Loading...

Table of Content

    15 January 2022, Volume 17 Issue 1 Previous Issue   
    For Selected: Toggle Thumbnails
    Genes for RNA-binding proteins involved in neural-specific functions and diseases are downregulated in Rubinstein-Taybi iNeurons
    Lidia Larizza, Luciano Calzari, Valentina Alari, Silvia Russo
    2022, 17 (1):  5-14.  doi: 10.4103/1673-5374.314286
    Abstract ( 201 )   PDF (2436KB) ( 208 )   Save
    Taking advantage of the fast-growing knowledge of RNA-binding proteins (RBPs) we review  the  signature of downregulated genes for RBPs in the transcriptome of induced pluripotent stem cell neurons (iNeurons) modelling the neurodevelopmental Rubinstein Taybi Syndrome (RSTS) caused by mutations in the genes encoding CBP/p300 acetyltransferases. We discuss top and functionally connected downregulated genes sorted to “RNA processing” and “Ribonucleoprotein complex biogenesis” Gene Ontology clusters. The first set of downregulated RBPs includes members of hnRNHP (A1, A2B1, D, G, H2-H1, MAGOHB, PAPBC), core subunits of U small nuclear ribonucleoproteins and Serine-Arginine splicing regulators families, acting in precursor messenger RNA alternative splicing and processing. Consistent with literature findings on reduced transcript levels of serine/arginine repetitive matrix 4 (SRRM4) protein, the main regulator of the neural-specific microexons splicing program upon depletion of Ep300 and Crebbp in mouse neurons, RSTS iNeurons show downregulated genes for proteins  impacting this network. We link downregulated genes to neurological disorders including the new HNRNPH1-related intellectual disability syndrome with clinical overlap to RSTS. The set of downregulated genes for Ribosome biogenesis includes several components of ribosomal subunits and nucleolar proteins, such NOP58 and fibrillarin that form complexes with snoRNAs with a central role in guiding post-transcriptional modifications needed for rRNA maturation. These nucleolar proteins are “dual” players as fibrillarin is also required for epigenetic regulation of ribosomal genes and conversely NOP58-associated snoRNA levels are under the control of NOP58 interactor BMAL1, a transcriptional regulator of the circadian rhythm. Additional downregulated genes for “dual specificity” RBPs such as RUVBL1 and METTL1 highlight the links between chromatin and the RBP-ome and the contribution of perturbations in their cross-talk to RSTS. We underline the hub position of CBP/p300 in chromatin regulation, the impact of its defect on neurons’ post-transcriptional regulation of gene expression and the potential use of epidrugs in therapeutics of RBP-caused neurodevelopmental disorders.
    Related Articles | Metrics
    Research advances on how metformin improves memory impairment in “chemobrain”
    Ahmad Alhowail, Sridevi Chigurupati
    2022, 17 (1):  15-19.  doi: 10.4103/1673-5374.314284
    Abstract ( 205 )   PDF (355KB) ( 143 )   Save
    Cognitive impairment caused by chemotherapy, referred to as “chemobrain,” is observed in approximately 70% of cancer survivors. However, it is not completely understood how chemotherapy induces cognitive dysfunction, and clinical treatment strategies for this problem are lacking. Metformin, used as a first-line treatment for type 2 diabetes mellitus, is reported to reduce the effects of chemobrain. Recently, several studies have examined the effect of metformin in rescuing chemobrain. This review discusses recent clinical/preclinical studies that addressed some mechanisms of chemobrain and evaluates the effect of metformin in rescuing chemobrain and its potential mechanisms of action.
    Related Articles | Metrics
    Dendritic spine density changes and homeostatic synaptic scaling: a meta-analysis of animal studies
    Thiago C. Moulin, Danielle Rayêe, Helgi B. Schiöth
    2022, 17 (1):  20-24. 
    Abstract ( 239 )   PDF (744KB) ( 121 )   Save
    Mechanisms of homeostatic plasticity promote compensatory changes of cellular excitability in response to chronic changes in the network activity. This type of plasticity is essential for the maintenance of brain circuits and is involved in the regulation of neural regeneration and the progress of neurodegenerative disorders. One of the most studied homeostatic processes is synaptic scaling, where global synaptic adjustments take place to restore the neuronal firing rate to a physiological range by the modulation of synaptic receptors, neurotransmitters, and morphology. However, despite the comprehensive literature on the electrophysiological properties of homeostatic scaling, less is known about the structural adjustments that occur in the synapses and dendritic tree. In this study, we performed a meta-analysis of articles investigating the effects of chronic network excitation (synaptic downscaling) or inhibition (synaptic upscaling) on the dendritic spine density of neurons. Our results indicate that spine density is consistently reduced after protocols that induce synaptic scaling, independent of the intervention type. Then, we discuss the implication of our findings to the current knowledge on the morphological changes induced by homeostatic plasticity.
    Related Articles | Metrics
    Optogenetic activation of intracellular signaling based on light-inducible protein-protein homo-interactions
    Peiyuan Huang, Zhihao Zhao, Liting Duan
    2022, 17 (1):  25-30.  doi: 10.4103/1673-5374.314293
    Abstract ( 200 )   PDF (552KB) ( 243 )   Save
    Dynamic protein-protein interactions are essential for proper cell functioning. Homo-interaction events—physical interactions between the same type of proteins—represent a pivotal subset of protein-protein interactions that are widely exploited in activating intracellular signaling pathways. Capacities of modulating protein-protein interactions with spatial and temporal resolution are greatly desired to decipher the dynamic nature of signal transduction mechanisms. The emerging optogenetic technology, based on genetically encoded light-sensitive proteins, provides promising opportunities to dissect the highly complex signaling networks with unmatched specificity and spatiotemporal precision. Here we review recent achievements in the development of optogenetic tools enabling light-inducible protein-protein homo-interactions and their applications in optical activation of signaling pathways.
    Related Articles | Metrics
    Presenilin mutations and their impact on neuronal differentiation in Alzheimer’s disease
    Mercedes A. Hernández-Sapiéns, Edwin E. Reza-Zaldívar, Ana L. Márquez-Aguirre, Ulises Gómez-Pinedo, Jorge Matias-Guiu, Ricardo R. Cevallos, Juan C. Mateos-Díaz, Víctor J. Sánchez-González, Alejandro A. Canales-Aguirre
    2022, 17 (1):  31-37.  doi: 10.4103/1673-5374.313016
    Abstract ( 275 )   PDF (945KB) ( 144 )   Save
    The presenilin genes (PSEN1 and PSEN2) are mainly responsible for causing early-onset familial Alzheimer’s disease, harboring ~300 causative mutations, and representing ~90% of all mutations associated with a very aggressive disease form. Presenilin 1 is the catalytic core of the γ-secretase complex that conducts the intramembranous proteolytic excision of multiple transmembrane proteins like the amyloid precursor protein, Notch-1, N- and E-cadherin, LRP, Syndecan, Delta, Jagged, CD44, ErbB4, and Nectin1a. Presenilin 1 plays an essential role in neural progenitor maintenance, neurogenesis, neurite outgrowth, synaptic function, neuronal function, myelination, and plasticity. Therefore, an imbalance caused by mutations in presenilin 1/γ-secretase might cause aberrant signaling, synaptic dysfunction, memory impairment, and increased Aβ42/Aβ40 ratio, contributing to neurodegeneration during the initial stages of Alzheimer’s disease pathogenesis. This review focuses on the neuronal differentiation dysregulation mediated by PSEN1 mutations in Alzheimer’s disease. Furthermore, we emphasize the importance of Alzheimer’s disease-induced pluripotent stem cells models in analyzing PSEN1 mutations implication over the early stages of the Alzheimer’s disease pathogenesis throughout neuronal differentiation impairment.
    Related Articles | Metrics
    Growth differentiation factor 5: a neurotrophic factor with neuroprotective potential in Parkinson’s disease
    Susan R. Goulding, Jayanth Anantha, Louise M. Collins, Aideen M. Sullivan, Gerard W. O’Keeffe
    2022, 17 (1):  38-44.  doi: 10.4103/1673-5374.314290
    Abstract ( 189 )   PDF (645KB) ( 102 )   Save
    Parkinson’s disease is the most common movement disorder worldwide, affecting over 6 million people. It is an age-related disease, occurring in 1% of people over the age of 60, and 3% of the population over 80 years. The disease is characterized by the progressive loss of midbrain dopaminergic neurons from the substantia nigra, and their axons, which innervate the striatum, resulting in the characteristic motor and non-motor symptoms of Parkinson’s disease. This is paralleled by the intracellular accumulation of α-synuclein in several regions of the nervous system. Current therapies are solely symptomatic and do not stop or slow disease progression. One promising disease-modifying strategy to arrest the loss of dopaminergic neurons is the targeted delivery of neurotrophic factors to the substantia nigra or striatum, to protect the remaining dopaminergic neurons of the nigrostriatal pathway. However, clinical trials of two well-established neurotrophic factors, glial cell line-derived neurotrophic factor and neurturin, have failed to meet their primary end-points. This failure is thought to be at least partly due to the downregulation by α-synuclein of Ret, the common co-receptor of glial cell line-derived neurorophic factor and neurturin. Growth/differentiation factor 5 is a member of the bone morphogenetic protein family of neurotrophic factors, that signals through the Ret-independent canonical Smad signaling pathway. Here, we review the evidence for the neurotrophic potential of growth/differentiation factor 5 in in vitro and in vivo models of Parkinson’s disease. We discuss new work on growth/differentiation factor 5’s mechanisms of action, as well as data showing that viral delivery of growth/differentiation factor 5 to the substantia nigra is neuroprotective in the α-synuclein rat model of Parkinson’s disease. These data highlight the potential for growth/differentiation factor 5 as a disease-modifying therapy for Parkinson’s disease. 
    Related Articles | Metrics
    The promise of neuroprotection by dietary restriction in glaucoma 
    Rossella Russo, Carlo Nucci, Annagrazia Adornetto
    2022, 17 (1):  45-47.  doi: 10.4103/1673-5374.314308
    Abstract ( 154 )   PDF (1645KB) ( 257 )   Save
    Glaucoma, a progressive age-related optic neuropathy characterized by the death of retinal ganglion cells, is the most common neurodegenerative cause of irreversible blindness worldwide. The therapeutic management of glaucoma, which is limited to lowering intraocular pressure, is still a challenge since visual loss progresses in a significant percentage of treated patients. Restricted dietary regimens have received considerable attention as adjuvant strategy for attenuating or delaying the progression of neurodegenerative diseases. Here we discuss the literature exploring the effects of modified eating patterns on retinal aging and resistance to stressor stimuli.
    Related Articles | Metrics
    New perspectives for mesenchymal stromal cells as an adjuvant therapy for infectious disease-associated encephalopathies
    Maiara N. Lima, Maria C. Barbosa-Silva, Tatiana Maron-Gutierrez
    2022, 17 (1):  48-52.  doi: 10.4103/1673-5374.314292
    Abstract ( 179 )   PDF (910KB) ( 104 )   Save
    Knowledge of the mechanisms that trigger infection-related encephalopathies is still very limited and cell therapies are one of the most promising alternatives for neurodegenerative diseases, and its application in infectious diseases can be of great relevance. Mesenchymal stromal cells are cells with great immunomodulatory potential; therefore, their use in clinical and preclinical studies in a variety of diseases, including central nervous system diseases, increased in the last decade. Mesenchymal stromal cells can exert their beneficial effects through several mechanisms, such as direct cell contact, through surface receptors, and also through paracrine or endocrine mechanisms. The paracrine mechanism is widely accepted by the scientific community and involves the release of soluble factors, which include cytokines, chemokines and trophic factors, and extracellular vesicles. This mini review discusses mesenchymal stromal cells mechanisms of action in neurological disorders, the neuroinflammatory process that takes place in the brain as a result of peripheral inflammation and changes in the brain’s cellular scenario as a common factor in central nervous system diseases, and mesenchymal stromal cells therapy in encephalopathies. Mesenchymal stromal cells have been shown to act in neuroinflammation processes, leading to improved survival and mitigating behavioral damage. More recently, these cells have been tested in preclinical models of infectious diseases-associated encephalopathies (e.g., cerebral malaria and sepsis associated encephalopathy) and have shown satisfactory results.
    Related Articles | Metrics
    The gut microbiome: implications for neurogenesis and neurological diseases
    Cheng Liu, Shang-Yu Yang, Long Wang, Fang Zhou
    2022, 17 (1):  53-58. 
    Abstract ( 211 )   PDF (984KB) ( 234 )   Save
    There is an increasing recognition of the strong links between the gut microbiome and the brain, and there is persuasive evidence that the gut microbiome plays a role in a variety of physiological processes in the central nervous system. This review summarizes findings that gut microbial composition alterations are linked to hippocampal neurogenesis, as well as the possible mechanisms of action; the existing literature suggests that microbiota influence neurogenic processes, which can result in neurological disorders. We consider this evidence from the perspectives of neuroinflammation, microbial-derived metabolites, neurotrophins, and neurotransmitters. Based on the existing research, we propose that the administration of probiotics can normalize the gut microbiome. This could therefore also represent a promising treatment strategy to counteract neurological impairment.
    Related Articles | Metrics
    Central nervous system stimulation therapies in phantom limb pain: a systematic review of clinical trials
    M. Ángeles García-Pallero, Diana Cardona, Lola Rueda-Ruzafa, Miguel Rodriguez-Arrastia, Pablo Roman
    2022, 17 (1):  59-64.  doi: 10.4103/1673-5374.314288
    Abstract ( 217 )   PDF (409KB) ( 172 )   Save

    Phantom limb pain is a chronic pain syndrome that is difficult to cope with. Despite neurostimulation treatment is indicated for refractory neuropathic pain, there is scant evidence from randomized controlled trials to recommend it as the treatment choice. Thus, a systematic review was performed to analyze the efficacy of central nervous system stimulation therapies as a strategy for pain management in patients with phantom limb pain. A literature search for studies conducted between 1970 and September 2020 was carried out using the MEDLINE and Embase databases. Principles of The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guideline were followed. There were a total of 10 full-text articles retrieved and included in this review. Deep brain stimulation, repetitive transcranial magnetic stimulation, transcranial direct current stimulation, and motor cortex stimulation were the treatment strategies used in the selected clinical trials. Repetitive transcranial magnetic stimulation and transcranial direct current stimulation were effective therapies to reduce pain perception, as well as to relieve anxiety and depression symptoms in phantom limb pain patients. Conversely, invasive approaches were considered the last treatment option as evidence in deep brain stimulation and motor cortex stimulation suggests that the value of phantom limb pain treatment remains controversial. However, the findings on use of these treatment strategies in other forms of neuropathic pain suggest that these invasive approaches could be a potential option for phantom limb pain patients.

    Related Articles | Metrics
    Lower and upper motor neuron involvement and their impact on disease prognosis in amyotrophic lateral sclerosis
    Maria N. Zakharova, Anna A. Abramova
    2022, 17 (1):  65-73.  doi: 10.4103/1673-5374.314289
    Abstract ( 335 )   PDF (410KB) ( 166 )   Save
    Amyotrophic lateral sclerosis is a fatal neurodegenerative disease characterized by progressive muscle wasting, breathing and swallowing difficulties resulting in patient’s death in two to five years after disease onset. In amyotrophic lateral sclerosis, both upper and lower motor neurons of the corticospinal tracts are involved in the process of neurodegeneration, accounting for great clinical heterogeneity of the disease. Clinical phenotype has great impact on the pattern and rate of amyotrophic lateral sclerosis progression and overall survival prognosis. Creating more homogenous patient groups in order to study the effects of drug agents on specific manifestations of the disease is a challenging issue in amyotrophic lateral sclerosis clinical trials. Since amyotrophic lateral sclerosis has low incidence rates, conduction of multicenter trials requires certain standardized approaches to disease diagnosis and staging. This review focuses on the current approaches in amyotrophic lateral sclerosis classification and staging system based on clinical examination and additional instrumental methods, highlighting the role of upper and lower motor neuron involvement in different phenotypes of the disease. We demonstrate that both clinical and instrumental findings can be useful in evaluating severity of upper motor neuron and lower motor neuron involvement and predicting the following course of the disease. Addressing disease heterogeneity in amyotrophic lateral sclerosis clinical trials could lead to study designs that will assess drug efficacy in specific patient groups, based on the disease pathophysiology and spatiotemporal pattern. Although clinical evaluation can be a sufficient screening method for dividing amyotrophic lateral sclerosis patients into clinical subgroups, we provide proof that instrumental studies could provide valuable insights in the disease pathology.
    Related Articles | Metrics
    Positron emission tomography imaging for the assessment of mild traumatic brain injury and chronic traumatic encephalopathy: recent advances in radiotracers
    Chu-Xin Huang, Yan-Hui Li, Wei Lu, Si-Hong Huang, Meng-Jun Li, Li-Zhi Xiao, Jun Liu
    2022, 17 (1):  74-81.  doi: 10.4103/1673-5374.314285
    Abstract ( 149 )   PDF (886KB) ( 162 )   Save
    A chronic phase following repetitive mild traumatic brain injury can present as chronic traumatic encephalopathy in some cases, which requires a neuropathological examination to make a definitive diagnosis. Positron emission tomography (PET) is a molecular imaging modality that has high sensitivity for detecting even very small molecular changes, and can be used to quantitatively measure a range of molecular biological processes in the brain using different radioactive tracers. Functional changes have also been reported in patients with different forms of traumatic brain injury, especially mild traumatic brain injury and subsequent chronic traumatic encephalopathy. Thus, PET provides a novel approach for the further evaluation of mild traumatic brain injury at molecular levels. In this review, we discuss the recent advances in PET imaging with different radiotracers, including radioligands for PET imaging of glucose metabolism, tau, amyloid-beta, γ-aminobutyric acid type A receptors, and neuroinflammation, in the identification of altered neurological function. These novel radiolabeled ligands are likely to have widespread clinical application, and may be helpful for the treatment of mild traumatic brain injury. Moreover, PET functional imaging with different ligands can be used in the future to perform large-scale and sequential studies exploring the time-dependent changes that occur in mild traumatic brain injury.

    Related Articles | Metrics
    Deciphering the transcriptomic signature of synaptic activity
    Guido Hermey
    2022, 17 (1):  82-84.  doi: 10.4103/1673-5374.315229
    Abstract ( 217 )   PDF (544KB) ( 134 )   Save
    Neurons undergo activity-dependent changes in their molecular composition and structure in order to regulate cellular processes such as dendritic growth, synapse elimination, spine maturation and synaptic strength. Such synaptic plasticity plays an important role during a critical period in brain development and contributes to sensory adaptation and to learning and memory in the mature nervous system. Its dysregulation underlies a number of pathological processes in psychiatric and neurodegenerative disorders, such as addiction, depression, anxiety, schizophrenia, epilepsy and traumatic brain injury. Short-term activity-dependent synaptic changes rely mostly on post-translational modifications of pre-existing proteins, whereas the long-term maintenance of synaptic adaptations depends on gene induction. Signals from the synapse to the nucleus activate gene expression. Such signals are thought to be encoded in calcium waves or conveyed by macromolecular signaling complexes translocated retrogradely by motor proteins. The induced gene transcription and protein synthesis alters the composition of synaptic protein networks and provides a mechanism for translating synaptic activity into persistent synaptic changes. In accordance, large sets of genes whose expression levels are regulated by synaptic activity have been described in the past decades. As expected, several of these genes encode proteins that modulate synaptic function and play a role in neuronal plasticity-related processes. However, it has to be considered that altered gene expression levels are only part of the complex activity-regulated transcriptional signature. Alternative splicing, the differential inclusion and exclusion of exonic sequence, in combination with other related processes such as the use of alternative transcriptional initiation sites and alternative polyadenylation sites as well as mRNA editing ensure the transcriptomic and proteomic diversity required for the regulation and diversification of synaptic functions.
    Related Articles | Metrics
    Barriers to axonal regeneration after spinal cord injury: a current perspective
    Jacek M. Kwiecien
    2022, 17 (1):  85-86.  doi: 10.4103/1673-5374.314299
    Abstract ( 175 )   PDF (305KB) ( 100 )   Save
    Regeneration of long axons after the spinal cord injury (SCI) will benefit patients with extensive traumatic damage to the white matter pathways who experience intolerable, permanent, neurologic deficits even after neuroprotective treatment with anti-inflammatory agents (Kwiecien, 2021a). This short paper attempts to synthetize pathologic mechanisms or barriers involved in inhibition of axonal regeneration in the SCI and provides suggestions of therapeutic interventions enabling this regeneration in animal models.
    Related Articles | Metrics
    TC10 as an essential molecule in axon regeneration through membrane supply and microtubule stabilization
    Takeshi Nakamura, Shingo Koinuma
    2022, 17 (1):  87-88.  doi: 10.4103/1673-5374.314297
    Abstract ( 190 )   PDF (881KB) ( 122 )   Save
    Mammalian central nervous system (CNS) neurons lose axon regenerative ability as they mature. This failure to regenerate shows a clear contrast to a remarkable potential of axon growth during embryonic development and after an injury in the peripheral nervous system (PNS) (Hilton and Bradke, 2017). The absence of regeneration in the mature CNS neurons is caused by an inhibitory influence of the environment of the injured axons and the deficit of intrinsic factors that enable regeneration in the PNS (He and Jin, 2016). In the last two decades, gene manipulation strategies and compound screening have identified several neuron-intrinsic players involved in axon regeneration (Ribas and Costa, 2017). The central players are the PTEN/mTOR pathway, which contributes to protein synthesis, and transcription factors (such as SOCS3, KLF family, and SOX11) which control the cell differentiation/de-differentiation status. In addition, cytoskeletal dynamics at growth cones and material transport in axons are essential for axon regrowth; however, it is unclear how the components that regulate these functions are modulated in injured CNS neurons. In this study, we have discussed our recent discovery of an indispensable role of TC10 in CNS and PNS axon regeneration (Koinuma et al., 2020).
    Related Articles | Metrics
    Ferroptosis: copper-iron connection in cuprizone-induced demyelination
    Priya Jhelum, Samuel David
    2022, 17 (1):  89-90.  doi: 10.4103/1673-5374.314300
    Abstract ( 332 )   PDF (516KB) ( 136 )   Save
    Redox active metals such as iron, copper, zinc, and manganese play important roles in promoting a variety of biochemical reactions essential for cellular function. This is made possible by the ability of these metals to accept and donate electrons. Iron in the form of iron-sulfur clusters and heme plays a key role in adenosine triphosphate generation in mitochondria as well as numerous other enzymatic reactions. On the other hand, disruption in the normal homeostatic levels of these metals, either excess or reduction, results in damage to cells and to tissue pathology. Excess copper accumulation in the central nervous system (CNS) in Wilson’s disease results in damage to the basal ganglia, and excess iron deposition in the CNS in aceruloplasminemia results in damage to various regions of the brain and retina. Such damage is thought to be induced by free radicals generated via Fenton chemistry. Here, we focus on our recent work that revealed a role for ferroptosis, a form of iron-mediated cell death, in cuprizone-induced oligodendrocyte loss and demyelination (Jhelum et al., 2020). Cuprizone is a copper chelator that induces demyelination in experimental animals and is widely used to study demyelination and remyelination in the CNS (Zhan et al., 2020), often in the context of multiple sclerosis (MS), the prototypical demyelination disease in humans. Another key finding in this work is the potential role of ferritinophagy in ferroptosis, i.e., the release of iron from cytosolic ferritin (Dixon et al., 2012; Mancias et al., 2014). This work also shows the link between copper and iron metabolism, in which chelating copper leads to dysregulation of iron metabolism. Importantly, ferroptosis may also play a role in other neurological conditions and deserves further study.
    Related Articles | Metrics
    Nogo-A receptor internalization by cyclic adenosine monophosphate in overcoming axonal growth inhibitors after stroke
    Rayudu Gopalakrishna, Charlotte Lin, Mark S. Kindy, William J. Mack
    2022, 17 (1):  91-92.  doi: 10.4103/1673-5374.314298
    Abstract ( 175 )   PDF (544KB) ( 129 )   Save
    Currently, there are no clinically proven drugs for recovery from stroke and other neuronal injuries such as traumatic brain injury and spinal cord injury. Recovery therapy requires axonal regeneration, which is inhibited by diverse axonal growth inhibitors, such as Nogo-A, myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (OMgp), and chondroitin sulfate proteoglycans (CSPGs) (Chaudhry and Filbin, 2007; Carmichael, 2010; Schwab and Strittmatter, 2014). A cell-surface receptor for Nogo-A, NgR1, mediates the inhibitory action of not only Nogo-A but also other axonal inhibitors (MAG, OMgp, and CSPGs) as well. Intracellular cyclic adenosine monophosphate (cAMP) overcomes neuronal growth inhibition caused by myelin and improves functional recovery from neuronal injuries (Chaudhry and Filbin, 2007). It may block the actions of axonal growth inhibitors by inducing a transcriptional activation of specific genes.  In some cases, cAMP rapidly prevents axonal growth inhibitors from acting through an unknown mechanism (Murray and Shewan, 2008).  
    Related Articles | Metrics
    Estrogens still represent an attractive therapeutic approach for Alzheimer’s disease
    Elena Tamagno, Michela Guglielmotto
    2022, 17 (1):  93-94.  doi: 10.4103/1673-5374.314295
    Abstract ( 250 )   PDF (290KB) ( 132 )   Save
    Alzheimer’s disease (AD) is a progressive neurodegenerative condition that goes from mild cognitive impairment in prodromal disease to severely disabling deficits in advanced stages. The risk for AD development, as well as progression and severity, clearly differ between men and women (Pike, 2017). Epidemiological studies have shown that there is a significantly increased prevalence in the development of AD in women compared to men, which is usually explained by the longer lifespan of women. This increased frequency may be due to the interplay between age and sex, in which genetic factors together with hormonal and metabolic patterns play a crucial role. Moreover, cognitive impairment has been confirmed to be greater in women than in men at the same stage of AD, likely due to reduced estrogen levels in post-menopausal women (Laws et al., 2016).
    Related Articles | Metrics
    The new wave of p75 neurotrophin receptor targeted therapies 
    Amanda M. Crooks, Rick B. Meeker
    2022, 17 (1):  95-96.  doi: 10.4103/1673-5374.314304
    Abstract ( 179 )   PDF (291KB) ( 166 )   Save
    Neurotrophins have been recognized for decades for their beneficial effects on growth, survival, and maintenance in the central nervous system, all of which suggest potential therapeutic utility. Although understanding and harnessing the activity of neurotrophins has proven difficult, the past several years have seen significant strides in the development of deliverable therapies that modulate neurotrophin activity (Shen et al., 2019; Yang et al., 2020; Xie et al., 2021). These recent studies have primarily focused on the multifunctional p75 neurotrophin receptor (p75NTR) which is upregulated in central nervous system disease and injury, thus offering a unique target for intervention. Animal studies focusing on neurodegenerative diseases, infection and injury have all illustrated the potential benefit of p75NTR modulation, such as prevention of neural damage via restoration of calcium homeostasis, facilitation of pro-survival signaling, and reduction of inflammation. In addition, new studies have revealed important interactions of p75NTR with microtubule-associated protein, Tau, relevant to Alzheimer’s disease (AD) pathogenesis, tauopathies, injury, and infection. Importantly, these investigational therapies have also been shown to be effectively deliverable as well as tolerable, with few side effects in animal models. Although the actions of p75NTR are complex and studies have only begun to reveal their potential utility, these new developments have paved the way for clinical trials of focused, modulatory interventions that have the potential to be broadly applicable in the treatment and prevention of central nervous system disease. 
    Related Articles | Metrics
    Fibrosis as a common trait in amyotrophic lateral sclerosis tissues
    Savina Apolloni, Nadia D’Ambrosi
    2022, 17 (1):  97-98.  doi: 10.4103/1673-5374.314302
    Abstract ( 298 )   PDF (951KB) ( 118 )   Save
    Amyotrophic lateral sclerosis (ALS) is a highly aggressive adult-onset neurodegenerative disease caused by the progressive loss of upper and lower motor neurons. Clinically, it causes irreversible muscle atrophy and spasticity, leading to death due to respiratory failure, usually within 2–5 years after the first symptom onset. Approximately 85% of ALS cases are classified as sporadic, while the remaining 15% are of familial origin, but the overall clinical and molecular features of the disease are almost undistinguishable in the two forms. The majority of familial ALS cases are caused by pathogenic variants of C9orf72, SOD1, TARDBP, FUS, ANG and OPTN genes that are inherited by a Mendelian pattern and display high penetrance (Kiernan et al., 2020).
    Related Articles | Metrics
    Small scale adeno-associated virus-vector production for preclinical gene delivery based on chloroform precipitation
    Marcus Davidsson, Andreas Heuer
    2022, 17 (1):  99-100.  doi: 10.4103/1673-5374.314309
    Abstract ( 168 )   PDF (348KB) ( 139 )   Save
    Gene therapy aims to introduce genetic information into a cell-type of interest to replace, correct, silence, or modify defective genes. Gene therapy in its broadest sense can theoretically prevent, halt, or cure any condition that affects mankind. In addition to that, the introduction and/or manipulation of genes is one of the major research areas in biological sciences, aimed to deepen our knowledge on how biological systems work. Scientific advances have made it possible to induce changes ranging from manipulations of large stretches of the genome to the change of single nucleotides. The gold-standard vehicles to bring this genetic information into the target cells are viral vectors, amongst which the adeno-associated virus (AAV) is the most commonly used. AAV-vectors are small single stranded DNA viruses that naturally infect cells in humans and other primate species, thereby making them a perfect candidate for gene-therapy. In contrast to retroviruses, such as lentiviral vectors, AAVs are replication deficient and do not integrate into the host genome thus reducing the risk of insertional mutagenesis. Furthermore, AAVs are currently not known to cause any diseases in humans. The AAVs have an excellent biosafety profile and approximately 80–90% of the human population is already carrying the virus. There are currently about 13 naturally occurring variants known (serotypes), each with a different tropism profile to specific cell/tissue types (Vance et al., 2015). 
    Related Articles | Metrics
    Investigating the role of imprinted genes in pediatric sporadic brain arteriovenous malformations
    Concetta Scimone, Luigi Donato, Antonina Sidoti
    2022, 17 (1):  101-102.  doi: 10.4103/1673-5374.314296
    Abstract ( 141 )   PDF (905KB) ( 84 )   Save
    Arteriovenous malformation (AVM) is a vascular congenital defect affecting microvasculature of both brain and peripheral organs. Arteriovenous malformation of the brain (bAVM, OMIM #108010), in particular, affects up to 15 per 100,000 persons with no sex predominance. Almost 50% of the patients manifest intracerebral hemorrhage and epileptic seizures, as main clinical symptoms. Anatomically, lesions exhibit the direct shunt from arterioles to venules, lacking the normal capillary bed. Arterioles and venules are curled forming a tangle called nidus. At the nidus, pericytes are reduced. Feeding arteries and draining veins show impaired expression of vessel differentiation markers. These features result in loss of endothelial cells properties and increased permeability of the affected vessels. The high pressure of blood perfusing from arteries to the nidus increases risk of lesion rupture, resulting in intracerebral hemorrhage. Moreover, at the nidus, arterial and venous blood mixes, altering the normal oxygenation of the central nervous system (Barbosa Do Prado et al., 2019). 
    Related Articles | Metrics
    Current and future therapeutic strategies for the treatment of retinal neurodegenerative diseases
    Victoria Maneu, Pedro Lax, Nicolás Cuenca
    2022, 17 (1):  103-104.  doi: 10.4103/1673-5374.314305
    Abstract ( 314 )   PDF (5876KB) ( 203 )   Save
    The complex and mostly multiple and unknown aetiology of neurodegenerative diseases always give way to an intricate scenario of dying tissue that involves multiple cell mediators and cell types. All neurodegenerative diseases of the central nervous system (CNS) share common mechanisms, regardless their origin: oxidative stress, neuroinflammation and cell death. Accordingly, retinal degenerative diseases, with or without a genetic cause, as retinitis pigmentosa (RP), glaucoma, age-related macular degeneration (AMD) or diabetic retinopathy (DR) do not differ in their basic mechanisms of cell death neither one to another, nor from those observed in other CNS diseases as Parkinson’s or Alzheimer’s (Cuenca et al., 2014). Indeed, the therapeutic findings should be able to be more or less easily extrapolated between these conditions, as far as they are directed to common dartboards. Gene therapy, in which we have very high hopes to solve genetic disorders, is currently being traslated from preclinical assays to the clinic for some retinal degenerative diseases, with successful achievement up today for the Leber congenital amaurosis, due to mutations in the RPE65 gene (Garafalo et al., 2020). But our promising therapies still face relevant challenges. In this sense, CRISP/Cas editing tools used to amend genetic missenses, need to fix secondary effects, as those related to the immune response (Yu et al., 2017); stem cell approaches have to procure the functionality of transplanted cells in the recipient, to assure the accurate establishment of synaptic connectivity and cell contacts, and gain success in precise image processing (Cuevas et al., 2019; Garita-Hernandez et al., 2019); and optogenetics also needs to find appropriate vectors for the delivery and expression in suitable cell types, avoiding immunological rejection of the vector systems (Shen et al., 2020). While gene- and cell-based therapies evolve through the tortuous pathway of biological success, combined therapies with antioxidant (as lutein or zeaxanthin), antiinflammatory (as corticosteroids or cannabinoids), and antiapoptotic (as tauroursodeoxycholic acid or proinsulin) molecules appear currently as the widest approach to pharmacologically treat a wide spectrum of retinal degenerative diseases. These compounds provide several advantages. They can slow down the progression of the degenerative process, so preserving the visual capacity for a certain time. Moreover, the administration of neuroprotective factors is essential even when the vision has been completely lost, as they can improve non-visual functions, like the control of circadian rhythms and pupil contraction, as the cannabinoid-mediated improvement of circadian rhythmicity in P23H rats, which are mediated by the melanopsin-containing photosensitive ganglion cells (Lax et al., 2019). Non-visual retinal functions have also effects on memory and depression. Therefore, the preservation of this subset of cells, although will not improve the visual function, will surely improve the quality of life of the patients and should not be underestimated. But, far beyond, these molecules will surely increase the success of the new therapies, as they can provide an adequate environment of healthy cells, as a substrate for gene transplant or optogenetic approaches, which could hardly be successful in a damaged tissue surrounded by dying cells. Genetic material can be potentially incorporated to the retina and eventually restore the visual functionality in the zone in which it is injected but, without a global actuation on the whole retina by maintaining the health of the adjacent cells, an inflamed surrounding could end in a complete failure of any therapy. Hence, the concomitant use of antiinflammatory, antioxidant and antiapoptotic agents, as well as neurotrophic and growth factors, will provide an adequate environment of healthy cells that will help to achieve a sustained functional restoration of the visual function (Figure 1), as it has been shown for the combination of progesterone and lipoic acid in a mouse model of RP (Ramirez-Lamelas et al., 2018).
    Related Articles | Metrics
    Hypoxia inducible factor and diffuse white matter injury in the premature brain: perspectives from genetic studies in mice
    Fuzheng Guo, Sheng Zhang
    2022, 17 (1):  105-107.  doi: 10.4103/1673-5374.314301
    Abstract ( 191 )   PDF (483KB) ( 100 )   Save
    Hypoxia-inducible factors (HIFs) are transcriptional regulators playing important roles in adapting various types of cells to physiological and pathological hypoxia cues. Three structurally related, oxygen-sensitive HIFα proteins have been identified (HIF1α, HIF2α, and HIF3α), among which HIF3α has weak transcriptional capacity because of the absence of the C-terminal transactivation domain as present in HIF1α and HIF2α. The role of HIFα in regulating diverse biological processes is primarily through the actions of its downstream target genes and/or signaling pathways (Figure 1). The HIFα signaling is subjected to regulation at the multiple levels as detailed in Figure 1. Previous studies have shown that HIF1α and HIF2α activate both common (canonical) and distinct (non-canonical) sets of target genes in cell-type and context dependent manners. The importance of HIFα (HIF1α and HIF2α) in embryonic development is manifested by the lethality of early embryos or neonates of HIF1α–/– mice and HIF2α–/– mice due to the cardiovascular and lung malformation. In the developing central nervous system (CNS) where the maturation of the vascular network is still ongoing, the local oxygen concentration ranges from 0.5% to 7% (Ivanovic, 2009). The physiologically hypoxic environment in the CNS suggests that HIFα may play an important role in neural development. 
    Related Articles | Metrics
    Inhibition of CXCR4/CXCL12 signaling: a translational perspective for Alzheimer’s disease treatment
    Yuval Gavriel, Inna Rabinovich-Nikitin, Beka Solomon
    2022, 17 (1):  108-109.  doi: 10.4103/1673-5374.314303
    Abstract ( 240 )   PDF (240KB) ( 148 )   Save
    The mechanism of AD remains uncovered: The current mainstream doctrine in Alzheimer’s disease (AD) is the amyloid cascade hypothesis. According to this hypothesis, amyloid-β (Aβ) deposition is the main reason for neurofibrillary tangles formation and synaptic dysfunction. However, treatments with antibodies against such targets did not manage to improve cognitive decline and leave some questions regarding the theory of amyloid plaques. Extensive evidence indicates that several pathology changes occur before the appearance of Aβ and tau aggregates. One of the changes is the assault in oligodendrocytes and as a consequence a breakdown of the myelin sheath which is associated with early AD (Dong et al., 2018). Notably, chronic neuroinflammation with sustained activation of microglia and astrocytes may lead to lesion in white matter tracts and disrupt the communication between neurons. Microglia activation and their associated chronic release of inflammatory cytokines in AD subjects attenuate its capacity to clear toxic and harmful substances from the brain which may also underlie the myelin damage. In addition, it was suggested that microglia and immune-related pathways can act as early mediators of synapse loss and dysfunction that occur in AD models before plaques formation. Microglia can exhibit a classically activated phenotype (M1) which exerts toxic effects by secreting proinflammatory cytokines or an alternative activated phenotype (M2) involved in the maintenance of central nervous system homeostasis, phagocytosis of apoptotic bodies or cells, releasing neurotrophic factors, and reducing proinflammatory cytokines. Studies demonstrated that M2 phenotype markers, in contrast to M1 markers, could be modulated in adult microglia, depending on the microenvironment and therefore, have a potential therapeutic effect in neuroinflammation for review see (Tang and Le, 2016.) Apart from resident microglia, another type of microglia has been identified in the brain that originates from monocyte precursor cells from the bone marrow referred as ‘microglia-like” cells. The bone marrow-derived microglia-like cells may cross the blood-brain barrier and migrate into the brain in a chemokine-dependent manner (Kawanishi et al., 2018). In AD, bone marrow-derived cells can access the Aβ-laden brain in higher numbers, as demonstrated in APP23 transgenic mice versus age-matched non-transgenic control mice. Microglia, especially bone marrow derived microglia, has been recently thought to play important roles in internalizing and phagocytizing Aβ oligomers. The invading cells exhibit hematopoietic phenotypes heterogeneously scattered throughout the brain. Hematopoietic stem cells that enter the brain affect brain homeostasis through the secretion of hematopoietic growth factors and cytokines and promote brain repair by increasing neurogenesis 
    Related Articles | Metrics
    Pharmacological strategies for treating misfolded rhodopsin-associated autosomal dominant retinitis pigmentosa
    Yibo Xi, Yuanyuan Chen
    2022, 17 (1):  110-112.  doi: 10.4103/1673-5374.314306
    Abstract ( 176 )   PDF (5876KB) ( 80 )   Save
    Mutations that cause protein misfolding are implicated in conditions such as retinitis pigmentosa (RP), Usher Syndrome, and myocilin associated primary open angle glaucoma. The aggregation and continuous degradation of a highly abundant misfolded protein add proteolytic load of the affected cells. The subtle balance of cellular homeostasis, once disrupted by an overwhelmed proteolytic system, will lead to cell death and tissue degeneration. This perspective uses RHODOPSIN (RHO)-associated RP to review pharmacologic strategies for modifying protein misfolding-associated abnormalities with the goal of bringing insights to the treatment of other proteinopathies. 
    Related Articles | Metrics
    Analyzing neural degeneration of the retina with connectomics
    Charles L. Zucker, John E. Dowling
    2022, 17 (1):  113-114.  doi: 10.4103/1673-5374.314307
    Abstract ( 191 )   PDF (11267KB) ( 46 )   Save
    Electron microscopy (EM) provides a unique ability to visualize structural detail with a resolution orders of magnitude better than other imaging techniques. Applied conventionally, its limitation is that each acquired image represents a small area with a section thickness significantly less than 100 nm. Recently, techniques have been developed that allow thousands of relatively large serial-sections to be collected and efficiently imaged at full EM resolution, with the images then being stitched back together to produce a 3D volume.  Within such a volume, every subcellular structure or cellular connection can be identified and mapped, i.e. connectomics. These methods offer the opportunity of revealing a comprehensive view of large volumes of neural tissue. With the increasing use of automated technologies, it is now possible to use large-scale serial-section electron microscopy to generate reconstructions of various brain regions with a resolution of 4 nm or better (Kasthuri et al., 2015; Baena et al., 2019). At this resolution, excitatory and inhibitory chemical synapses can be seen; the existence of electrical synapses (gap junctions), the presence of neuromodulatory peptides and biogenic amines, and the identification of local microcircuits can all be observed (Swanson and Lichtman, 2016).  Furthermore, relationships with various types of glial cells are readily seen, and cells associated with vascular elements and non-neuronal/glial cell types can be distinguished.  Further, the fine structure of organelles, including mitochondria, endoplasmic reticulum, lysosomes, and autophagosomes, along with cytoskeletal elements are within the resolution of these techniques. Thus, a continuum of scale, from sub-organelle structure, through the cellular level, up to a wide field tissue perspective can be viewed and analyzed simultaneously.  Such techniques have been used to map nerve regeneration in 3D (Leckenby et al., 2019), to investigate developmental rewiring in the cerebellum (Wilson et al., 2019); to explore the network connectivity in visual thalamus (Morgan and Lichtman, 2020), and to define the neuronal connectivity and relationships with glial cells in the human fovea (Dacey et al., 2017; Packer et al., 2017).
    Related Articles | Metrics
    Traumatic brain injury induced by exposure to blast overpressure via ear canal
    Yang Ou, Brad A. Clifton, Jinghui Li, David Sandlin, Na Li, Li Wu, Chunming Zhang, Tianwen Chen, Jun Huang, Yue Yu, Jerome Allison, Fan Fan, Richard J. Roman, James Shaffery, Wu Zhou, Yi Pang, Hong Zhu
    2022, 17 (1):  115-121.  doi: 10.4103/1673-5374.314311
    Abstract ( 199 )   PDF (8781KB) ( 45 )   Save
    Exposure to explosive shockwave often leads to blast-induced traumatic brain injury in military and civilian populations. Unprotected ears are most often damaged following exposure to blasts. Although there is an association between tympanic membrane perforation and TBI in blast exposure victims, little is known about how and to what extent blast energy is transmitted to the central nervous system via the external ear canal. The present study investigated whether exposure to blasts directed through the ear canal causes brain injury in Long-Evans rats. Animals were exposed to a single blast (0–30 pounds per square inch (psi)) through the ear canal, and brain injury was evaluated by histological and behavioral outcomes at multiple time-points. Blast exposure not only caused tympanic membrane perforation but also produced substantial neuropathological changes in the brain, including increased expression of c-Fos, induction of a profound chronic neuroinflammatory response, and apoptosis of neurons. The blast-induced injury was not limited only to the brainstem most proximal to the source of the blast, but also affected the forebrain including the hippocampus, amygdala and the habenula, which are all involved in cognitive functions. Indeed, the animals exhibited long-term neurological deficits, including signs of anxiety in open field tests 2 months following blast exposure, and impaired learning and memory in an 8-arm maze 12 months following blast exposure. These results suggest that the unprotected ear canal provides a locus for blast waves to cause TBI. This study was approved by the Institutional Animal Care and Use Committee at the University of Mississippi Medical Center (Animal protocol# 0932E, approval date: September 30, 2016 and 0932F, approval date: September 27, 2019). 
    Related Articles | Metrics
    Functionality of a bicistronic construction containing HEXA and HEXB genes encoding β-hexosaminidase A for cell-mediated therapy of GM2 gangliosidoses
    Alisa A. Shaimardanova, Daria S. Chulpanova, Valeriya V. Solovyeva, Aleksandr M. Aimaletdinov, Albert A. Rizvanov
    2022, 17 (1):  122-129.  doi: 10.4103/1673-5374.314310
    Abstract ( 221 )   PDF (1546KB) ( 148 )   Save
    Tay-Sachs disease and Sandhoff disease are severe hereditary neurodegenerative disorders caused by a deficiency of β-hexosaminidase A (HexA) enzyme, which results in the accumulation of GM2 gangliosides in the nervous system cells. In this work, we analyzed the efficacy and safety of cell-mediated gene therapy for Sandhoff disease and Sandhoff disease using a bicistronic lentiviral vector encoding cDNA of HexA α- and β-subunit genes separated by the nucleotide sequence of a P2A peptide (HEXA-HEXB). The functionality of the bicistronic construct containing the HEXA-HEXB genetic cassette was analyzed in a culture of HEK293T cells and human umbilical cord blood mononuclear cells (hUCBMCs). Our results showed that the enzymatic activity of HexA in the conditioned medium harvested from genetically modified HEK293T-HEXA-HEXB and hUCBMCs-HEXA-HEXB was increased by 23 and 8 times, respectively, compared with the conditioned medium of native cells. Western blot analysis showed that hUCBMCs-HEXA-HEXB secreted both completely separated HEXA and HEXB proteins, and an uncleaved protein containing HEXA + HEXB linked by the P2A peptide. Intravenous injection of genetically modified hUCBMCs-HEXA-HEXB to laboratory Wistar rats was carried out, and the HexA enzymatic activity in the blood plasma of experimental animals, as well as the number of live cells of immune system organs (spleen, thymus, bone marrow, lymph nodes) were determined. A significant increase in the enzymatic activity of HexA in the blood plasma of laboratory rats on days 6 and 9 (by 2.5 and 3 times, respectively) after the administration of hUCBMCs-HEXA-HEXB was shown. At the same time, the number of live cells in the studied organs remained unchanged. Thus, the functionality of the bicistronic genetic construct encoding cDNA of the HEXA and HEXB genes separated by the nucleotide sequence of the P2A peptide was shown in vitro and in vivo. We hypothesize that due to the natural ability of hUCBMCs to overcome biological barriers, such a strategy can restore the activity of the missing enzyme in the central nervous system of patients with GM2 gangliosidoses. Based on the obtained data, it can be concluded that intravenous administration of hUCBMCs with HexA overexpression is a promising method of the therapy for GM2 gangliosidoses. The animal protocol was approved by the Animal Ethics Committee of the Kazan Federal University (No. 23) on June 30, 2020.

    Related Articles | Metrics
    Cell cycle exit and neuronal differentiation 1-engineered embryonic neural stem cells enhance neuronal differentiation and neurobehavioral recovery after experimental traumatic brain injury
    Ren Wang, Dian-Xu Yang, Ying-Liang Liu, Jun Ding, Yan Guo, Wan-Hai Ding, Heng-Li Tian, Fang Yuan
    2022, 17 (1):  130-136.  doi: 10.4103/1673-5374.314316
    Abstract ( 161 )   PDF (2184KB) ( 137 )   Save
    Our previous study showed that cell cycle exit and neuronal differentiation 1 (CEND1) may participate in neural stem cell cycle exit and oriented differentiation. However, whether CEND1-transfected neural stem cells can improve the prognosis of traumatic brain injury remained unclear. In this study, we performed quantitative proteomic analysis and found that after traumatic brain injury, CEND1 expression was downregulated in mouse brain tissue. Three days after traumatic brain injury, we transplanted CEND1-transfected neural stem cells into the area surrounding the injury site. We found that at 5 weeks after traumatic brain injury, transplantation of CEND1-transfected neural stem cells markedly alleviated brain atrophy and greatly improved neurological function. In vivo and in vitro results indicate that CEND1 overexpression inhibited the proliferation of neural stem cells, but significantly promoted their neuronal differentiation. Additionally, CEND1 overexpression reduced protein levels of Notch1 and cyclin D1, but increased levels of p21 in CEND1-transfected neural stem cells. Treatment with CEND1-transfected neural stem cells was superior to similar treatment without CEND1 transfection. These findings suggest that transplantation of CEND1-transfected neural stem cells is a promising cell therapy for traumatic brain injury. This study was approved by the Animal Ethics Committee of the School of Biomedical Engineering of Shanghai Jiao Tong University, China (approval No. 2016034) on November 25, 2016. 
    Related Articles | Metrics
    Ki20227 aggravates apoptosis, inflammatory response, and oxidative stress after focal cerebral ischemia injury
    Cheng Jiang, Ze-Ning Wang, Yu-Chen Kang, Yi Chen, Wei-Xin Lu, Hai-Jun Ren, Bo-Ru Hou
    2022, 17 (1):  137-143.  doi: 10.4103/1673-5374.314318
    Abstract ( 187 )   PDF (8320KB) ( 28 )   Save
    The survival of microglia depends on the colony-stimulating factor-1 receptor (CSF1R) signaling pathway under physiological conditions. Ki20227 is a highly selective CSF1R inhibitor that has been shown to change the morphology of microglia. However, the effects of Ki20227 on the progression of ischemic stroke are unclear. In this study, male C57BL/6 mouse models of focal cerebral ischemic injury were established through the occlusion of the middle cerebral artery and then administered 3 mg/g Ki20227 for 3 successive days. The results revealed that the number of ionized calcium-binding adaptor molecule 1/bromodeoxyuridine double positive cells in the infarct tissue was reduced, the degree of edema was increased, neurological deficits were aggravated, infarct volume was increased, and the number of peri-infarct Nissl bodies was reduced. The number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells in the peri-infarct tissue was increased. The expression levels of Bax and Cleaved caspase-3 were up-regulated. Bcl-2 expression was downregulated. The expression levels of inflammatory factors and oxidative stress-associated factors were increased. These findings suggested that Ki20227 blocked microglial proliferation and aggravated the pathological progression of ischemia/reperfusion injury in a transient middle cerebral artery occlusion model. This study was approved by the Animal Ethics Committee of Lanzhou University Second Hospital (approval No. D2020-68) on March 6, 2020.
    Related Articles | Metrics
    Down-regulating Circular RNA Prkcsh suppresses the inflammatory response after spinal cord injury
    Jia-Nan Chen, Yi-Ning Zhang, Li-Ge Tian, Ying Zhang, Xin-Yu Li, Bin Ning
    2022, 17 (1):  144-151.  doi: 10.4103/1673-5374.314114
    Abstract ( 197 )   PDF (2775KB) ( 132 )   Save
    Circular RNAs (circRNAs) are a class of conserved, endogenous non-coding RNAs that are involved in transcriptional and post-transcriptional gene regulation and are highly enriched in the nervous system. They participate in the survival and differentiation of multiple nerve cells, and may even promote the recovery of neurological function after stroke. However, their role in the inflammatory response after spinal cord injury remains unclear. In the present study, we established a mouse model of T9 spinal cord injury using the modified Allen’s impact method, and identified 16,013 circRNAs and 960 miRNAs that were differentially expressed after spinal cord injury. Of these, the expression levels of circPrkcsh were significantly different between injured and sham-treated mice. We then treated astrocytes with tumor necrosis factor-α in vitro to simulate the inflammatory response after spinal cord injury. Our results revealed an elevated expression of circPrkcsh with a concurrent decrease in miR-488 expression in injured cells. We also found that circPrkcsh regulated the expression of the inflammation-related gene Ccl2. Furthermore, in tumor necrosis factor-α-treated astrocytes, circPrkcsh knockdown decreased the expression of Ccl2 by upregulating miR-488 expression, and reduced the secretion of inflammatory cytokines in vitro. These findings suggest that differentially expressed circRNAs participate in the inflammatory response after spinal cord injury and act as the regulators of certain microRNAs. Furthermore, circPrkcsh may be used as an miR-488 sponge to regulate Ccl2 expression, which might provide a new potential therapy for SCI. The study was approved by the Animal Ethics Committee of Shandong University of China (approval No. KYLL-20170303) on March 3, 2017.
    Related Articles | Metrics
    Neural stem cell transplantation alleviates functional cognitive deficits in a mouse model of tauopathy
    He-Ao Zhang, Chun-Xu Yuan, Ke-Fu Liu, Qi-Fan Yang, Juan Zhao, Hui Li, Qing-Hu Yang, Da Song, Zhen-Zhen Quan, Hong Qing
    2022, 17 (1):  152-162.  doi: 10.4103/1673-5374.314324
    Abstract ( 196 )   PDF (2393KB) ( 119 )   Save
    The mechanisms of the transplantation of neural stem cells (NSCs) in the treatment of Alzheimer’s disease remain poorly understood. In this study, NSCs were transplanted into the hippocampal CA1 region of the rTg (tau P301L) 4510 mouse model, a tauopathy model that is thought to reflect the tau pathology associated with Alzheimer’s disease. The results revealed that NSC transplantation reduced the abnormal aggregation of tau, resulting in significant improvements in the short-term memory of the tauopathy model mice. Compared with wild-type and phosphate-buffered saline (PBS)-treated mice, mice that received NSC transplantations were characterized by changes in the expression of multiple proteins in brain tissue, particularly those related to the regulation of tau aggregation or misfolding. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) function analysis revealed that these proteins were primarily enriched in pathways associated with long-term potentiation, neurogenesis, and other neurobiological processes. Changes in the expression levels of key proteins were verified by western blot assays. These data provided clues to improve the understanding of the functional capacity associated with NSC transplantation in Alzheimer’s disease treatment. This study was approved by the Beijing Animal Ethics Association and Ethics Committee of Beijing Institute of Technology (approval No. SYXK-BIT-school of life science-2017-M03) in 2017.
    Related Articles | Metrics
    Neat1 decreases neuronal apoptosis after oxygen and glucose deprivation
    Wei-Na Chai, Yi-Fan Wu, Zhi-Min Wu, Yan-Feng Xie, Quan-Hong Shi, Wei Dan, Yan Zhan, Jian-Jun Zhong, Wei Tang, Xiao-Chuan Sun, Li Jiang
    2022, 17 (1):  163-169.  doi: 10.4103/1673-5374.314313
    Abstract ( 234 )   PDF (6740KB) ( 134 )   Save
    Studies have shown that downregulation of nuclear-enriched autosomal transcript 1 (Neat1) may adversely affect the recovery of nerve function and the increased loss of hippocampal neurons in mice. Whether Neat1 has protective or inhibitory effects on neuronal cell apoptosis after secondary brain injury remains unclear. Therefore, the effects of Neat1 on neuronal apoptosis were observed. C57BL/6 primary neurons were obtained from the cortices of newborn mice and cultured in vitro, and an oxygen and glucose deprivation cell model was established to simulate the secondary brain injury that occurs after traumatic brain injury in vitro. The level of Neat1 expression in neuronal cells was regulated by constructing a recombinant adenovirus to infect neurons, and the effects of Neat1 expression on neuronal apoptosis after oxygen and glucose deprivation were observed. The experiment was divided into four groups: the control group, without any treatment, received normal culture; the oxygen and glucose deprivation group were subjected to the oxygen and glucose deprivation model protocol; the Neat1 overexpression and Neat1 downregulation groups were treated with Neat1 expression intervention techniques and were subjected to the in oxygen and glucose deprivation protocol. The protein expression levels of neurons p53-induced death domain protein 1 (PIDD1, a pro-apoptotic protein), caspase-2 (an apoptotic priming protein), cytochrome C (a pro-apoptotic protein), and cleaved caspase-3 (an apoptotic executive protein) were measured in each group using the western blot assay. To observe changes in the intracellular distribution of cytochrome C, the expression levels of cytochrome C in the cytoplasm and mitochondria of neurons from each group were detected by western blot assay. Differences in the cell viability and apoptosis rate between groups were detected by cell-counting kit 8 assay and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, respectively. The results showed that the apoptosis rate, PIDD1, caspase-2, and cleaved caspase-3 expression levels significantly decreased, and cell viability significantly improved in the Neat1 overexpression group compared with the oxygen and glucose deprivation group; however, Neat1 downregulation reversed these changes. Compared with the Neat1 downregulation group, the cytosolic cytochrome C level in the Neat1 overexpression group significantly decreased, and the mitochondrial cytochrome C level significantly increased. These data indicate that Neat1 upregulation can reduce the release of cytochrome C from the mitochondria to the cytoplasm by inhibiting the PIDD1-caspase-2 pathway, reducing the activation of caspase-3, and preventing neuronal apoptosis after oxygen and glucose deprivation, which might reduce secondary brain injury after traumatic brain injury. All experiments were approved by the Animal Ethics Committee of the First Affiliated Hospital of Chongqing Medical University, China, on December 19, 2020 (approval No. 2020-895). 
    Related Articles | Metrics
    Ghrelin alleviates 6-hydroxydopamine-induced neurotoxicity in SH-SY5Y cells
    Xin He, Wei Yuan, Chun-Qing Yang, Lu Zhu, Fei Liu, Juan Feng, Yi-Xue Xue
    2022, 17 (1):  170-177.  doi: 10.4103/1673-5374.314314
    Abstract ( 252 )   PDF (3064KB) ( 190 )   Save
    Ghrelin is a neuropeptide that has various physiological functions and has been demonstrated to be neuroprotective in a number of neurological disease models. However, the underlying mechanisms of ghrelin in Parkinson’s disease remain largely unexplored. The current study aimed to study the effects of ghrelin in a 6-hydroxydopamine (6-OHDA)-induced Parkinson’s disease model and evaluate the potential underlying mechanisms. In the present study, we treated an SH-SY5Y cell model with 6-OHDA, and observed that pretreatment with different concentrations of ghrelin (1, 10, and 100 nM) for 30 minutes relieved the neurotoxic effects of 6-OHDA, as revealed by Cell Counting Kit-8 and Annexin V/propidium iodide (PI) apoptosis assays. Reverse transcription quantitative polymerase chain reaction and western blot assay results demonstrated that 6-OHDA treatment upregulated α-synuclein and lincRNA-p21 and downregulated TG-interacting factor 1 (TGIF1), which was predicted as a potential transcription regulator of the gene encoding α-synuclein (SNCA). Ghrelin pretreatment was able to reverse the trends caused by 6-OHDA. The Annexin V/PI apoptosis assay results revealed that inhibiting either α-synuclein or lincRNA-p21 expression with small interfering RNA (siRNA) relieved 6-OHDA-induced cell apoptosis. Furthermore, inhibiting lincRNA-p21 also partially upregulated TGIF1. By retrieving information from a bioinformatics database and performing both double luciferase and RNA immunoprecipitation assays, we found that lincRNA-p21 and TGIF1 were able to form a double-stranded RNA-binding protein Staufen homolog 1 (STAU1) binding site and further activate the STAU1-mediated mRNA decay pathway. In addition, TGIF1 was able to transcriptionally regulate α-synuclein expression by binding to the promoter of SNCA. The Annexin V/PI apoptosis assay results showed that either knockdown of TGIF1 or overexpression of lincRNA-p21 notably abolished the neuroprotective effects of ghrelin against 6-OHDA-induced neurotoxicity. Collectively, these findings suggest that ghrelin exerts neuroprotective effects against 6-OHDA-induced neurotoxicity via the lincRNA-p21/TGIF1/α-synuclein pathway. 
    Related Articles | Metrics
    Polygalasaponin F protects hippocampal neurons against glutamate-induced cytotoxicity
    Chong Sun, Xin-Cheng Cao, Zhi-Yang Liu, Chao-Lin Ma, Bao-Ming Li
    2022, 17 (1):  178-184.  doi: 10.4103/1673-5374.314321
    Abstract ( 185 )   PDF (1332KB) ( 94 )   Save
    Excess extracellular glutamate leads to excitotoxicity, which induces neuronal death through the overactivation of N-methyl-D-aspartate receptors (NMDARs). Excitotoxicity is thought to be closely related to various acute and chronic neurological disorders, such as stroke and Alzheimer’s disease. Polygalasaponin F (PGSF) is a triterpenoid saponin monomer that can be isolated from Polygala japonica, and has been reported to protect cells against apoptosis. To investigate the mechanisms underlying the neuroprotective effects of PGSF against glutamate-induced cytotoxicity, PGSF-pretreated hippocampal neurons were exposed to glutamate for 24 hours. The results demonstrated that PGSF inhibited glutamate-induced hippocampal neuron death in a concentration-dependent manner and reduced glutamate-induced Ca2+ overload in the cultured neurons. In addition, PGSF partially blocked the excess activity of NMDARs, inhibited both the downregulation of NMDAR subunit NR2A expression and the upregulation of NMDAR subunit NR2B expression, and upregulated the expression of phosphorylated cyclic adenosine monophosphate-responsive element-binding protein and brain-derived neurotrophic factor. These findings suggest that PGSF protects cultured hippocampal neurons against glutamate-induced cytotoxicity by regulating NMDARs. The study was approved by the Institutional Animal Care Committee of Nanchang University (approval No. 2017-0006) on December 29, 2017.
    Related Articles | Metrics
    Neuroprotective effects of Alda-1 mitigate spinal cord injury in mice: involvement of Alda-1-induced ALDH2 activation-mediated suppression of reactive aldehyde mechanisms
    Mushfiquddin Khan, Fei Qiao, Pavan Kumar, S.M. Touhidul Islam, Avtar K. Singh, Jeseong Won, Inderjit Singh
    2022, 17 (1):  185-193.  doi: 10.4103/1673-5374.314312
    Abstract ( 198 )   PDF (4235KB) ( 154 )   Save
    Spinal cord injury (SCI) is associated with high production and excessive accumulation of pathological 4-hydroxy-trans-2-nonenal (4-HNE), a reactive aldehyde, formed by SCI-induced metabolic dysregulation of membrane lipids. Reactive aldehyde load causes redox alteration, neuroinflammation, neurodegeneration, pain-like behaviors, and locomotion deficits. Pharmacological scavenging of reactive aldehydes results in limited improved motor and sensory functions. In this study, we targeted the activity of mitochondrial enzyme aldehyde dehydrogenase 2 (ALDH2) to detoxify 4-HNE for accelerated functional recovery and improved pain-like behavior in a male mouse model of contusion SCI. N-(1,3-benzodioxol-5-ylmethyl)-2,6-dichlorobenzamide (Alda-1), a selective activator of ALDH2, was used as a therapeutic tool to suppress the 4-HNE load. SCI was induced by an impactor at the T9–10 vertebral level. Injured animals were initially treated with Alda-1 at 2 hours after injury, followed by once-daily treatment with Alda-1 for 30 consecutive days. Locomotor function was evaluated by the Basso Mouse Scale, and pain-like behaviors were assessed by mechanical allodynia and thermal algesia. ALDH2 activity was measured by enzymatic assay. 4-HNE protein adducts and enzyme/protein expression levels were determined by western blot analysis and histology/immunohistochemistry. SCI resulted in a sustained and prolonged overload of 4-HNE, which parallels with the decreased activity of ALDH2 and low functional recovery. Alda-1 treatment of SCI decreased 4-HNE load and enhanced the activity of ALDH2 in both the acute and the chronic phases of SCI. Furthermore, the treatment with Alda-1 reduced neuroinflammation, oxidative stress, and neuronal loss and increased adenosine 5′-triphosphate levels stimulated the neurorepair process and improved locomotor and sensory functions. Conclusively, the results provide evidence that enhancing the ALDH2 activity by Alda-1 treatment of SCI mice suppresses the 4-HNE load that attenuates neuroinflammation and neurodegeneration, promotes the neurorepair process, and improves functional outcomes. Consequently, we suggest that Alda-1 may have therapeutic potential for the treatment of human SCI. Animal procedures were approved by the Institutional Animal Care and Use Committee (IACUC) of MUSC (IACUC-2019-00864) on December 21, 2019.
    Related Articles | Metrics
    Exosomes derived from bone marrow mesenchymal stem cells protect the injured spinal cord by inhibiting pericyte pyroptosis
    Yan Zhou, Lu-Lu Wen, Yan-Fei Li, Kai-Min Wu, Ran-Ran Duan, Yao-Bing Yao, Li-Jun Jing, Zhe Gong, Jun-Fang Teng, Yan-Jie Jia
    2022, 17 (1):  194-202.  doi: 10.4103/1673-5374.314323
    Abstract ( 249 )   PDF (3945KB) ( 314 )   Save
    Mesenchymal stem cell (MSC) transplantation is a promising treatment strategy for spinal cord injury, but immunological rejection and possible tumor formation limit its application. The therapeutic effects of MSCs mainly depend on their release of soluble paracrine factors. Exosomes are essential for the secretion of these paracrine effectors. Bone marrow mesenchymal stem cell-derived exosomes (BMSC-EXOs) can be substituted for BMSCs in cell transplantation. However, the underlying mechanisms remain unclear. In this study, a rat model of T10 spinal cord injury was established using the impact method. Then, 30 minutes and 1 day after spinal cord injury, the rats were administered 200 μL exosomes via the tail vein (200 μg/mL; approximately 1 × 106 BMSCs). Treatment with BMSC-EXOs greatly reduced neuronal cell death, improved myelin arrangement and reduced myelin loss, increased pericyte/endothelial cell coverage on the vascular wall, decreased blood-spinal cord barrier leakage, reduced caspase 1 expression, inhibited interleukin-1β release, and accelerated locomotor functional recovery in rats with spinal cord injury. In the cell culture experiment, pericytes were treated with interferon-γ and tumor necrosis factor-α. Then, Lipofectamine 3000 was used to deliver lipopolysaccharide into the cells, and the cells were co-incubated with adenosine triphosphate to simulate injury in vitro. Pre-treatment with BMSC-EXOs for 8 hours greatly reduced pericyte pyroptosis and increased pericyte survival rate. These findings suggest that BMSC-EXOs may protect pericytes by inhibiting pyroptosis and by improving blood-spinal cord barrier integrity, thereby promoting the survival of neurons and the extension of nerve fibers, and ultimately improving motor function in rats with spinal cord injury. All protocols were conducted with the approval of the Animal Ethics Committee of Zhengzhou University on March 16, 2019.
    Related Articles | Metrics
    Lycium barbarum extract promotes M2 polarization and reduces oligomeric amyloid-β-induced inflammatory reactions in microglial cells
    Zhong-Qing Sun, Jin-Feng Liu, Wei Luo, Ching-Hin Wong, Kwok-Fai So, Yong Hu, Kin Chiu
    2022, 17 (1):  203-209.  doi: 10.4103/1673-5374.314325
    Abstract ( 197 )   PDF (40461KB) ( 105 )   Save
    Lycium barbarum (LB) is a traditional Chinese medicine that has been demonstrated to exhibit a wide variety of biological functions, such as antioxidation, neuroprotection, and immune modulation. One of the main mechanisms of Alzheimer’s disease is that microglia activated by amyloid beta (Aβ) transform from the resting state to an M1 state and release pro-inflammatory cytokines to the surrounding environment. In the present study, immortalized microglial cells were pretreated with L. barbarum extract for 1 hour and then treated with oligomeric Aβ for 23 hours. The results showed that LB extract significantly increased the survival of oligomeric Aβ-induced microglial cells, downregulated the expression of M1 pro-inflammatory markers (inducible nitric oxide synthase, tumor necrosis factor α, interleukin-6, and interleukin-1β), and upregulated the expression of M2 anti-inflammatory markers (arginase-1, chitinase-like protein 3, and interleukin-4). LB extract also inhibited the oligomeric Aβ-induced secretion of tumor necrosis factor α, interleukin-6, and interleukin-1β in microglial cells. The results of in vitro cytological experiments suggest that, in microglial cells, LB extract can inhibit oligomeric Aβ-induced M1 polarization and concomitant inflammatory reactions, and promote M2 polarization.
    Related Articles | Metrics
    Intranasal insulin ameliorates neurological impairment after intracerebral hemorrhage in mice
    Yuan Zhu, Yi Huang, Jin Yang, Rong Tu, Xin Zhang, Wei-Wei He, Chang-Yue Hou, Xiao-Ming Wang, Ju-Ming Yu, Guo-Hui Jiang
    2022, 17 (1):  210-216.  doi: 10.4103/1673-5374.314320
    Abstract ( 191 )   PDF (2666KB) ( 138 )   Save
    In Alzheimer’s disease and ischemic stroke, intranasal insulin can act as a neuroprotective agent. However, whether intranasal insulin has a neuroprotective effect in intracerebral hemorrhage and its potential mechanisms remain poorly understood. In this study, a mouse model of autologous blood-induced intracerebral hemorrhage was treated with 0.5, 1, or 2 IU insulin via intranasal delivery, twice per day, until 24 or 72 hours after surgery. Compared with saline treatment, 1 IU intranasal insulin treatment significantly reduced hematoma volume and brain edema after cerebral hemorrhage, decreased blood-brain barrier permeability and neuronal degeneration damage, reduced neurobehavioral deficits, and improved the survival rate of mice. Expression levels of p-AKT and p-GSK3β were significantly increased in the perihematoma tissues after intranasal insulin therapy. Our findings suggest that intranasal insulin therapy can protect the neurological function of mice after intracerebral hemorrhage through the AKT/GSK3β signaling pathway. The study was approved by the Ethics Committee of the North Sichuan Medical College of China (approval No. NSMC(A)2019(01)) on January 7, 2019.
    Related Articles | Metrics
    Inhibition of LncRNA Vof-16 expression promotes nerve regeneration and functional recovery after spinal cord injury 
    Xiao-Min Zhang, Li-Ni Zeng, Wan-Yong Yang, Lu Ding, Kang-Zhen Chen, Wen-Jin Fu, Si-Quan Zeng, Yin-Ru Liang, Gan-Hai Chen, Hong-Fu Wu
    2022, 17 (1):  217-227.  doi: 10.4103/1673-5374.314322
    Abstract ( 221 )   PDF (24041KB) ( 74 )   Save
    Our previous RNA sequencing study showed that the long non-coding RNA ischemia-related factor Vof-16 (lncRNA Vof-16) was upregulated after spinal cord injury, but its precise role in spinal cord injury remains unclear. Bioinformatics predictions have indicated that lncRNA Vof-16 may participate in the pathophysiological processes of inflammation and apoptosis. PC12 cells were transfected with a pHBLV-U6-MCS-CMV-ZsGreen-PGK-PURO vector to express an lncRNA Vof-16 knockdown lentivirus and a pHLV-CMVIE-ZsGree-Puro vector to express an lncRNA Vof-16 overexpression lentivirus. The overexpression of lncRNA Vof-16 inhibited PC12 cell survival, proliferation, migration, and neurite extension, whereas lncRNA Vof-16 knockdown lentiviral vector resulted in the opposite effects in PC12 cells. Western blot assay results showed that the overexpression of lncRNA Vof-16 increased the protein expression levels of interleukin 6, tumor necrosis factor-α, and Caspase-3 and decreased Bcl-2 expression levels in PC12 cells. Furthermore, we established rat models of spinal cord injury using the complete transection at T10. Spinal cord injury model rats were injected with the lncRNA Vof-16 knockdown or overexpression lentiviral vectors immediately after injury. At 7 days after spinal cord injury, rats treated with lncRNA Vof-16 knockdown displayed increased neuronal survival and enhanced axonal extension. At 8 weeks after spinal cord injury, rats treated with the lncRNA Vof-16 knockdown lentiviral vector displayed improved neurological function in the hind limb. Notably, lncRNA Vof-16 knockdown injection increased Bcl-2 expression and decreased tumor necrosis factor-α and Caspase-3 expression in treated animals. Rats treated with the lncRNA Vof-16 overexpression lentiviral vector displayed opposite trends. These findings suggested that lncRNA Vof-16 is associated with the regulation of inflammation and apoptosis. The inhibition of lncRNA Vof-16 may be useful for promoting nerve regeneration and functional recovery after spinal cord injury. The experiments were approved by the Institutional Animal Care and Use Committee of Guangdong Medical University, China.

    Related Articles | Metrics
    The anatomical, electrophysiological and histological observations of muscle contraction units in rabbits: a new perspective on nerve injury and regeneration
    Ting-Min Xu, Bo Chen, Zong-Xue Jin, Xiao-Feng Yin, Pei-Xun Zhang, Bao-Guo Jiang
    2022, 17 (1):  228-232.  doi: 10.4103/1673-5374.315228
    Abstract ( 253 )   PDF (1418KB) ( 119 )   Save
    In the conventional view a muscle is composed of intermediate structures before its further division into microscopic muscle fibers. Our experiments in mice have confirmed this intermediate structure is composed of the lamella cluster formed by motor endplates, the innervating nerve branches and the corresponding muscle fibers, which can be viewed as an independent structural and functional unit. In this study, we verified the presence of these muscle construction units in rabbits. The results showed that the muscular branch of the femoral nerve sent out 4–6 nerve branches into the quadriceps and the tibial nerve sent out 4–7 nerve branches into the gastrocnemius. When each nerve branch of the femoral nerve was stimulated from the most lateral to the medial, the contraction of the lateral muscle, intermediate muscle and medial muscle of the quadriceps could be induced by electrically stimulating at least one nerve branch. When stimulating each nerve branch of the tibial nerve from the lateral to the medial, the muscle contraction of the lateral muscle 1, lateral muscle 2, lateral muscle 3 and medial muscle of the gastrocnemius could be induced by electrically stimulating at least one nerve branch. Electrical stimulation of each nerve branch resulted in different electromyographical waves recorded in different muscle subgroups. Hematoxylin-eosin staining showed most of the nerve branches around the neuromuscular junctions consisted of one individual neural tract, a few consisted of two or more neural tracts. The muscles of the lower limb in the rabbit can be subdivided into different muscle subgroups, each innervated by different nerve branches, thereby allowing much more complex muscle activities than traditionally stated. Together, the nerve branches and the innervated muscle subgroups can be viewed as an independent structural and functional unit. This study was approved by the Animal Ethics Committee of Peking University People’s Hospital (approval No. 2019PHE027) on October 20, 2019.
    Related Articles | Metrics