Loading...

Table of Content

    15 December 2023, Volume 18 Issue 12 Previous Issue    Next Issue
    For Selected: Toggle Thumbnails
    The importance of laminin at the blood-brain barrier
    Sebok K. Halder, Arjun Sapkota, Richard Milner
    2023, 18 (12):  2557-2563.  doi: 10.4103/1673-5374.373677
    Abstract ( 135 )   PDF (731KB) ( 105 )   Save
    The blood-brain barrier is a unique property of central nervous system blood vessels that protects sensitive central nervous system cells from potentially harmful blood components. The mechanistic basis of this barrier is found at multiple levels, including the adherens and tight junction proteins that tightly bind adjacent endothelial cells and the influence of neighboring pericytes, microglia, and astrocyte endfeet. In addition, extracellular matrix components of the vascular basement membrane play a critical role in establishing and maintaining blood-brain barrier integrity, not only by providing an adhesive substrate for blood-brain barrier cells to adhere to, but also by providing guidance cues that strongly influence vascular cell behavior. The extracellular matrix protein laminin is one of the most abundant components of the basement membrane, and several lines of evidence suggest that it plays a key role in directing blood-brain barrier behavior. In this review, we describe the basic structure of laminin and its receptors, the expression patterns of these molecules in central nervous system blood vessels and how they are altered in disease states, and most importantly, how genetic deletion of different laminin isoforms or their receptors reveals the contribution of these molecules to blood-brain barrier function and integrity. Finally, we discuss some of the important unanswered questions in the field and provide a “to-do” list of some of the critical outstanding experiments. 
    Related Articles | Metrics
    Repair and regeneration of peripheral nerve injuries that ablate branch points
    JuliAnne E. Allgood, George D. Bittner, Jared S. Bushman
    2023, 18 (12):  2564-2568.  doi: 10.4103/1673-5374.373679
    Abstract ( 118 )   PDF (1508KB) ( 92 )   Save
    The peripheral nervous system has an extensive branching organization, and peripheral nerve injuries that ablate branch points present a complex challenge for clinical repair. Ablations of linear segments of the PNS have been extensively studied and routinely treated with autografts, acellular nerve allografts, conduits, wraps, and nerve transfers. In contrast, segmental-loss peripheral nerve injuries, in which one or more branch points are ablated so that there are three or more nerve endings, present additional complications that have not been rigorously studied or documented. This review discusses: (1) the branched anatomy of the peripheral nervous system, (2) case reports describing how peripheral nerve injuries with branched ablations have been surgically managed, (3) factors known to influence regeneration through branched nerve structures, (4) techniques and models of branched peripheral nerve injuries in animal models, and (5) conclusions regarding outcome measures and studies needed to improve understanding of regeneration through ablated branched structures of the peripheral nervous system. 
    Related Articles | Metrics
    Cytokines, synaptic plasticity and network dynamics: a matter of balance#br#
    Laura Bellingacci, Jacopo Canonichesi#, Andrea Mancini, Lucilla Parnetti, Massimiliano Di Filippo
    2023, 18 (12):  2569-2572.  doi: 10.4103/1673-5374.371344
    Abstract ( 147 )   PDF (538KB) ( 110 )   Save
    The modern view of the immune system as a sensitizing and modulating machinery of the central nervous system is now well recognized. However, the specific mechanisms underlying this fine crosstalk have yet to be fully disentangled. To control cognitive function and behavior, the two systems are engaged in a subtle interacting act. In this scenario, a dual action of pro-inflammatory cytokines in the modulation of brain network connections is emerging. Pro-inflammatory cytokines are indeed required to express physiological plasticity in the hippocampal network while being detrimental when over-expressed during uncontrolled inflammatory processes. In this dynamic equilibrium, synaptic functioning and the performance of neural networks are ensured by maintaining an appropriate balance between pro- and anti-inflammatory molecules in the central nervous system microenvironment.
    Related Articles | Metrics
    Axonal growth inhibitors and their receptors in spinal cord injury: from biology to clinical translation
    Sílvia Sousa Chambel, Célia Duarte Cruz
    2023, 18 (12):  2573-2581.  doi: 10.4103/1673-5374.373674
    Abstract ( 162 )   PDF (1006KB) ( 132 )   Save
    Axonal growth inhibitors are released during traumatic injuries to the adult mammalian central nervous system, including after spinal cord injury. These molecules accumulate at the injury site and form a highly inhibitory environment for axonal regeneration. Among these inhibitory molecules, myelin-associated inhibitors, including neurite outgrowth inhibitor A, oligodendrocyte myelin glycoprotein, myelin-associated glycoprotein, chondroitin sulfate proteoglycans and repulsive guidance molecule A are of particular importance. Due to their inhibitory nature, they represent exciting molecular targets to study axonal inhibition and regeneration after central injuries. These molecules are mainly produced by neurons, oligodendrocytes, and astrocytes within the scar and in its immediate vicinity. They exert their effects by binding to specific receptors, localized in the membranes of neurons. Receptors for these inhibitory cues include Nogo receptor 1, leucine-rich repeat, and Ig domain containing 1 and p75 neurotrophin receptor/tumor necrosis factor receptor superfamily member 19 (that form a receptor complex that binds all myelin-associated inhibitors), and also paired immunoglobulin-like receptor B. Chondroitin sulfate proteoglycans and repulsive guidance molecule A bind to Nogo receptor 1, Nogo receptor 3, receptor protein tyrosine phosphatase σ and leucocyte common antigen related phosphatase, and neogenin, respectively. Once activated, these receptors initiate downstream signaling pathways, the most common amongst them being the RhoA/ROCK signaling pathway. These signaling cascades result in actin depolymerization, neurite outgrowth inhibition, and failure to regenerate after spinal cord injury. Currently, there are no approved pharmacological treatments to overcome spinal cord injuries other than physical rehabilitation and management of the array of symptoms brought on by spinal cord injuries. However, several novel therapies aiming to modulate these inhibitory proteins and/or their receptors are under investigation in ongoing clinical trials. Investigation has also been demonstrating that combinatorial therapies of growth inhibitors with other therapies, such as growth factors or stem-cell therapies, produce stronger results and their potential application in the clinics opens new venues in spinal cord injury treatment. 
    Related Articles | Metrics
    The role of natural flavonoids on neuroinflammation as a therapeutic target for Alzheimer’s disease: a narrative review
    Qian Zhang, Yaping Yan
    2023, 18 (12):  2582-2591.  doi: 10.4103/1673-5374.373680
    Abstract ( 334 )   PDF (1041KB) ( 208 )   Save
    Alzheimer’s disease is a neurodegenerative disease that affects a large proportion of older adult people and is characterized by memory loss, progressive cognitive impairment, and various behavioral disturbances. Although the pathological mechanisms underlying Alzheimer’s disease are complex and remain unclear, previous research has identified two widely accepted pathological characteristics: extracellular neuritic plaques containing amyloid beta peptide, and intracellular neurofibrillary tangles containing tau. Furthermore, research has revealed the significant role played by neuroinflammation over recent years. The inflammatory microenvironment mainly consists of microglia, astrocytes, the complement system, chemokines, cytokines, and reactive oxygen intermediates; collectively, these factors can promote the pathological process and aggravate the severity of Alzheimer’s disease. Therefore, the development of new drugs that can target neuroinflammation will be a significant step forward for the treatment of Alzheimer’s disease. Flavonoids are plant-derived secondary metabolites that possess various bioactivities. Previous research found that multiple natural flavonoids could exert satisfactory treatment effects on the neuroinflammation associated with Alzheimer’s disease. In this review, we describe the pathogenesis and neuroinflammatory processes of Alzheimer’s disease, and summarize the effects and mechanisms of 13 natural flavonoids (apigenin, luteolin, naringenin, quercetin, morin, kaempferol, fisetin, isoquercitrin, astragalin, rutin, icariin, mangiferin, and anthocyanin) derived from plants or medicinal herbs on neuroinflammation in Alzheimer’s disease. As an important resource for the development of novel compounds for the treatment of critical diseases, it is essential that we focus on the exploitation of natural products. In particular, it is vital that we investigate the effects of flavonoids on the neuroinflammation associated with Alzheimer’s disease in greater detail. 
    Related Articles | Metrics
    Thinking outside the black box: are the brain endothelial cells the new main target in Alzheimer’s disease?
    Enrique Estudillo, Adolfo López-Ornelas, Alejandro Rodríguez-Oviedo, Neptali Gutiérrez de la Cruz, Marco Antonio Vargas-Hernández, Adriana Jiménez
    2023, 18 (12):  2592-2598.  doi: 10.4103/1673-5374.373672
    Abstract ( 111 )   PDF (649KB) ( 85 )   Save
    The blood-brain barrier is the interface through which the brain interacts with the milieu and consists mainly of a sophisticated network of brain endothelial cells that forms blood vessels and selectively moves molecules inside and outside the brain through multiple mechanisms of transport. Although brain endothelial cell function is crucial for brain homeostasis, their role in neurodegenerative diseases has historically not been considered with the same importance as other brain cells such as microglia, astroglia, neurons, or even molecules such as amyloid beta, Tau, or alpha-synuclein. Alzheimer’s disease is the most common neurodegenerative disease, and brain endothelial cell dysfunction has been reported by several groups. However, its impairment has barely been considered as a potential therapeutic target. Here we review the most recent advances in the relationship between Alzheimer’s disease and brain endothelial cells commitment and analyze the possible mechanisms through which their alterations contribute to this neurodegenerative disease, highlighting their inflammatory phenotype and the possibility of an impaired secretory pattern of brain endothelial cells that could contribute to the progression of this ailment. Finally, we discuss why shall brain endothelial cells be appreciated as a therapeutic target instead of solely an obstacle for delivering treatments to the injured brain in Alzheimer’s disease.
    Related Articles | Metrics
    CDP-choline to promote remyelination in multiple sclerosis: the need for a clinical trial
    Viktoria Gudi, Paweł Grieb, Ralf A. Linker, Thomas Skripuletz
    2023, 18 (12):  2599-2605.  doi: 10.4103/1673-5374.373671
    Abstract ( 129 )   PDF (1116KB) ( 64 )   Save
    Multiple sclerosis is a multifactorial chronic inflammatory disease of the central nervous system that leads to demyelination and neuronal cell death, resulting in functional disability. Remyelination is the natural repair process of demyelination, but it is often incomplete or fails in multiple sclerosis. Available therapies reduce the inflammatory state and prevent clinical relapses. However, therapeutic approaches to increase myelin repair in humans are not yet available. The substance cytidine-5′-diphosphocholine, CDP-choline, is ubiquitously present in eukaryotic cells and plays a crucial role in the synthesis of cellular phospholipids. Regenerative properties have been shown in various animal models of diseases of the central nervous system. We have already shown that the compound CDP-choline improves myelin regeneration in two animal models of multiple sclerosis. However, the results from the animal models have not yet been studied in patients with multiple sclerosis. In this review, we summarise the beneficial effects of CDP-choline on biolipid metabolism and turnover with regard to inflammatory and regenerative processes. We also explain changes in phospholipid and sphingolipid homeostasis in multiple sclerosis and suggest a possible therapeutic link to CDP-choline. 
    Related Articles | Metrics
    Saliva: a challenging human fluid to diagnose brain disorders with a focus on Alzheimer’s disease
    Christine Zürcher, Christian Humpel
    2023, 18 (12):  2606-2610.  doi: 10.4103/1673-5374.373675
    Abstract ( 129 )   PDF (641KB) ( 172 )   Save
    Biomarkers are molecules of biological processes that help in both the diagnosis of human diseases and in follow-up assessments of therapeutic responses. Biomarkers can be measured in many human fluids, such as blood, cerebrospinal fluid, urine, and saliva. The -omics methods (genomics, RNomics, proteomics, and metabolomics) are useful at measuring thousands of markers in a small volume. Saliva is a human fluid that is easily accessible, without any ethical concerns. Yet, saliva remains unexplored in regard to many human disease biomarkers. In this review, we will give an overview on saliva and how it can be influenced by exogenous factors. As we focus on the potential use of saliva as a diagnostic tool in brain disorders (especially Alzheimer’s disease), we will cover how saliva is linked to the brain. We will discuss that saliva is a heterogeneous human fluid, yet useful for the discovery of biomarkers in human disorders. However, a procedure and consensus that is controlled, validated, and standardized for the collection and processing of saliva is required, followed by a highly sensitive diagnostic approach.
    Related Articles | Metrics
    Gut-microbiome-brain axis: the crosstalk between the vagus nerve, alpha-synuclein and the brain in Parkinson’s disease
    Júlio César Claudino dos Santos, Leandro Freitas Oliveira, Felipe Micelli Noleto, Camilla Teixeira Pinheiro Gusmão, Gerly Anne de Castro Brito, Glauce Socorro de Barros Viana
    2023, 18 (12):  2611-2614.  doi: 10.4103/1673-5374.373673
    Abstract ( 119 )   PDF (1044KB) ( 86 )   Save
    This critical review of the literature shows that there is a close link between the microbiome, the gut, and the brain in Parkinson’s disease. The vagus nerve, the main component of the parasympathetic nervous system, is involved in the regulation of immune response, digestion, heart rate, and control of mood. It can detect microbiota metabolites through its afferents, transferring this gut information to the central nervous system. Preclinical and clinical studies have shown the important role played by the gut microbiome and gut-related factors in disease development and progression, as well as treatment responses. These findings suggest that the gut microbiome may be a valuable target for new therapeutic strategies for Parkinson’s disease. More studies are needed to better understand the underlying biology and how this axis can be modulated for the patient’s benefit.
    Related Articles | Metrics
    Animal models of vascularized nerve grafts: a systematic review
    Francesca Toia, Daniele Matta, Federico De Michele, Roberto Pirrello, Adriana Cordova
    2023, 18 (12):  2615-2618.  doi: 10.4103/1673-5374.358604
    Abstract ( 93 )   PDF (878KB) ( 41 )   Save
    The aim of this review is to present and compare the various animal models of vascularized nerve grafts described in the literature as well as to summarize preclinical evidence for superior functional results compared to non-vascularized free nerve grafts. We also will present the state of the art on prefabricated vascularized nerve grafts. A systematic literature review on vascularized nerve graft models was conducted via the retrieval with the PubMed database on March 30, 2019. Data on the animal, nerve, and vascularization model, the recipient bed, the evaluation time points and methods, and the results of the study results were extracted and analyzed from selected articles. The rat sciatic nerve was the most popular model for vascularized nerve grafts, followed by the rabbit; however, rabbit models allow for longer nerve grafts, which are suitable for translational evaluation, and produced more cautious results on the superiority of vascularized nerve grafts. Compared to free nerve grafts, vascularized nerve grafts have better early but similar long-term results, especially in an avascular bed. There are few studies on avascular receiving beds and prefabricated nerve grafts. The clinical translation potential of available animal models is limited, and current experimental knowledge cannot fully support that the differences between vascularized nerve grafts and free nerve grafts yield a clinical advantage that justifies the complexity of the procedure. 
    Related Articles | Metrics
    Retinoprotective compounds, current efficacy, and future prospective
    Rachele Marino, Rebecca Sappington, Marco Feligioni
    2023, 18 (12):  2619-2622.  doi: 10.4103/1673-5374.373662
    Abstract ( 122 )   PDF (832KB) ( 57 )   Save
    Retinal dysfunction is the most common cause of vision loss in several retinal disorders. It has been estimated a great increase in these pathologies that are becoming more globally widespread and numerous over time, also supported by the life expectancy increment. Among different types of retinopathies, we can account some that share causes, symptoms, and treatment including diabetic retinopathy, age-related macular degeneration, glaucoma, and retinitis pigmentosa. Molecular changes, environmental factors, and genetic predisposition might be some of the main causes that drive retinal tissue to chronic inflammation and neurodegeneration in these retinopathies. The treatments available on the market contain compounds that efficiently ameliorate some of the important clinical features of these pathologies like stabilization of the intraocular pressure, reduction of eye inflammation, control of eye oxidative stress which are considered the major molecular mechanisms related to retinal dysfunction. Indeed, the most commonly used drugs are anti-inflammatories, such as corticosteroids, antioxidant, hypotonic molecules and natural neuroprotective compounds. Unfortunately, these drugs, which are fundamental to treating disease symptoms, are not capable to cure the pathologies and so they are not life-changing for patients. This review provided an overview of current treatments on the market, but more interestingly, wants to be a quick window on the new treatments that are now in clinical trials. Additionally, it has been here highlighted that the recent technical enhancement of the investigation methods to identify the various retinopathies causes might be used as a sort of “precise medicine” approach to tailor the identification of molecular pathways involved and potentially study a dedicated treatment for each patient. This approach includes the use of cutting-edge technologies like gene therapy and metabolomics.
    Related Articles | Metrics
    Potential role of Lycium barbarum polysaccharides in glaucoma management: evidence from preclinical in vivo studies
    Yamunadevi Lakshmanan, Francisca Siu Yin Wong, Kwok-Fai So, Henry Ho-Lung Chan
    2023, 18 (12):  2623-2632.  doi: 10.4103/1673-5374.355977
    Abstract ( 130 )   PDF (578KB) ( 108 )   Save
    In recent years, the pharmacological benefits of herbal extracts have been revisited for their potential neuroprotective effects in glaucoma. The polysaccharides extracted from the fruits of Lycium barbarum L., or Lycium barbarum polysaccharides, exert their anti-aging effect through reducing oxidative stress, modulating the immune response, enhancing neuronal responses, and promoting cytoprotection. The therapeutic efficacy of Lycium barbarum polysaccharides in preserving retinal ganglion cells and their functions was demonstrated in a range of experimental models of optic neuropathies. These include the acute and chronic ocular hypertension models, the partial optic nerve transection model, and the ischemic-reperfusion injuries model. Based on these findings, Lycium barbarum polysaccharides appear to be a good candidate to be developed as a neuroprotective agent for treating multifactorial diseases. This review aims to present a comprehensive review on the latest preclinical evidence on the pre- and post-treatment benefits of Lycium barbarum polysaccharides in retinal ganglion cell neuroprotection. The possible mechanisms of Lycium barbarum polysaccharides mediating retinal ganglion cell neuroprotection will also be described. Moreover, the potential research gaps in the effective translation of Lycium barbarum polysaccharides treatment into clinical glaucoma management will be discussed. 
    Related Articles | Metrics
    Therapies for Tau-associated neurodegenerative disorders: targeting molecules, synapses, and cells
    Miranda Robbins
    2023, 18 (12):  2633-2637.  doi: 10.4103/1673-5374.373670
    Abstract ( 99 )   PDF (399KB) ( 149 )   Save
    Advances in experimental and computational technologies continue to grow rapidly to provide novel avenues for the treatment of neurodegenerative disorders. Despite this, there remain only a handful of drugs that have shown success in late-stage clinical trials for Tau-associated neurodegenerative disorders. The most commonly prescribed treatments are symptomatic treatments such as cholinesterase inhibitors and N-methyl-D-aspartate receptor blockers that were approved for use in Alzheimer’s disease. As diagnostic screening can detect disorders at earlier time points, the field needs pre-symptomatic treatments that can prevent, or significantly delay the progression of these disorders (Koychev et al., 2019). These approaches may be different from late-stage treatments that may help to ameliorate symptoms and slow progression once symptoms have become more advanced should early diagnostic screening fail. This mini-review will highlight five key avenues of academic and industrial research for identifying therapeutic strategies to treat Tau-associated neurodegenerative disorders. These avenues include investigating (1) the broad class of chemicals termed “small molecules”; (2) adaptive immunity through both passive and active antibody treatments; (3) innate immunity with an emphasis on microglial modulation; (4) synaptic compartments with the view that Tau-associated neurodegenerative disorders are synaptopathies. Although this mini-review will focus on Alzheimer’s disease due to its prevalence, it will also argue the need to target other tauopathies, as through understanding Alzheimer’s disease as a Tau-associated neurodegenerative disorder, we may be able to generalize treatment options. For this reason, added detail linking back specifically to Tau protein as a direct therapeutic target will be added to each topic.
    Related Articles | Metrics
    Molecular chaperones in stroke-induced immunosuppression
    Haoduo Qiao, Qing Xu, Yunfei Xu, Yao Zhao, Nina He, Jie Tang, Jie Zhao, Ying Liu
    2023, 18 (12):  2638-2644.  doi: 10.4103/1673-5374.373678
    Abstract ( 118 )   PDF (530KB) ( 61 )   Save
    Stroke-induced immunosuppression is a process that leads to peripheral suppression of the immune system after a stroke and belongs to the central nervous system injury-induced immunosuppressive syndrome. Stroke-induced immunosuppression leads to increased susceptibility to post-stroke infections, such as urinary tract infections and stroke-associated pneumonia, worsening prognosis. Molecular chaperones are a large class of proteins that are able to maintain proteostasis by directing the folding of nascent polypeptide chains, refolding misfolded proteins, and targeting misfolded proteins for degradation. Various molecular chaperones have been shown to play roles in stroke-induced immunosuppression by modulating the activity of other molecular chaperones, cochaperones, and their associated pathways. This review summarizes the role of molecular chaperones in stroke-induced immunosuppression and discusses new approaches to restore host immune defense after stroke.
    Related Articles | Metrics
    Role of exercise in the brain: focus on oligodendrocytes and remyelination
    Grazia Maugeri, Velia D’Agata, Giuseppe Musumeci
    2023, 18 (12):  2645-2646.  doi: 10.4103/1673-5374.373683
    Abstract ( 118 )   PDF (330KB) ( 39 )   Save
    The phrase “Mens sana in corpore sano” taken from Juvenal’s Satires (127 a. C.) represents one of the best-known and most used sentences of all time, whose meaning in the modern age refers to the importance of physical activity for mental health and wellbeing. Robust literature demonstrated the positive role of exercise in counteracting several diseases, such as diabetes, cardiovascular diseases, neurodegenerative diseases, cancers and age-related disorders including muscle atrophy, the reduction of aerobic capacity, bone and cartilage loss. Moreover, physical exercise ensures brain health and acts as a key player in preventing cognitive decline related to aging. Exercise represents a promising strategy to ensure optimal aging, whose benefits in healthy adults were observed in attention, processing speed, memory, and executive functions. Studies performed on human and animal models showed the ability of physical activity to increase the volume of different brain regions and enhance brain plasticity and neurogenesis through the stimulation of neural progenitor cell proliferation and the sustainment of the developing neurons. Interestingly, the systemic administration of blood plasma derived from exercised mice on aged animals increased the number of newly born neurons in the dentate gyrus region, ameliorating the impaired neurogenesis and cognition in the aged hippocampus (Horowitz et al., 2020). The strict association between physical exercise and neurogenesis was recently confirmed in adult zebrafish. Here, in a spinal cord injury model, the exercise showed to activate the nicotinic-ACh receptors and inhibit the GABAA receptors, by increasing the number of newborn neurons and promoting motor function restoration (Chang et al., 2021). This evidence also corroborates previous results showing the positive role of exercise to enhance motor and sensory functions in spinal cord injury-affected patients as well as axonal regeneration and sprouting in rodent models. The positive relationship between exercise and neurogenesis is also due to the ability of physical activity to increase the expression levels and the secretion of neurotrophic and growth factors, including brain-derived neurotrophic factor, insulin-like growth factor-1, nerve growth factor and vascular endothelial growth factor. Brain-derived neurotrophic factor plays a key role in cognition, neuroplasticity, and angiogenesis. It promotes neural connectivity and is involved in the development of learning and memory. Insulin-like growth factor-1 is a neuroprotective factor, which supports brain development, neural survival and vasculature. Nerve growth factor is considered to play an essential role in mediating neuronal development and survival, and its treatment inverts the effects of lesions and age-related degeneration. Exercise also stimulates the release of vascular endothelial growth factor, which through angiogenesis also directly enhances neurogenesis and synaptic function.
    Related Articles | Metrics
    GABAergic synaptic transmission and plasticity oscillate across sleep and wake
    Kunwei Wu, Wei Lu
    2023, 18 (12):  2647-2648.  doi: 10.4103/1673-5374.373665
    Abstract ( 119 )   PDF (374KB) ( 139 )   Save
    Sleep is a widely expressed behavior across the animal kingdom. In addition to the numerous health benefits that are associated with sleep, it is believed that sleep plays a pivotal role in mental processes such as learning and memory. Indeed, it has been demonstrated that learning and memory benefit from sleep, whereas sleep loss causes cognitive impairment (Rasch and Born, 2013). Changing the strength of synapses, the connections between neurons, has been proposed to be the basic memory mechanism. Therefore, it is not surprising that many studies about the memory functions of sleep have focused on its effects on synapses. An influential hypothesis regarding the function of sleep in learning and memory is the synaptic homeostasis hypothesis, which proposes that wake increases overall excitatory synaptic strength due to ongoing learning and sleep renormalizes them to facilitate memory consolidation and integration (Tononi and Cirelli, 2014). This view has received support by findings in a number of studies showing molecular, morphological and electrophysiological changes indicative of excitatory synaptic weakening during sleep and strengthening during wake (Tononi and Cirelli, 2014). However, several recent studies have shown that sleep can potentiate excitatory synaptic transmission (Chauvette et al., 2012), or have no impact on excitatory synaptic strength (Cary and Turrigiano, 2021). These studies indicate complex mechanisms underlying the possible roles of sleep in regulating excitatory synapses. Of note, given a neuron typically receives thousands of synaptic inputs and the interactions between excitatory and inhibitory synaptic inputs determine the level of activity in it, neuronal communication across sleep and wake is probably regulated as a balance between excitatory and inhibitory influences. However, compared to extensive studies on excitatory synapses, much less is known about the regulation of inhibitory synapses by sleep. 
    Related Articles | Metrics
    Implications of Olig2 silencing in oligodendrocyte precursor cells
    Li-Pao Fang, Xianshu Bai
    2023, 18 (12):  2649-2650.  doi: 10.4103/1673-5374.373666
    Abstract ( 173 )   PDF (516KB) ( 79 )   Save
    Oligodendrocytes (OLs) are the only myelin-forming cells in the central nervous system. Their differentiation from OL precursor cells (OPCs) occurs throughout life and is mediated by numerous intrinsic and extrinsic factors. OL transcription factor 2 (Olig2), a basic helix-loop-helix transcription factor, is one of the intrinsic factors that specify the OL lineage. It is expressed by both OPCs and OLs, and no variant of Olig2 has yet been identified in rodents. Although the function of Olig2 in OL maturation and myelination is still under debate, Olig2 is essential for OPC differentiation in health and disease. Because of its broad expression throughout the OL lineage, Olig2 is often used as a lineage marker. However, in the healthy perinatal and adult brain, a small population of NG2-positive (NG2pos) cells were found to be Olig2-negative (Olig2neg), and stab wound injury increased the population of NG2posOlig2neg cells. NG2 is a protein specifically expressed by OPCs and pericytes in the healthy brain and additionally by microglia after acute brain injury. Therefore, it remained unclear whether these NG2posOlig2neg cells are OPCs or other cell types, such as pericytes or microglia? If these cells are OPCs, are they functionally different from the Olig2pos OPCs? By immunostaining for platelet-derived growth factor receptor alpha (PDGFRα), the established marker of OPCs, we confirmed that a subset of OPCs does indeed not express Olig2. This population of OPCs could be detected throughout life, from the embryonic stage (embryonic day 14.5) to the aged mouse (44 weeks old). Fate mapping studies provided strong evidence that Olig2neg OPCs are derived from pre-existing Olig2pos OPCs (Fang et al., 2023). Therefore, it is conceivable that Olig2neg OPCs do not represent a separate cell type, but rather a distinct functional stage of OPCs in which Olig2 expression is transiently downregulated in response to microenvironmental changes. In other words, OPCs may dynamically up- and downregulate Olig2 expression in response to changes in brain activity.
    Related Articles | Metrics
    Focus on LPA signaling: a promising therapeutic target to foster regeneration in immune-mediated neuropathies
    Fabian Szepanowski, Mark Stettner
    2023, 18 (12):  2651-2652.  doi: 10.4103/1673-5374.373709
    Abstract ( 111 )   PDF (325KB) ( 46 )   Save
    Lysophospholipids are metabolites of glycerophospholipids and sphingolipids that are commonly found as lipid constituents of cell membranes. In addition to their function as structural membrane components, certain members of the lysophospholipid family have considerable cell signaling properties. Sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) represent the two major bioactive members of the lysophospholipid family that address specific G-protein coupled receptors, simply referred to as S1P (subtypes S1P1–5) and LPA (subtypes LPA1–6) receptors (Kano et al., 2022). Lysophospholipid signaling has a wide range of physiological functions and potential pathophysiological effects in the adult organism, but also plays important roles during embryonic development, especially in the development of the nervous, vascular and immune system (Kano et al., 2022).
    Related Articles | Metrics
    The presence of functional blood vessels in the ischemic core provides a therapeutic target for stroke recovery
    Gary P. Morris, Brad A. Sutherland
    2023, 18 (12):  2653-2654.  doi: 10.4103/1673-5374.373703
    Abstract ( 120 )   PDF (638KB) ( 41 )   Save
    Ischemic stroke occurs when a blood vessel in the brain is occluded. In the immediate aftermath, blood flow becomes deficient in regions supplied by the blocked vessel. This leads to the development of a core region, where blood flow is reduced to < 30% and injury occurs rapidly, and a penumbral region, where blood flow is reduced to a level that is not capable of supporting neuronal function, but is sufficient to maintain cell viability (Morris et al., 2023). In the core, tissue injury occurs within minutes and this region becomes walled off from the surrounding tissue through the formation of a glial scar (Morris et al., 2023). Ultimately, the core becomes fibrotic, devoid of neurons, with a limited vascular supply (Kanazawa et al., 2019). In the penumbra, residual blood flow can maintain tissue homeostasis for several hours, but if blood flow is not restored, irreversible cell death also occurs in this region. 
    Related Articles | Metrics
    Unexpected role of complement component 8 gamma chain in the inflamed brain
    Jong-Heon Kim, Kyoungho Suk
    2023, 18 (12):  2655-2656.  doi: 10.4103/1673-5374.373706
    Abstract ( 100 )   PDF (410KB) ( 38 )   Save
    Neuroinflammation, an intricate inflammatory process occurring in the central nervous system (CNS), plays an important role in host defense. Glial cells, including astrocytes and microglia, along with cytokines, chemokines, and the complement system are important components of neuroinflammation. A low level of neuroinflammation is associated with and contributes to various homeostatic and neuroprotective processes, such as removing pathogens or cellular debris and promoting tissue repair after brain injury. However, prolonged or maladaptive neuroinflammation has been implicated in neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease, multiple sclerosis, and major depression. Increasing evidence indicates that targeting neuroinflammation may be a potential therapeutic intervention against neurodegeneration for delaying disease onset or progression. However, our current understanding of neuroinflammation remains limited.
    Related Articles | Metrics
    ‘Hippocampal innate inflammatory gliosis only’ – the future role of surgery in a novel temporal lobe epilepsy syndrome
    Alexander Grote, Daniel Delev
    2023, 18 (12):  2657-2658.  doi: 10.4103/1673-5374.373707
    Abstract ( 94 )   PDF (576KB) ( 48 )   Save
    Epilepsy is one of the most common neurological conditions affecting more than 50 million people worldwide (https://www.who.int/news-room/fact-sheets/detail/epilepsy). Despite numerous antiseizure medications (ASM), approximately 30% of all patients will develop drug-resistant epilepsy (DRE). DRE leads to devastating health and socio-economic consequences (Luoni et al., 2011).
    Related Articles | Metrics
    Restoration of GABAB receptor expression in cerebral ischemia: a promising novel neuroprotective strategy
    Musadiq A. Bhat, Mohammad Hleihil, Dietmar Benke
    2023, 18 (12):  2659-2600.  doi: 10.4103/1673-5374.373704
    Abstract ( 144 )   PDF (525KB) ( 49 )   Save
    Although stroke is a major global health problem, a pharmacological treatment to inhibit ongoing neuronal death in patients is still lacking. In cerebral ischemia, the prevailing form of stroke, severely reduced blood supply by obstruction of blood vessels deprives neurons from oxygen and glucose, eventually leading to metabolic derailment and death of neurons in the affected brain area. Currently, the only available treatment for acute ischemic stroke is thrombolytic drugs (Alteplase, Tenecteplase) to break down blood clots to restore blood circulation and thrombectomy to mechanically remove the occlusion. Even after the successful restoration of blood flow, progressive neuronal death remains a major concern. One of the main causes for ongoing neuronal death is excitotoxicity induced by excessive glutamate release and overactivation of glutamate receptors resulting in chronic overexcitation of neurons (Choi, 2020). Under physiological conditions, neuronal excitability and elevated glutamate release are controlled by the heterodimeric G protein-coupled γ-aminobutyric acid type B (GABAB) receptors. However, under ischemic conditions, GABAB receptors are downregulated and can no longer counteract the overexcitation of neurons. Therefore, restoring GABAB receptor expression to normal levels after an ischemic insult is a promising strategy to limit overexcitation and the associated excitotoxic death of neurons. We recently showed that targeting protein-protein interactions involved in the aberrant downregulation of GABAB receptors with specific interfering peptides restored GABAB receptor expression, counteracted neuronal overexcitation, and provided neuroprotection with a potentially wide time window (Balakrishnan et al., 2022; Bhat et al., 2022; Hleihil et al., 2022). Besides cerebral ischemia, this approach could also be a promising strategy to address other neurological diseases associated with an excitation/inhibition imbalance and downregulation of GABAB receptors, e.g. addiction and neurodegenerative diseases.
    Related Articles | Metrics
    Advancing spinal cord injury research with optical clearing, light sheet microscopy, and artificial intelligence-based image analysis
    Qiang Li, Alfredo Sandoval Jr, Bo Chen
    2023, 18 (12):  2661-2662.  doi: 10.4103/1673-5374.373708
    Abstract ( 107 )   PDF (481KB) ( 33 )   Save
    From the days of Ramon y Cajal’s first sketches, neuroscientists have recognized the importance of visualizing the complex architecture of the central nervous system. In the past century, we have come to appreciate how the rich structural and functional complementarity of axons and cell types in the spinal cord make it uniquely suited for information transfer between the periphery and the brain. However, appreciating these relationships has been limited by the low-throughput histology techniques in common usage. For example, axon projections span many spinal segments, traversing centimeters to meters (depending on the species), with many spinal nuclei also occupying more than one spinal level. Spinal cord injuries also disrupt many levels of the spinal cord, with secondary inflammatory responses reaching far beyond the epicenter and surrounding penumbra after injury. These injuries affect the primary injury site, the penumbra, and descending and ascending white matter tracts that pass through the injury site, ultimately impacting communication between the periphery, the spinal cord, and the supraspinal structures. Therefore, holistically examining spinal structures or injuries that span multiple segments with two-dimensional (2D) sections is extremely challenging. At best, 2D sections can give a snapshot of axons and neighboring cells, however, they provide no information about axon trajectory and only offer variable representations of cells in any given cross-section. Additionally, these approaches are labor-intensive and can only produce short reconstructions of projection axons in sparsely labeled samples due to the optical limitations associated with imaging thick tissue sections. These drawbacks may severely limit the novelty, scope, and impact of studies, as useful data is left uncollected due to the biased nature and technical limitations of traditional 2D histology and imaging techniques. In light of these disadvantages and the difficulty in acquiring samples from complex SCI experiments, tools that can effectively resolve structures across multiple scales, from the tissue to the subcellular level, while preserving spatial relationships, are highly needed. 
    Related Articles | Metrics
    Recent advances in RNA-targeting therapy for neurological diseases
    Satheesh Kumar, Guei-Sheung Liu
    2023, 18 (12):  2663-2664.  doi: 10.4103/1673-5374.373658
    Abstract ( 126 )   PDF (574KB) ( 75 )   Save
    Advances in sequencing and molecular technology now allow us to understand the genetic underpinnings of complex diseases such as neurological disorders. Genetic variations (or mutations) in the DNA sequence of single genes have been implicated in neurological diseases such as Huntington’s disease and spinal muscular atrophy. As a result, the development of gene therapies for neurological diseases is now a feasible endeavor. Indeed, gene therapy for neurological diseases has recently been invigorated by the market approvals of Zolgensma® (onasemnogene abeparvovec) in 2019 and UpstazaTM (eladocagene exuparvovec) in 2022. These gene therapies deliver a transgene to compensate for an aberrant or missing gene for the therapeutic benefit of neurological diseases and have demonstrated significant clinical potential. However, current gene therapy is limited to loss-of-function genetic diseases, and the delivery approach for large genes has been a challenge.
    Related Articles | Metrics
    Axon guidance in regeneration of the mature central nervous system: step by step
    Julia Schaeffer, Noemie Vilallongue, Stephane Belin, Homaira Nawabi
    2023, 18 (12):  2665-2666.  doi: 10.4103/1673-5374.373663
    Abstract ( 133 )   PDF (383KB) ( 59 )   Save
    Unlocking axon regeneration in the injured central nervous system: In adult mammals, central nervous system (CNS) neurons fail to regenerate after a lesion, whether it is traumatic – after spinal cord injury for example – or in the case of neurodegenerative diseases. This causes axons to degenerate and neurons to die, leading to permanent motor and/or cognitive impairment. One of the reasons behind this regeneration failure lies in the mature CNS environment, where a number of growth-inhibitory factors, at the lesion site, contributes to axon regrowth inhibition (He and Jin, 2016). In this context, removing such extrinsic factors should alleviate the growth-inhibitory barrier. Yet, surprisingly, no robust regeneration is achieved past the lesion site. These results led researchers to investigate the intrinsic regrowth properties of adult neurons themselves. Indeed, adult CNS neurons lose their capacity to grow an axon, not only because of the switch-off of developmental pro-growth programs during maturation, but also in response to the injury itself (Belin et al., 2015; He and Jin, 2016).
    Related Articles | Metrics
    Exploration of C-terminal CX3CL1 for reducing age-dependent neurodegeneration
    Jacob Hudobenko, Manoshi Gayen, Marc R. Benoit, Neeraj Singh, Riqiang Yan
    2023, 18 (12):  2667-2668.  doi: 10.4103/1673-5374.373702
    Abstract ( 106 )   PDF (577KB) ( 34 )   Save
    Owing to the increasing life expectancy, the prevalence of age-dependent neurodegenerative diseases will exert an enormous toll on our aging population, their caregivers and the healthcare system. It is anticipated that numbers of patients suffering from diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and other neurodegenerative diseases are poised to increase markedly over the next few decades. Therefore, discovering treatments that mitigate the deleterious effects of these diseases is imperative. The common feature of neurodegenerative diseases is the loss of neurons, due to the accumulation of abnormal protein aggregates, which eventually cause dysfunction and death of neurons (Wareham et al., 2022). Stem cell niches are present in the adult brain, but the majority of neurogenesis takes place before adulthood, meaning that the terminally differentiated neurons, lost to neurodegenerative diseases, are unlikely to be replaced under normal physiological conditions. The loss of functional neurons will lead to permanent disability for patients. To date, the drug discovery effort for treating individual neurodegenerative diseases including AD, PD, and Huntington’s disease (HD) has been enormous, but the therapeutic strategy for preventing neuron loss in multiple neurodegenerative diseases is scarce.  In this perspective, we will discuss a recently discovered small peptide (Fan et al., 2019), named CX3CL1-ICD, which is derived from the intracellular domain of CX3CL1, for its translational potential through both enhancing neurogenesis and reducing neuronal apoptosis in neurodegenerative diseases. 
    Related Articles | Metrics
    Calcium-permeable AMPA receptors: an underestimated pharmacological target for the therapy of brain pathologies
    Sergei G. Gaidin, Artem M. Kosenkov
    2023, 18 (12):  2669-2670.  doi: 10.4103/1673-5374.373714
    Abstract ( 127 )   PDF (576KB) ( 49 )   Save
    Excitotoxicity resulting from the accumulation of extracellular glutamate (the main excitatory neurotransmitter in the brain) is one of the main causes of neuronal death in various brain pathologies, including traumatic brain injury, epilepsy, stroke, and a number of neurodegenerative diseases such as Parkinson’s and Alzheimer’s diseases. An acute increase in extracellular glutamate concentration leads to neuronal dysfunction accompanied by oxidative stress, calcium homeostasis failure, a drop in energy metabolism, and loss of plasma membrane integrity. However, despite a more than 30-year history of studying glutamate excitotoxicity, there are no safe and effective drugs that prevent cell death under these pathological conditions. Numerous promising neuroprotective compounds have been rejected in clinical trials due to low efficacy and/or side effects. N-methyl-D-aspartate receptors (NMDARs) are considered the main source of Ca2+ influx during excitotoxicity, so various NMDAR antagonists have been primarily tested. However, the paradigm of NMDAR-Ca2+-mediated excitotoxicity formulated in the 1990s has outlived itself in terms of pharmacological potential. In this regard, the researchers have focused on α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), the antagonists of which also demonstrate neuroprotective effects in in vitro studies. 
    Related Articles | Metrics
    Historic recurrences in medicinal chemistry: nature-inspired structures as a new opportunity for novel multi-target anti-Alzheimer’s drugs
    Luca Piemontese
    2023, 18 (12):  2671-2672.  doi: 10.4103/1673-5374.373685
    Abstract ( 87 )   PDF (251KB) ( 24 )   Save
    Alzheimer’s disease (AD) is an alarming non-communicable, multi-factorial, and non-treatable disease. Its underlying neurodegenerative events have not yet been fully explained and its early diagnosis is very difficult. The appearance of the disease is associated with clinical features such as the degeneration of several cholinergic nuclei of the brain, causing lower levels of the neurotransmitter acetylcholine and the formation of protein aggregates in the inter-synaptic space (amyloid plaques) or inside the cells (neurofibrillary tangles, Brunetti et al., 2020). 
    Related Articles | Metrics
    CD74: a prospective marker for reactive microglia?
    Phani Sankar Potru, Björn Spittau
    2023, 18 (12):  2673-2674.  doi: 10.4103/1673-5374.371350
    Abstract ( 294 )   PDF (541KB) ( 139 )   Save
    Cluster of differentiation 74 (CD74), also called as major histocompatibility complex class II (MHCII) invariant chain, is involved in trafficking MHCII cell surface molecules on antigen-presenting cells and has been implicated in many signaling pathways. For example, the interaction between CD74 and macrophage migration inhibitory factor cytokine (MIF) leads to the activation of a plethora of pathways such as extracellular regulated protein kinases, phosphoinositide 3-kinase, and nuclear factor-κB which are essential for cell survival, differentiation, and proliferation. Structurally, CD74 is a type 2 transmembrane receptor with a short N-terminal cytoplasmic tail consisting of 28 amino acids, a 24-amino acid transmembrane region, and a luminal domain of approximately 150 amino acids. In mice, Cd74 occurs in two isoforms, namely p31, and p41, due to alternative splicing. In humans, along with the p33 and p41 isoforms that correspond to the p31 and p41 isoforms of mice, two additional isoforms p35 and p43 can also be found (Farr et al., 2020). Microglia are central nervous system (CNS)-specific antigen-presenting cells with diverse functional repertoire. Microglia remain in a resting, albeit a surveying state under normal physiological conditions. Such microglia are deemed to be in their homeostatic state with a characteristic expression of Tmem119, Olfml3, P2ry12, Sall1, Hexb, Gpr34, or Fcrls. They however become reactive under pathological states assuming phenotypes with increased expression of genes such as ApoE, Axl, Clec7a, Cst7, Cybb, and Ctsb. It is now widely accepted that this microglial activation and the resultant dysregulation is an inevitable component of almost all CNS pathologies (Pettas et al., 2022).
    Related Articles | Metrics
    Cyclic adenosine monophosphate-elevating agents inhibit amyloid-beta internalization and neurotoxicity: their action in Alzheimer’s disease prevention
    Rayudu Gopalakrishna, Andrew Oh, Narayan R. Bhat, William J. Mack
    2023, 18 (12):  2675-2676.  doi: 10.4103/1673-5374.373664
    Abstract ( 107 )   PDF (401KB) ( 59 )   Save
    Recently, we have found that various intracellular cyclic adenosine monophosphate (cAMP)-elevating agents, both pharmacological (dibutyryl-cAMP, forskolin, and rolipram) and physiological (pituitary adenylate cyclase-activating polypeptide), decrease cell-surface levels of 67-kDa laminin receptor (67LR) and cellular prion protein (PrPC). Thereby, they inhibit the internalization of amyloid-β oligomer (AβO) and attenuate AβO-induced neuronal death (Figure 1; Gopalakrishna et al., 2022). We postulate that the 67LR-PrPC-mediated AβO mechanism may be important in understanding the Alzheimer’s disease (AD)-preventive actions of green tea polyphenol, epigallocatechin-3-gallate (EGCG), which is known to bind 67LR at a site within the PrPC-binding site and induce 67LR internalization. This mechanism may also be relevant in understanding the anti-AD actions of dietary agents such as resveratrol and quercetin as well as synthetic drugs (including the ones in clinical trials) that elevate intracellular cAMP by inhibiting cyclic nucleotide phosphodiesterases (PDEs).   
    Related Articles | Metrics
    Newfound physiological function of FAIM protein offers hope of novel disease-modifying therapy for Alzheimer’s disease
    Allison R. Balaj, Hiroaki Kaku
    2023, 18 (12):  2677-2679.  doi: 10.4103/1673-5374.374003
    Abstract ( 127 )   PDF (971KB) ( 63 )   Save
    Introduction of Fas apoptosis inhibitory molecule (FAIM): FAIM was originally discovered in FAS-resistant mouse primary B lymphocytes in 1999, and was thought of as a FAS-apoptosis inhibitor based on overexpression studies (Schneider et al., 1999). FAIM is an approximately 20 kDa intracellular protein, but a subsequent study identified an alternatively spliced form, termed FAIM-Long (L), which has 22 additional amino acids at the N-terminus (Zhong et al., 2001). Thus, the originally identified FAIM was renamed FAIM-Short (S). FAIM is also termed FAIM1 in some publicly available gene/genome databases. Although two other gene products were confusingly termed FAIM2 (also termed lifeguard) and FAIM3 (also termed TOSO), neither FAIM2 nor FAIM3 are related to FAIM-S and FAIM-L in terms of physiological functions or gene/protein homology. FAIM-S is ubiquitously expressed in the body, but FAIM-L is expressed almost exclusively in the brain and testis (Zhong et al., 2001). Overexpression studies using B lymphocytes showed that FAIM inhibits FAS-mediated apoptosis presumably by enhancing NF-κB activation (Schneider et al., 1999; Kaku and Rothstein, 2009a, b). However, the physiological role of FAIM (hereafter FAIM indicates both FAIM-S and FAIM-L) was unknown for many years, partly because no abnormality was detected in FAIM-deficient mice. It was not until our recent studies that we discovered FAIM-deficient cells are more susceptible to cellular stress (Kaku and Rothstein, 2020). In retrospect, its role might have previously been obscured by the lack of stress in vivarium mouse life. 
    Related Articles | Metrics
    TREM2 as an evolving therapeutic target in Alzheimer’s disease
    Jacob George
    2023, 18 (12):  2680-2681.  doi: 10.4103/1673-5374.371360
    Abstract ( 87 )   PDF (253KB) ( 41 )   Save
    Related Articles | Metrics
    Trans-synaptic degeneration as a mechanism of neurodegeneration in multiple sclerosis
    Olwen C. Murphy, Peter A. Calabresi, Shiv Saidha
    2023, 18 (12):  2682-2684.  doi: 10.4103/1673-5374.373661
    Abstract ( 106 )   PDF (951KB) ( 58 )   Save
    Multiple sclerosis (MS) is regarded as an immune-mediated, demyelinating disorder of the central nervous system, however neuroaxonal degeneration is accepted as the principal substrate of disability accumulation (Dutta and Trapp, 2011). Neurodegeneration occurs throughout the course of MS and is detectable even in the earliest stages of the disease (Azevedo et al., 2018). Mechanisms of neurodegeneration in MS are complex and not completely understood. Neuro-axonal transection or degeneration can occur within acutely or chronically demyelinated MS lesions, and also within normal-appearing white and gray matter (Dutta and Trapp, 2011). At a tissue level, pathologic contributors to neuro-axonal degeneration may include inflammatory injury, loss of trophic support, retrograde and anterograde degeneration, failure of remyelination, impaired axonal transport, microglial activation, mitochondrial injury, energy failure, oxidative injury, iron accumulation and tissue hypoxia (Dutta and Trapp, 2011; Mahad et al., 2015). One putative mechanism of neurodegeneration is trans-synaptic degeneration – whereby injury to a neuron or axon leads to the degeneration of synaptically-connected neurons. Theoretically, trans-synaptic degeneration may proceed anterogradely (“dying forward”) or retrogradely (“dying back”), ultimately resulting in the loss of neurons in discrete but distant central nervous system locations (Figures 1 and 2). Based on this mechanism, trans-synaptic degeneration could occur in MS both in demyelinated tissue and in normal-appearing tissue. However, while trans-synaptic degeneration is biologically plausible in MS, and supported by indirect evidence (Rocca et al., 2013; Gabilondo et al., 2014), the “real time” documentation of trans-synaptic degeneration has been challenging. Pathological studies of MS are typically not suitable for assessing trans-synaptic changes, since post-mortem studies can only assess a single pathological timepoint, and post-mortem samples of MS brains are usually acquired from older patients – often years after the phase of the disease involving overt inflammatory activity. Furthermore, radiological capturing of trans-synaptic degeneration in vivo has been a major challenge due to the limitations of imaging technology and the complex synaptically-connected central nervous system networks which can be affected by many MS lesions.
    Related Articles | Metrics
    Calcium handling: a strategy to fight neurodegeneration in Alzheimer’s disease
    Livia La Barbera, Elena Spoleti, Marcello D’Amelio
    2023, 18 (12):  2685-2686.  doi: 10.4103/1673-5374.374004
    Abstract ( 72 )   PDF (845KB) ( 62 )   Save
    In the last few years, preclinical and clinical studies identified the ventral tegmental area (VTA) as one of the first brain regions to be affected in the prodromal phase of Alzheimer’s disease (AD). 
    The VTA is a deep midbrain nucleus rich in dopaminergic neurons that innervates and releases dopamine (DA) in several cortical and subcortical brain regions. DA plays a crucial role in the modulation of both cognitive and non-cognitive functions and failure of its release underlies both cognitive and neuropsychiatric symptoms in patients with AD. 
    Related Articles | Metrics
    Meningeal T-cells in the cross-talk between memory consolidation and sleep
    Rodrigo Pacheco, Valentina Ugalde
    2023, 18 (12):  2687-2688.  doi: 10.4103/1673-5374.373686
    Abstract ( 78 )   PDF (641KB) ( 34 )   Save
    Some years ago, the infiltration of T-cells into the central nervous system (CNS) was considered a pathological condition exclusively. However, during the last two decades, a growing body of studies has shown that T-cells are essential players regulating several physiological processes exerted by the CNS, including stress resilience, social behavior, anxiety, learning, and memory. Notably, the discovery of the meningeal lymphatics (Louveau et al., 2015) provided critical anatomical clues to a better understanding of the mechanisms by which T-cells collaborate with the CNS in cognitive and psychological processes. For instance, a number of mouse strains devoid of functional T-cells present worst adaptation to stress than immunocompetent counterparts, and the adoptive transfer of T-cells isolated from stress-experienced mice improves the psychological ability to adapt to stress in non-experienced mice appropriately (Brachman et al., 2015). Moreover, it has been demonstrated that T-cells deficiency results in anti-social behavior. The mechanistic analysis provided evidence indicating that interferon-gamma produced by meningeal T-cells stimulates GABAergic neurons, triggering inhibitory circuits that prevent hyperexcitability in the prefrontal cortex, thus promoting social behavior (Filiano et al., 2016). In addition, it has been shown that, through the production of interleukin (IL)-17, meningeal γδT-cells might stimulate cortical glutamatergic neurons inducing anxiety-like behavior in mice (Alves de Lima et al., 2020). Furthermore, another set of studies has shown that T-cells, particularly those CD4+ T-cells producing IL-4 and IL-13, are required for the proper learning and acquisition of memory (Derecki et al., 2010; Brombacher et al., 2017).
    Related Articles | Metrics
    The tip of the iceberg? The underestimated potential of non-canonical beta-amyloids for Alzheimer’s disease
    Lukas Busch, Bernd Bufe
    2023, 18 (12):  2689-2690.  doi: 10.4103/1673-5374.374013
    Abstract ( 111 )   PDF (1111KB) ( 88 )   Save
    Formation and deposition of amyloid-beta (Aβ) are considered one of the main drivers of Alzheimer’s disease (AD). For more than 30 years, Aβ has challenged researchers through its complex physicochemical properties and multiple peptide processing steps that involve several proteases (Andreasson et al., 2007), ultimately creating many Aβ variants that trigger various physiological effects in neurons, glia, and peripheral immune cells (Busch et al., 2022a). Recent research identified various additional naturally occurring Aβ species with different lengths and chemical modifications. Their impact on AD pathology is currently not well understood, although some of them occur at much higher abundance than Aβ1–42 and Aβ1–40. Here, we highlight new findings that challenge traditional views and summarize recent findings that argue for a significant contribution of previously neglected Aβ variants to the development and progression of AD. Next, we provide an overview on the technical challenges and open questions associated with their study and highlight how different Aβ variants such as Aβ11–40 and Aβ17–40 may use formyl peptide receptor signaling to trigger distinct cellular effects in neurons, glia, and immune cells. 
    Related Articles | Metrics
    Implication of salivary lactoferrin and periodontal-mediated infections in Alzheimer’s disease
    Cristina Municio, Eva Carro
    2023, 18 (12):  2691-2692.  doi: 10.4103/1673-5374.373712
    Abstract ( 128 )   PDF (455KB) ( 34 )   Save
    Lactoferrin is an antimicrobial protein characterized by the exertion of many protective functions, including antibacterial, antifungal, antiviral, and antiparasitic properties, as well as anti-inflammatory and immunomodulatory activities (Kruzel et al., 2017). Lactoferrin is one of the major proteins present in exocrine secretions, including saliva, and is therefore associated with host defense against oral pathogens and control of the oral microbiome. In recent years, it has become clear that alterations in the oral microbiome may contribute to opportunistic pathogen infections in the brains of Alzheimer’s disease (AD) patients and thus participate in or contribute to the development of this neurodegenerative disease (Sureda et al., 2020). Pathogenic oral microbes can affect neurological processes by entering brain tissue through various pathways and directly damaging the central nervous system. In the central nervous system, oral microbes may trigger an immune response that increases amyloid β (Aβ) production and may even trigger the Aβ cascade to promote the onset of AD, as we discuss in our previous study supporting the “infectious hypothesis” in AD (González-Sánchez et al., 2020).
    Related Articles | Metrics
    Cell replacement for Parkinson’s disease: advances and challenges
    Bin Xiao, Eng-King Tan
    2023, 18 (12):  2693-2694.  doi: 10.4103/1673-5374.373710
    Abstract ( 168 )   PDF (402KB) ( 71 )   Save
    Parkinson’s disease (PD), characterized by the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain, is a prototype neurological disease that is suitable for cellular replacement therapy. Levodopa has been utilized to replace the insufficient dopamine released by degenerating DA neurons since the 1960s and it remains the cornerstone of PD treatment. However, as the disease progresses, the diminishing DA neurons become inadequate to convert administered levodopa to functional dopamine, and the affected axonal projections fail to deliver dopamine to target brain regions. As a result, the dopamine replacement eventually loses its efficacy after the initial honeymoon period, and chronic levodopa therapy is associated with debilitating side effects, including motor and non-motor complications.  
    Related Articles | Metrics
    On the role of RNA binding proteins in polyglutamine diseases: from pathogenesis to therapeutics
    André Conceição, Rebekah Koppenol, Clévio Nóbrega
    2023, 18 (12):  2695-2696.  doi: 10.4103/1673-5374.373711
    Abstract ( 84 )   PDF (743KB) ( 62 )   Save
    Polyglutamine (polyQ) diseases are a group of different neurodegenerative disorders characterized by an abnormal expansion of the trinucleotide cytosine-adenine-guanine (CAG) within coding regions of each disease-associated gene. The abnormal expansion translates into a protein bearing an abnormally long tract of glutamines. The expanded proteins are prone to aggregate, promote aberrant interaction with other proteins and mRNAs and contribute to cellular pathway disruption (Matos et al., 2019). To date, nine different polyQ diseases are described, including among others, Huntington’s disease, and six different spinocerebellar ataxias (SCA). Patients affected by polyQ diseases, suffer a myriad of motor symptoms that include ataxia, dysphagia, tremors, dysarthria, and even dementia. Unfortunately, there is no cure nor treatment able to delay the disease and patients rely only on symptomatic and supportive treatments culminating in premature death (Takahashi et al., 2010).
    Related Articles | Metrics
    Impaired lysosomes in the retinal pigment epithelium play a central role in the degeneration of the neuroretina
    Rebecca D. Miller, J. Arjuna Ratnayaka
    2023, 18 (12):  2697-2698.  doi: 10.4103/1673-5374.373713
    Abstract ( 104 )   PDF (738KB) ( 48 )   Save
    Lysosomes are highly dynamic, single membrane-bound compartments that are critical for maintaining cellular homeostasis. These organelles, which appear to vary between 200 nm–1 μm in diameter, originate from the maturation of endocytic vesicles and via the enrichment of newly synthesized lysosomal proteins in the trans-Golgi network. Lysosomes mediate the targeted degradation of intracellular and extracellular cargoes, including those trafficked by the endosomal and autophagic pathways, to produce monosaccharides, amino acids, and free fatty acids, among other molecules. However, recent discoveries have revealed additional lysosomal functions, including roles in nutrient and metabolic sensing, gene regulation, inflammation, membrane repair, and effects on the extracellular environment. Lysosomes are found throughout the cytoplasm, but may have a preferential distribution depending on their specific activity or cell state. For example, due to their origin, lysosomes predominantly localize to the perinuclear region, but also maintain this distribution under conditions of proteolytic stress. However, secretory lysosomes localize to the cell periphery and also appear to be less acidic (Keeling et al., 2018). Lysosomal positioning is also influenced by contact with the endoplasmic reticulum, mitochondria and the Golgi apparatus. Lysosomes are rarely stationary and are trafficked bidirectionally along microtubules, with anterograde and retrograde movements mediated by kinesin or dynein. While lysosomes are known to play a causative role in storage diseases, their broader dysfunction contributes to a variety of neurodegenerative diseases, including Alzheimer’s disease, frontotemporal dementia, age-related macular degeneration (AMD), amyotrophic lateral sclerosis, Parkinson’s disease, and Huntington’s disease, among others (Keeling et al., 2018; Malik et al., 2019). Retinal pigment epithelial (RPE) cells provide a unique model to study lysosomal biology and the consequences of their dysfunction. The RPE forms a highly specialized monolayer in the retina that is intimately associated with photoreceptors. Overlying photoreceptors shed outer segments (photoreceptor outer segments, POS) as part of the daily photoreceptor renewal, which are engulfed and trafficked for degradation in lysosomes of RPE cells. Here, we review our novel findings showing how disease-related pathways in the retina affect these organelles and how lysosomal defects may contribute to RPE damage as part of a clinically well-defined pathway of RPE atrophy leading to irreversible vision loss in AMD and other retinopathies.
    Related Articles | Metrics
    Implication of gut microbiome in age-related macular degeneration
    Wendy Luo, Dimitra Skondra
    2023, 18 (12):  2699-2670.  doi: 10.4103/1673-5374.373687
    Abstract ( 89 )   PDF (696KB) ( 36 )   Save
    Age-related macular degeneration (AMD) is the most common cause of blindness in the United States in adults over 55 years of age and is one of the leading global causes of blindness: at least 196 million of the worldwide population have AMD, and prevalence is projected to rise to 288 million by 2040 (Lin et al., 2021). As cases and disease burden increase, improvements in the characterization of AMD pathobiology and exploration of potential therapeutic solutions are necessary first steps in addressing this global health concern.
    Related Articles | Metrics
    The pathology behind glaucoma: what we already know using omics technologies
    Caroline May, Sabrina Reinehr
    2023, 18 (12):  2701-2702.  doi: 10.4103/1673-5374.373667
    Abstract ( 113 )   PDF (527KB) ( 46 )   Save
    The term glaucoma encompasses a spectrum of eye diseases pathologically characterized by an irreversible loss of retinal ganglion cells and their associated axons. Patients initially suffer from gaps in their visual field, the part of the visual range that can be perceived without eye movement. In the early stages of the disease, this damage can be compensated very well by eye movements. This, in turn, means that glaucoma is often not diagnosed until the disease is more advanced. However, existing lesions cannot be reversed. High intraocular pressure (IOP) remains one of the main risk factors and the only available therapy is based on its lowering to slow down the progression. It must be emphasized here that there are forms of glaucoma without elevated IOP (normal tension glaucoma) and that the disease continues to progress despite IOP-lowering treatment (Klein et al., 1992). Currently, a cure for glaucoma is not available. The persistent damage to the optic nerve eventually leads to severe visual impairment and even blindness in the patient. This imposes significant limitations on the patient’s independence and quality of life. In addition, it has immense socioeconomic implications. In Western Europe, the annual cost of monthly disability benefits for the blind, staff attrition, and early retirement due to glaucoma is higher than the medical cost of treating the disease. By 2040, Europe is projected to have 7.85 million glaucoma patients (Tham et al., 2014). Therefore, it is of great societal interest to find therapeutic strategies that significantly influence the progression of the disease. A deeper understanding of the underlying pathomechanisms is essential for glaucoma, as they are not fully understood yet. Today, it is already known that various mechanisms contribute to the development of the different forms of glaucoma. Among them, several studies have clearly demonstrated the importance of inflammatory processes in the pathogenesis of glaucoma. For example, differences in systemic as well as ocular antibody profiles were observed in patients suffering from glaucoma (Grus et al., 2004). In addition, antibody depositions were detected in glaucomatous retinae. To further elucidate this, the experimental autoimmune glaucoma (EAG) model was developed. Here, systemic immunization with ocular antigens, such as optic nerve homogenate antigen (ONA) or S100B protein, as part of ONA, leads to optic nerve degeneration and retinal ganglion cell loss (Laspas et al., 2011; Noristani et al., 2016)
    Related Articles | Metrics
    Is a mirror necessary for mirror therapy?
    Richard G. Carson, Alannah Morley
    2023, 18 (12):  2703-2704.  doi: 10.4103/1673-5374.373701
    Abstract ( 180 )   PDF (664KB) ( 43 )   Save
    Since it was first described as a method to treat phantom limb pain, mirror therapy (MT) has been applied in many areas of rehabilitation. Its seminal application with the aim of restoring motor function following stroke is considered to be that of Altschuler et al. (1999). The key features of MT in this context are that the stroke survivor performs movements of the less-impaired limb while looking in a mirror, that is positioned such that the reflected image gives rise to the impression that the more-impaired limb is also moving. In most discussions of the means through which MT exerts therapeutic effects, emphasis is placed upon an instrumental role of the visual feedback provided by the mirror. Although it is known that such feedback increases the excitability of corticospinal projections to the quiescent limb, there is scant evidence that this in itself gives rise to subsequent increases in functional movement capacity (Carson et al., 2016).
    Related Articles | Metrics
    Repurposing niclosamide for the treatment of neurological disorders
    Savina Apolloni, Nadia D’Ambrosi
    2023, 18 (12):  2705-2706.  doi: 10.4103/1673-5374.373705
    Abstract ( 119 )   PDF (451KB) ( 70 )   Save
    Neurological disorders are still one of the major causes of death, and the vast need to find efficacious therapy is nowadays an essential goal of the scientific community. For Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), spinal cord injury, and intracerebral hemorrhage, current pharmacological treatments are solely symptomatic, so there is a need to identify agents that can slow or stop neurodegeneration. Neurodegenerative diseases are caused by interactions between genetic, epigenetic, and environmental factors with consequent dysfunction of multiple cellular and molecular pathways. The multifactorial nature of the disorders could explain the modest results obtained by the treatments proposed so far. Moreover, the biochemical complexity of the pathological mechanisms highlights the need for multitarget therapies acting synergistically on different aspects of the diseases. Niclosamide, marketed as Yomesan for human use in 1962, is a Food and Drug Administration (FDA)-approved anti-helminthic drug used for over 50 years with considerable safety (Chen et al., 2018), and it is included in the World Health Organization’s list of essential medicines. Niclosamide is a member of the salicylanilide class of pharmacologic agents with an aryl β-hydroxy-carbonyl pharmacophore motif, usually present in many biological natural products. The pharmacophore motif confers to this small molecule its pleiotropic activities and the potential to interact with multiple biological targets. Its first documented action is to translocate protons across the mitochondrial membrane, resulting in mild mitochondrial uncoupling. This action is sufficient to kill tapeworms in the gastrointestinal tract but is generally well tolerated by human cells. In addition, niclosamide modulates Wnt/β-catenin, signal transducer and activator of transcription 3 (STAT3), mammalian target of rapamycin (mTOR), nuclear factor-kappa B (NF-κB), transmembrane protein 16 (TMEM16), and Notch signaling pathways (Chen et al., 2018). Since these molecules drive the transcription of multiple genes, it is possible that the broad biological activity displayed by the compound is the result of direct or indirect effects on these signaling pathways. Thanks to its pleiotropic actions, in recent years, niclosamide has been repurposed for several diseases. Preclinical validation proved that niclosamide has efficacy against solid cancers, rheumatoid arthritis, and fibrotic conditions, and it is currently in phase II–III clinical trial for metastatic colorectal cancer, prostate cancer, and coronavirus disease 2019 (COVID-19; Singh et al., 2022). 
    Related Articles | Metrics
    Etiology matters: genetic and acquired prion diseases engage different mechanisms at a presymptomatic stage
    Walker S. Jackson
    2023, 18 (12):  2707-2708.  doi: 10.4103/1673-5374.373684
    Abstract ( 91 )   PDF (354KB) ( 95 )   Save
    One of the most enigmatic problems in biomedical research surrounds the phenomenon that neurodegenerative diseases target specific cell types and brain regions. This is difficult to explain because the proteins that cause them are widely expressed, often highest in resistant regions. This mystery is further complicated by the fact that some disease-causing proteins are associated with multiple diseases. For example, the protein alpha-synuclein forms toxic aggregates in multiple diseases including Parkinson’s disease, dementia with Lewy bodies, and multiple systems atrophy, whereas the protein TDP43 can cause amyotrophic lateral sclerosis or frontotemporal lobar degeneration (Alegre-Abarrategui et al., 2019; Schweingruber and Hedlund, 2022). Alzheimer’s disease is associated with aggregates from two proteins, the amyloid precursor protein (APP) and Tau. Familial forms of Alzheimer’s disease are often associated with mutations in APP and enzymes that process APP. In contrast, mutations in Tau are not linked to Alzheimer’s disease but instead to frontal temporal dementia, whereas non-mutated Tau is also associated with several other neurodegenerative diseases (Carroll et al., 2021). Huntington’s disease is one of the few types of neurodegenerative disease that is strictly genetic. It is caused by an abnormally long polyglutamine tract in the Huntingtin protein, and the longer the tract the earlier the disease emerges. Remarkably, very long polyglutamine tracts also cause a wider distribution of affected brain regions, presenting with different clinical signs. The prion protein (PrP) also causes multiple, distinct diseases, collectively called prion diseases (Jackson, 2014).
    Related Articles | Metrics
    OxInflammation in Alzheimer’s disease
    Carlo Cervellati, Giovanni Zuliani, Giuseppe Valacchi
    2023, 18 (12):  2709-2710.  doi: 10.4103/1673-5374.374144
    Abstract ( 108 )   PDF (520KB) ( 48 )   Save
    OxInflammation: definition and role in disease: OxInflammation has been introduced as a new term in the biomedical field to highlight the deep and mutual relationship between the dysregulation of redox and immune homeostasis (Valacchi et al., 2018). Regardless of which is the initial spark, inflammation cannot take place without a concomitant oxidative stress response and vice-versa. In other words, they are sides of the same coin. 
    Related Articles | Metrics
    Quantitative proteomic and phosphoproteomic analyses of the hippocampus reveal the involvement of NMDAR1 signaling in repetitive mild traumatic brain injury
    Zhicheng Tian, Zixuan Cao, Erwan Yang, Juan Li, Dan Liao, Fei Wang, Taozhi Wang, Zhuoyuan Zhang, Haofuzi Zhang, Xiaofan Jiang, Xin Li, Peng Luo
    2023, 18 (12):  2711-2719.  doi: 10.4103/1673-5374.374654
    Abstract ( 196 )   PDF (2885KB) ( 142 )   Save
    The cumulative damage caused by repetitive mild traumatic brain injury can cause long-term neurodegeneration leading to cognitive impairment. This cognitive impairment is thought to result specifically from damage to the hippocampus. In this study, we detected cognitive impairment in mice 6 weeks after repetitive mild traumatic brain injury using the novel object recognition test and the Morris water maze test. Immunofluorescence staining showed that p-tau expression was increased in the hippocampus after repetitive mild traumatic brain injury. Golgi staining showed a significant decrease in the total density of neuronal dendritic spines in the hippocampus, as well as in the density of mature dendritic spines. To investigate the specific molecular mechanisms underlying cognitive impairment due to hippocampal damage, we performed proteomic and phosphoproteomic analyses of the hippocampus with and without repetitive mild traumatic brain injury. The differentially expressed proteins were mainly enriched in inflammation, immunity, and coagulation, suggesting that non-neuronal cells are involved in the pathological changes that occur in the hippocampus in the chronic stage after repetitive mild traumatic brain injury. In contrast, differentially expressed phosphorylated proteins were mainly enriched in pathways related to neuronal function and structure, which is more consistent with neurodegeneration. We identified N-methyl-D-aspartate receptor 1 as a hub molecule involved in the response to repetitive mild traumatic brain injury , and western blotting showed that, while N-methyl-D-aspartate receptor 1 expression was not altered in the hippocampus after repetitive mild traumatic brain injury, its phosphorylation level was significantly increased, which is consistent with the omics results. Administration of GRP78608, an N-methyl-D-aspartate receptor 1 antagonist, to the hippocampus markedly improved repetitive mild traumatic brain injury-induced cognitive impairment. In conclusion, our findings suggest that N-methyl-D-aspartate receptor 1 signaling in the hippocampus is involved in cognitive impairment in the chronic stage after repetitive mild traumatic brain injury and may be a potential target for intervention and treatment.
    Related Articles | Metrics
    Ca2+-induced myelin pathology precedes axonal spheroid formation and is mediated in part by store-operated Ca2+ entry after spinal cord injury#br#
    Spencer Ames, Kia Adams, Mariah E. Geisen, David P. Stirling
    2023, 18 (12):  2720-2726.  doi: 10.4103/1673-5374.373656
    Abstract ( 135 )   PDF (2707KB) ( 70 )   Save
    The formation of axonal spheroid is a common feature following spinal cord injury. To further understand the source of Ca2+ that mediates axonal spheroid formation, we used our previously characterized ex vivo mouse spinal cord model that allows precise perturbation of extracellular Ca2+. We performed two-photon excitation imaging of spinal cords isolated from Thy1YFP+ transgenic mice and applied the lipophilic dye, Nile red, to record dynamic changes in dorsal column axons and their myelin sheaths respectively. We selectively released Ca2+ from internal stores using the Ca2+ ionophore ionomycin in the presence or absence of external Ca2+. We reported that ionomycin dose-dependently induces pathological changes in myelin and pronounced axonal spheroid formation in the presence of normal 2 mM Ca2+ artificial cerebrospinal fluid. In contrast, removal of external Ca2+ significantly decreased ionomycin-induced myelin and axonal spheroid formation at 2 hours but not at 1 hour after treatment. Using mice that express a neuron-specific Ca2+ indicator in spinal cord axons, we confirmed that ionomycin induced significant increases in intra-axonal Ca2+, but not in the absence of external Ca2+. Periaxonal swelling and the resultant disruption in the axo-myelinic interface often precedes and is negatively correlated with axonal spheroid formation. Pretreatment with YM58483 (500 nM), a well-established blocker of store-operated Ca2+ entry, significantly decreased myelin injury and axonal spheroid formation. Collectively, these data reveal that ionomycin-induced depletion of internal Ca2+ stores and subsequent external Ca2+ entry through store-operated Ca2+ entry contributes to pathological changes in myelin and axonal spheroid formation, providing new targets to protect central myelinated fibers. 
    Related Articles | Metrics
    Fidgetin interacting with microtubule end binding protein EB3 affects axonal regrowth in spinal cord injury
    Chao Ma, Junpei Wang, Qifeng Tu, Weijuan Bo, Zunlu Hu, Run Zhuo, Ronghua Wu, Zhangji Dong, Liang Qiang, Yan Liu, Mei Liu
    2023, 18 (12):  2727-2732.  doi: 10.4103/1673-5374.373716
    Abstract ( 126 )   PDF (2455KB) ( 93 )   Save
    Fidgetin, a microtubule-severing enzyme, regulates neurite outgrowth, axonal regeneration, and cell migration by trimming off the labile domain of microtubule polymers. Because maintenance of the microtubule labile domain is essential for axon initiation, elongation, and navigation, it is of interest to determine whether augmenting the microtubule labile domain via depletion of fidgetin serves as a therapeutic approach to promote axonal regrowth in spinal cord injury. In this study, we constructed rat models of spinal cord injury and sciatic nerve injury. Compared with spinal cord injury, we found that expression level of tyrosinated microtubules in the labile portion of microtubules continuously increased, whereas fidgetin decreased after peripheral nerve injury. Depletion of fidgetin enhanced axon regeneration after spinal cord injury, whereas expression level of end binding protein 3 (EB3) markedly increased. Next, we performed RNA interference to knockdown EB3 or fidgetin. We found that deletion of EB3 did not change fidgetin expression. Conversely, deletion of fidgetin markedly increased expression of tyrosinated microtubules and EB3. Deletion of fidgetin increased the amount of EB3 at the end of neurites and thereby increased the level of tyrosinated microtubules. Finally, we deleted EB3 and overexpressed fidgetin. We found that fidgetin trimmed tyrosinated tubulins by interacting with EB3. When fidgetin was deleted, the labile portion of microtubules was elongated, and as a result the length of axons and number of axon branches were increased. These findings suggest that fidgetin can be used as a novel therapeutic target to promote axonal regeneration after spinal cord injury. Furthermore, they reveal an innovative mechanism by which fidgetin preferentially severs labile microtubules. 
    Related Articles | Metrics
    Bexarotene improves motor function after spinal cord injury in mice
    Xingyu Wang, Zhihao Shen, Haojie Zhang, Hao-Jie Zhang, Feida Li, Letian Yu, Hua Chen, Kailiang Zhou, Hui Xu, Sunren Sheng
    2023, 18 (12):  2733-2742.  doi: 10.4103/1673-5374.373676
    Abstract ( 142 )   PDF (4997KB) ( 89 )   Save
    Spinal cord injury is a challenge in orthopedics because it causes irreversible damage to the central nervous system. Therefore, early treatment to prevent lesion expansion is crucial for the management of patients with spinal cord injury. Bexarotene, a type of retinoid, exerts therapeutic effects on patients with cutaneous T-cell lymphoma and Parkinson’s disease. Bexarotene has been proven to promote autophagy, but it has not been used in the treatment of spinal cord injury. To investigate the effects of bexarotene on spinal cord injury, we established a mouse model of T11–T12 spinal cord contusion and performed daily intraperitoneal injection of bexarotene for 5 consecutive days. We found that bexarotene effectively reduced the deposition of collagen and the number of pathological neurons in the injured spinal cord, increased the number of synapses of nerve cells, reduced oxidative stress, inhibited pyroptosis, promoted the recovery of motor function, and reduced death. Inhibition of autophagy with 3-methyladenine reversed the effects of bexarotene on spinal cord injury. Bexarotene enhanced the nuclear translocation of transcription factor E3, which further activated AMP-activated protein kinase-S-phase kinase-associated protein 2-coactivator-associated arginine methyltransferase 1 and AMP-activated protein kinase-mammalian target of rapamycin signaling pathways. Intravenous injection of transcription factor E3 shRNA or intraperitoneal injection of compound C, an AMP-activated protein kinase blocker, inhibited the effects of bexarotene. These findings suggest that bexarotene regulates nuclear translocation of transcription factor E3 through the AMP-activated protein kinase-S-phase kinase-associated protein 2-coactivator-associated arginine methyltransferase 1 and AMP-activated protein kinase-mammalian target of rapamycin signal pathways, promotes autophagy, decreases reactive oxygen species level, inhibits pyroptosis, and improves motor function after spinal cord injury.
    Related Articles | Metrics
    Transcriptomic analysis of spinal cord regeneration after injury in Cynops orientalis
    Di Wang, Man Zhao, Xiao Tang, Man Gao, Wenjing Liu, Minghui Xiang, Jian Ruan, Jie Chen, Bin Long, Jun Li
    2023, 18 (12):  2743-2750.  doi: 10.4103/1673-5374.373717
    Abstract ( 184 )   PDF (3619KB) ( 209 )   Save
    Cynops orientalis (C. orientalis) has a pronounced ability to regenerate its spinal cord after injury. Thus, exploring the molecular mechanism of this process could provide new approaches for promoting mammalian spinal cord regeneration. In this study, we established a model of spinal cord thoracic transection injury in C. orientalis, which is an endemic species in China. We performed RNA sequencing of the contused axolotl spinal cord at two early time points after spinal cord injury – during the very acute stage (4 days) and the subacute stage (7 days) – and identified differentially expressed genes; additionally, we performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses, at each time point. Transcriptome sequencing showed that 13,059 genes were differentially expressed during C. orientalis spinal cord regeneration compared with uninjured animals, among which 4273 were continuously down-regulated and 1564 were continuously up-regulated. Down-regulated genes were most enriched in the Gene Ontology term “multicellular organismal process” and in the ribosome pathway at 10 days following spinal cord injury. We found that multiple genes associated with energy metabolism were down-regulated and multiple genes associated with the lysosome were up-regulated after spinal cord injury, indicating the importance of low metabolic activity during wound healing. Immune response-associated pathways were activated during the early acute phase (4 days), while the expression of extracellular matrix proteins such as glycosaminoglycan and collagen, as well as tight junction proteins, was lower at 10 days post-spinal cord injury than 4 days post-spinal cord injury. However, compared with 4 days post-injury, at 10 days post-injury neuroactive ligand-receptor interactions were no longer down-regulated, up-regulated differentially expressed genes were enriched in pathways associated with cancer and the cell cycle, and SHH, VIM, and Sox2 were prominently up-regulated. Immunofluorescence staining showed that glial fibrillary acidic protein was up-regulated in axolotl ependymoglial cells after injury, similar to what is observed in mammalian astrocytes after spinal cord injury, even though axolotls do not form a glial scar during regeneration. We suggest that low intracellular energy production could slow the rapid amplification of ependymoglial cells, thereby inhibiting reactive gliosis, at early stages after spinal cord injury. Extracellular matrix degradation slows cellular responses, represses the expression of neurogenic genes, and reactivates a transcriptional program similar to that of embryonic neuroepithelial cells. These ependymoglial cells act as neural stem cells: they migrate and proliferate to repair the lesion and then differentiate to replace lost glial cells and neurons. This provides the regenerative microenvironment that allows axon growth after injury. 
    Related Articles | Metrics
    A novel mouse model of central cord syndrome
    Elzat Elham – Yilizati Yilihamu, Xiangchuang Fan, Zimeng Yang, Shiqing Feng
    2023, 18 (12):  2751-2756.  doi: 10.4103/1673-5374.373718
    Abstract ( 118 )   PDF (2257KB) ( 116 )   Save
    Patients with potential spinal stenosis are susceptible to central cord syndrome induced by blunt trauma. Suitable animal models are helpful for studying the pathogenesis and treatment of such injuries. In this study, we established a mouse model of acute blunt traumatic spinal cord injury by compressing the C6 spinal cord with 5 and 10 g/mm2 compression weights to simulate cervical central cord syndrome. Behavioral testing confirmed that this model exhibited the characteristics of central cord syndrome because motor function in the front paws was impaired, whereas basic motor and sensory functions of the lower extremities were retained. Hematoxylin-eosin staining showed that the diseased region of the spinal cord in this mouse model was restricted to the gray matter of the central cord, whereas the white matter was rarely affected. Magnetic resonance imaging showed a hypointense signal in the lesion after mild and severe injury. In addition, immunofluorescence staining showed that the degree of nerve tract injury in the spinal cord white matter was mild, and that there was a chronic inflammation reaction. These findings suggest that this mouse model of central cord syndrome can be used as a model for preclinical research, and that gray matter is most vulnerable to injury in central cord syndrome, leading to impaired motor function.
    Related Articles | Metrics
    Motor neuron-specific RhoA knockout delays degeneration and promotes regeneration of dendrites in spinal ventral horn after brachial plexus injury
    Mi Li, Jiawei Xu, Ying Zou, Jialing Lu, Aiyue Ou, Xinrui Ma, Jiaqi Zhang, Yizhou Xu, Lanya Fu, Jingmin Liu, Xianghai Wang, Libing Zhou, Jiasong Guo
    2023, 18 (12):  2757-2761.  doi: 10.4103/1673-5374.373657
    Abstract ( 134 )   PDF (5421KB) ( 149 )   Save
    Dendrites play irreplaceable roles in the nerve conduction pathway and are vulnerable to various insults. Peripheral axotomy of motor neurons results in the retraction of dendritic arbors, and the dendritic arbor can be re-expanded when reinnervation is allowed. RhoA is a target that regulates the cytoskeleton and promotes neuronal survival and axon regeneration. However, the role of RhoA in dendrite degeneration and regeneration is unknown. In this study, we explored the potential role of RhoA in dendrites. A line of motor neuronal RhoA conditional knockout mice was developed by crossbreeding HB9Cre+ mice with RhoAflox/flox mice. We established two models for assaying dendrite degeneration and regeneration, in which the brachial plexus was transection or crush injured, respectively. We found that at 28 days after brachial plexus transection, the density, complexity, and structural integrity of dendrites in the ventral horn of the spinal cord of RhoA conditional knockout mice were slightly decreased compared with that in Cre mice. Dendrites underwent degeneration at 7 and 14 days after brachial plexus transection and recovered at 28–56 days. The density, complexity, and structural integrity of dendrites in the ventral horn of the spinal cord of RhoA conditional knockout mice recovered compared with results in Cre mice. These findings suggest that RhoA knockout in motor neurons attenuates dendrite degeneration and promotes dendrite regeneration after peripheral nerve injury.
    Related Articles | Metrics
    Nerve function restoration following targeted muscle reinnervation after varying delayed periods
    Yuanheng Li, Jiangping Huang, Yuling Chen, Shanshan Zhu, Zhen Huang, Lin Yang, Guanglin Li
    2023, 18 (12):  2762-2766.  doi: 10.4103/1673-5374.373659
    Abstract ( 117 )   PDF (1813KB) ( 64 )   Save
    Targeted muscle reinnervation has been proposed for reconstruction of neuromuscular function in amputees. However, it is unknown whether performing delayed targeted muscle reinnervation after nerve injury will affect restoration of function. In this rat nerve injury study, the median and musculocutaneous nerves of the forelimb were transected. The proximal median nerve stump was sutured to the distal musculocutaneous nerve stump immediately and 2 and 4 weeks after surgery to reinnervate the biceps brachii. After targeted muscle reinnervation, intramuscular myoelectric signals from the biceps brachii were recorded. Signal amplitude gradually increased with time. Biceps brachii myoelectric signals and muscle fiber morphology and grooming behavior did not significantly differ among rats subjected to delayed target muscle innervation for different periods. Targeted muscle reinnervation delayed for 4 weeks can acquire the same nerve function restoration effect as that of immediate reinnervation. 
    Related Articles | Metrics
    Ketogenic diet alleviates cognitive dysfunction and neuroinflammation in APP/PS1 mice via the Nrf2/HO-1 and NF-κB signaling pathways
    Jingwen Jiang, Hong Pan, Fanxia Shen, Yuyan Tan, Shengdi Chen
    2023, 18 (12):  2767-2772.  doi: 10.4103/1673-5374.373715
    Abstract ( 237 )   PDF (1852KB) ( 94 )   Save
    Alzheimer’s disease is a progressive neurological disorder characterized by cognitive decline and chronic inflammation within the brain. The ketogenic diet, a widely recognized therapeutic intervention for refractory epilepsy, has recently been proposed as a potential treatment for a variety of neurological diseases, including Alzheimer’s disease. However, the efficacy of ketogenic diet in treating Alzheimer’s disease and the underlying mechanism remains unclear. The current investigation aimed to explore the effect of ketogenic diet on cognitive function and the underlying biological mechanisms in a mouse model of Alzheimer’s disease. Male amyloid precursor protein/presenilin 1 (APP/PS1) mice were randomly assigned to either a ketogenic diet or control diet group, and received their respective diets for a duration of 3 months. The findings show that ketogenic diet administration enhanced cognitive function, attenuated amyloid plaque formation and proinflammatory cytokine levels in APP/PS1 mice, and augmented the nuclear factor-erythroid 2-p45 derived factor 2/heme oxygenase-1 signaling pathway while suppressing the nuclear factor-kappa B pathway. Collectively, these data suggest that ketogenic diet may have a therapeutic potential in treating Alzheimer’s disease by ameliorating the neurotoxicity associated with Aβ-induced inflammation. This study highlights the urgent need for further research into the use of ketogenic diet as a potential therapy for Alzheimer’s disease.
    Related Articles | Metrics
    Selective deletion of zinc transporter 3 in amacrine cells promotes retinal ganglion cell survival and optic nerve regeneration after injury
    Zhe Liu, Jingfei Xue, Canying Liu, Jiahui Tang, Siting Wu, Jicheng Lin, Jiaxu Han, Qi Zhang, Caiqing Wu, Haishun Huang, Ling Zhao, Yehong Zhuo, Yiqing Li
    2023, 18 (12):  2773-2780.  doi: 10.4103/1673-5374.373660
    Abstract ( 138 )   PDF (2519KB) ( 106 )   Save
    Vision depends on accurate signal conduction from the retina to the brain through the optic nerve, an important part of the central nervous system that consists of bundles of axons originating from retinal ganglion cells. The mammalian optic nerve, an important part of the central nervous system, cannot regenerate once it is injured, leading to permanent vision loss. To date, there is no clinical treatment that can regenerate the optic nerve and restore vision. Our previous study found that the mobile zinc (Zn2+) level increased rapidly after optic nerve injury in the retina, specifically in the vesicles of the inner plexiform layer. Furthermore, chelating Zn2+ significantly promoted axonal regeneration with a long-term effect. In this study, we conditionally knocked out zinc transporter 3 (ZnT3) in amacrine cells or retinal ganglion cells to construct two transgenic mouse lines (VGATCreZnT3fl/fl and VGLUT2CreZnT3fl/fl, respectively). We obtained direct evidence that the rapidly increased mobile Zn2+ in response to injury was from amacrine cells. We also found that selective deletion of ZnT3 in amacrine cells promoted retinal ganglion cell survival and axonal regeneration after optic nerve crush injury, improved retinal ganglion cell function, and promoted vision recovery. Sequencing analysis of reginal ganglion cells revealed that inhibiting the release of presynaptic Zn2+ affected the transcription of key genes related to the survival of retinal ganglion cells in postsynaptic neurons, regulated the synaptic connection between amacrine cells and retinal ganglion cells, and affected the fate of retinal ganglion cells. These results suggest that amacrine cells release Zn2+ to trigger transcriptomic changes related to neuronal growth and survival in reginal ganglion cells, thereby influencing the synaptic plasticity of retinal networks. These results make the theory of zinc-dependent retinal ganglion cell death more accurate and complete and provide new insights into the complex interactions between retinal cell networks.
    Related Articles | Metrics
    Acute penetrating injury of the spinal cord by a wooden spike with delayed surgery: a case report
    James D. Guest, Zhuojing Luo, Yansheng Liu, Hongkun Gao, Dianchun Wang, Xiao-Ming Xu, Hui Zhu
    2023, 18 (12):  2781-2784.  doi: 10.4103/1673-5374.373668
    Abstract ( 80 )   PDF (854KB) ( 90 )   Save
    Rarely, penetrating injuries to the spinal cord result from wooden objects, creating unique challenges to mitigate neurological injury and high rates of infection and foreign body reactions. We report a man who sustained a penetrating cervical spinal cord injury from a sharpened stick. While initially tetraparetic, he rapidly recovered function. The risks of neurological deterioration during surgical removal made the patient reluctant to consent to surgery despite the impalement of the spinal cord. A repeat MRI on day 3 showed an extension of edema indicating progressive inflammation. On the 7th day after injury, fever and paresthesias occurred with a large increase in serum inflammatory indicators, and the patient agreed to undergo surgical removal of the wooden object. We discuss the management nuances related to wood, the longitudinal evolution of MRI findings, infection risk, surgical risk mitigation and technique, an inflammatory marker profile, long-term recovery, and the surprisingly minimal neurological deficits associated with low-velocity midline spinal cord injuries. The patient had an excellent clinical outcome. The main lessons are that a wooden penetrating central nervous system injury has a high risk for infection, and that surgical removal from the spinal cord should be performed soon after injury and under direct visualization. 
    Related Articles | Metrics