damage in mice
    Pedro Norat, Jingyi Gao, Sauson Soldozy, Hao F. Zhang, Xiaorong Liu
    2021, 16 (7):  1442-1443.  doi: 10.4103/1673-5374.301015
    Abstract ( 88 )   PDF (416KB) ( 139 )   Save
    In the mammalian central nervous system, neuronal loss induced by injuries or in neurodegenerative diseases is often irreversible (Quigley, 2016; Gan et al., 2018). Following the disease insult, the surviving neurons may continue to lose their functionality because their axons degenerate and fail to maintain proper synaptic connections, and the underlying molecular and cellular mechanisms remain to be investigated (Raff et al., 2002; Bei et al., 2016; Quigley, 2016). Retinal ganglion cells (RGCs), for example, the neurons conveying visual information from the retina to the brain via the optic nerve, is a great model system to study the neural circuit and function because of its relatively easy accessibility and manipulation of the tissue. Optic nerve crush (ONC) injury in mice, the mechanical damage of the RGC axons at 0.5–1 mm behind the eye globe (Li et al., 1999), is a widely used model to examine the neural degeneration and axonal regeneration (Nickells et al., 2012; Quigley 2016). Since the introduction of this mouse model in late nineties, major progress has been made to characterize the axon degeneration and regeneration, especially assisted by the powerful mouse genetic tools that allow examining the molecular mechanisms underneath (Duan et al., 2015; Yi et al., 2016; Feng et al., 2017; Tran et al., 2019).
    Related Articles | Metrics
    Are mitochondria the key to reduce the age-dependent decline in axon growth after spinal cord injury?
    Theresa C. Sutherland, Cédric G. Geoffroy
    2021, 16 (7):  1444-1445.  doi: 10.4103/1673-5374.301016
    Abstract ( 88 )   PDF (1282KB) ( 121 )   Save
    Spinal cord injury (SCI) is a debilitating condition resulting in varying degrees of functional impairment and exhibits only limited repair. Currently there is no cure for SCI, and no proven treatment to promote restoration of function. One area that has received extensive attention, with the goal of promoting functional recovery, is promoting axonal regeneration and growth in the injured cord. However, one factor that is likely to impede the translation of promising restorative therapies from the bench to the clinic is the lack of consideration of the aging factor in SCI research and its impact on axon regeneration in particular. In the United States, the average age of occurrence of SCI is 43 years old (National Spinal Cord Injury Statistical Center), with a peak in incidence in young (20–30 years) and in aging (≥ 65 years) adults (GBD 2016 Traumatic Brain Injury and Spinal Cord Injury Collaborators, 2019). Patients are also living longer with the injury, with approximately 80% of all people with a SCI being over 40 (One Degree of Separation, Christopher & Dana Reeve Foundation). This demographic change has largely not been addressed in pre-clinical research. Indeed, the bulk of pre-clinical research is performed in young adult rodents (2–4 months), despite 6 month old mice more appropriately representing the 20–25 year old population in humans, and less than 0.35% of experimental rodents being 12 months or older, mimicking 40 years of age in humans (Fouad et al., 2020). In fact, an age-dependent decline in axon growth has been reported in a variety of model organisms, mediated by both neuron-intrinsic (Geoffroy et al., 2017) and extrinsic mechanisms (Sutherland and Geoffroy, 2020). While there has been significant progress made in understanding and manipulating axon growth after injury, even genetic manipulations promoting growth seem age-sensitive (Geoffroy et al., 2017), suggesting that other factors are needed to enhance axon growth in aging neurons. One of these factors is mitochondria. The mitochondrial theory of aging is one of the main mechanisms proposed to explain the biological process of aging. With age, mitochondrial function is reduced, which has been associated with a wide range of age-related diseases, including neurodegeneration (Haas, 2019). Importantly, mitochondria are essential for axonal growth and cell maintenance. Both normal aging and traumatic injury to the central nervous system (CNS) are highly associated with mitochondrial dysfunction and oxidative stress, this poses a great challenge for an aging SCI population as the two elements can compound one another to worsen injury outcomes. Observations from our laboratory has found detrimental changes in mitochondria in the aging CNS across a range of functional areas that will have a significant effect on neuronal health and ability to promote axonal growth in the event of injury. These observations suggest an important role for mitochondria in the age-dependent decline in axon growth potential that has been previously observed, and also may suggest targeting mitochondria as a promising therapeutic avenue for SCI regardless of age.
    Related Articles | Metrics
    The future of adenoassociated viral vectors for optogenetic peripheral nerve interfaces
    Hans E. Anderson, Richard F. ff. Weir
    2021, 16 (7):  1446-1447.  doi: 10.4103/1673-5374.301017
    Abstract ( 70 )   PDF (382KB) ( 105 )   Save
    Prostheses have a several thousand year history for treating limb loss. With time, these prostheses have become more sophisticated and closer to replicating the natural limb. These advances have culminated in the myoelectrically controlled prosthesis, which employs the surface electromyogram to decode the user’s intent. But the surface electromyogram lacks fidelity and though convenient, suffers from several problems. The skin-electrode interface undergoes impedance changes throughout the day and electrode liftoff can cause signal loss. A better solution would be to interface directly with the residual nerves which still carry the descending and ascending neural impulses. Signals can be recorded through electrodes implanted within peripheral nerves or the spinal cord, however current electrode technologies generally trade specificity for longevity and reliability. Electrodes that have high specificity use penetrating approaches that often irritate, damage and become encapsulated with fibrotic tissue limiting their long term viability (see (Navarro et al., 2005) for a review of surface electromyogram and electrode based interfaces). Optical approaches may obviate this problem and provide high specificity with limited invasiveness. Through the use of optogenetic actuator and reporter proteins, an optogenetic peripheral nerve interface can circumvent these problems by using light to manipulate and detect neural activity. This interface could be deployed for both prosthesis control following limb loss or limb reanimation following spinal cord injury (for example, see optogenetic actuation in a nonhuman primate model by (Williams et al., 2019)). An additional advantage of using optogenetic actuators in limb reanimation is that they may have a more physiologic motor unit recruitment pattern, recruiting slow oxidative fibers at lower stimulus than fast glycolytic fibers, reducing fatigue compared to electrical stimulation which recruits larger fibers at lower stimulus (Llewellyn et al., 2010). As transgenes, these optogenetic proteins require a method of delivery to the nerve, which can be achieved using viral vectors, which offer a high transduction efficiency. Of these vectors, adenoassociated viral vectors (AAVs) are particularly interesting because they are relatively nonimmunogenic and have been approved by the United States Food and Drug Administration to treat several genetic diseases. Furthermore, unlike lentiviruses and their wild-type progenitors, AAVs rarely integrate into the host genome, yet still provide extended expression. Targeting of these vectors to the motor and sensory neurons, and ultimately the axons of the dorsal root ganglia and ventral horn of the spinal cord is critical. Off-target expression can cause toxicity and immunogenicity. Selectivity is achieved through route of administration, expression cassette design, and serotype selection and capsid engineering (Figure 1A).
    Related Articles | Metrics
    Backdoor intrusion: retrotoxicity can explain targeted motor neuron death in amyotrophic lateral sclerosis 
    Randall D. McKinnon
    2021, 16 (7):  1448-1448.  doi: 10.4103/1673-5374.301037
    Abstract ( 68 )   PDF (203KB) ( 68 )   Save
    Amyotrophic lateral sclerosis (ALS) is a fatal disease of unknown cause that selectively targets brain and spinal cord motor neurons (MNs). The lifetime risk is 1 in 2000, and most cases are sporadic although up to 10% of patients are predisposed by familial mutations in MN protection or repair genes (Bruijn et al., 2004). Risk factors include agrochemical exposure and trauma (Walters et al., 2019), although why they target MN is perplexing. Farmers are at a greater risk than non-farming rural residents (Kang et al, 2014), and ALS clusters occur in abrasion prone activities conducted on agrochemical treated fields such as baseball and soccer (Chio et al., 2005). These observations suggest that one mechanism for targeted loss of MNs may be the retrograde transport of neurotoxins subsequent to peripheral nerve injury, a process termed ‘retrotoxicity’. 
    Related Articles | Metrics