Loading...

Table of Content

    15 December 2021, Volume 16 Issue 12 Previous Issue   
    For Selected: Toggle Thumbnails
    Inhibition of extracellular vesicle pathway using neutral sphingomyelinase inhibitors as a neuroprotective treatment for brain injury
    Asit Kumar, Santosh Kumar
    2021, 16 (12):  2349-2352.  doi: 10.4103/1673-5374.313014
    Abstract ( 143 )   PDF (507KB) ( 364 )   Save
    Traumatic brain injury is a sudden trauma or blow on the head, and severe traumatic brain injury is a major cause of death and disability worldwide. The acute and chronic consequences following traumatic brain injury can lead to progressive secondary neurodegenerative changes and cognitive dysfunction. To date, there is no effective pharmaceutical products for the treatment to reduce secondary damage after brain injury. The discovery of extracellular vesicles has attracted considerable scientific attention due to their role in cell-to-cell communication. Extracellular vesicles have shown their potential to carry not only biological molecules but also as a drug delivery vehicle. As a carrier of molecular information, extracellular vesicles have been involved in physiological functions as well as in the modulation of immune responses. Here, we aim to provide new insights into the contrasting role of extracellular vesicles in the propagation of inflammatory responses after brain injury. As a carrier of pro-inflammatory molecules, their role as functional mediators in the pathophysiology of brain injury is discussed, addressing the inhibition of the extracellular vesicle pathway as an anti-inflammatory or neuroprotective approach to improve the outcome of both acute and chronic inflammation following brain injury. Here, we summarize therapeutic strategies to diminish the risk the neurodegeneration post brain injury and propose that neutral sphingomyelinase inhibitors could be used as potentially useful therapeutic agents for the treatment of brain injury associated neuroinflammation.
    Related Articles | Metrics
    Neuroglobin and neuroprotection: the role of natural and synthetic compounds in neuroglobin pharmacological induction
    Lidia Ciccone, Susanna Nencetti, Simone Socci, Elisabetta Orlandini
    2021, 16 (12):  2353-2358.  doi: 10.4103/1673-5374.300981
    Abstract ( 257 )   PDF (490KB) ( 161 )   Save
    Neuroglobin (Ngb) is a 17 kDa monomeric hexa-coordinated heme protein belonging to the globin family. Ngb is mainly expressed in neurons of the central and peripheral nervous system, although moderate levels of Ngb have been detected in non-nervous tissues. In the past decade, Ngb has been studied for its neuroprotective role in a large number of neurological disorders such as Alzheimer’s disease, Huntington’s disease, brain ischemia and hypoxia. This review discusses and summarizes the natural compounds and the small synthetic molecules capable of modulating Ngb expression that exhibits a protective role against various neurodegenerative diseases.
    Related Articles | Metrics
    Potential effects of mesenchymal stem cell derived extracellular vesicles and exosomal miRNAs in neurological disorders
    Masako Nakano, Mineko Fujimiya
    2021, 16 (12):  2359-2366.  doi: 10.4103/1673-5374.313026
    Abstract ( 268 )   PDF (440KB) ( 207 )   Save
    Mesenchymal stem cells are multipotent cells that possess anti-inflammatory, anti-apoptotic and immunomodulatory properties. The effects of existing drugs for neurodegenerative disorders such as Alzheimer’s disease are limited, thus mesenchymal stem cell therapy has been anticipated as a means of ameliorating neuronal dysfunction. Since mesenchymal stem cells are known to scarcely differentiate into neuronal cells in damaged brain after transplantation, paracrine factors secreted from mesenchymal stem cells have been suggested to exert therapeutic effects. Extracellular vesicles and exosomes are small vesicles released from mesenchymal stem cells that contain various molecules, including proteins, mRNAs and microRNAs. In recent years, administration of exosomes/extracellular vesicles in models of neurological disorders has been shown to improve neuronal dysfunctions, via exosomal transfer into damaged cells. In addition, various microRNAs derived from mesenchymal stem cells that regulate various genes and reduce neuropathological changes in various neurological disorders have been identified. This review summarizes the effects of exosomes/extracellular vesicles and exosomal microRNAs derived from mesenchymal stem cells on models of stroke, subarachnoid and intracerebral hemorrhage, traumatic brain injury, and cognitive impairments, including Alzheimer’s disease.
    Related Articles | Metrics
    Epidural electrical stimulation for spinal cord injury
    Elliot H. Choi, Sandra Gattas, Nolan J. Brown, John D. Hong, Joshua N. Limbo, Alvin Y. Chan, Michael Y. Oh
    2021, 16 (12):  2367-2375.  doi: 10.4103/1673-5374.313017
    Abstract ( 400 )   PDF (424KB) ( 252 )   Save
    A long-standing goal of spinal cord injury research is to develop effective repair strategies, which can restore motor and sensory functions to near-normal levels. Recent advances in clinical management of spinal cord injury have significantly improved the prognosis, survival rate and quality of life in patients with spinal cord injury. In addition, a significant progress in basic science research has unraveled the underlying cellular and molecular events of spinal cord injury. Such efforts enabled the development of pharmacologic agents, biomaterials and stem-cell based therapy. Despite these efforts, there is still no standard care to regenerate axons or restore function of silent axons in the injured spinal cord. These challenges led to an increased focus on another therapeutic approach, namely neuromodulation. In multiple animal models of spinal cord injury, epidural electrical stimulation of the spinal cord has demonstrated a recovery of motor function. Emerging evidence regarding the efficacy of epidural electrical stimulation has further expanded the potential of epidural electrical stimulation for treating patients with spinal cord injury. However, most clinical studies were conducted on a very small number of patients with a wide range of spinal cord injury. Thus, subsequent studies are essential to evaluate the therapeutic potential of epidural electrical stimulation for spinal cord injury and to optimize stimulation parameters. Here, we discuss cellular and molecular events that continue to damage the injured spinal cord and impede neurological recovery following spinal cord injury. We also discuss and summarize the animal and human studies that evaluated epidural electrical stimulation in spinal cord injury.
    Related Articles | Metrics
    Localization of the hydrogen sulfide and oxytocin systems at the depth of the sulci in a porcine model of acute subdural hematoma
    Oscar McCook, Angelika Scheuerle, Nicole Denoix, Thomas Kapapa, Peter Radermacher, Tamara Merz
    2021, 16 (12):  2376-2382.  doi: 10.4103/1673-5374.313018
    Abstract ( 121 )   PDF (947KB) ( 145 )   Save
    In the porcine model discussed in this review, the acute subdural hematoma was induced by subdural injection of autologous blood over the left parietal cortex, which led to a transient elevation of the intracerebral pressure, measured by bilateral neuromonitoring. The hematoma-induced brain injury was associated with albumin extravasation, oxidative stress, reactive astrogliosis and microglial activation in the ipsilateral hemisphere. Further proteins and injury markers were validated to be used for immunohistochemistry of porcine brain tissue. The cerebral expression patterns of oxytocin, oxytocin receptor, cystathionine-γ-lyase and cystathionine-β-synthase were particularly interesting: these four proteins all co-localized at the base of the sulci, where pressure-induced brain injury elicits maximum stress. In this context, the pig is a very relevant translational model in contrast to the rodent brain. The structure of the porcine brain is very similar to the human: the presence of gyri and sulci (gyrencephalic brain), white matter to grey matter proportion and tentorium cerebelli. Thus, pressure-induced injury in the porcine brain, unlike in the rodent brain, is reflective of the human pathophysiology. 
    Related Articles | Metrics
    Lithium beyond psychiatric indications: the reincarnation of a new old drug
    Matteo Haupt, Mathias Bähr, Thorsten R. Doeppner
    2021, 16 (12):  2383-2387.  doi: 10.4103/1673-5374.313015
    Abstract ( 159 )   PDF (589KB) ( 162 )   Save
    Lithium has been used in the treatment of bipolar disorders for decades, but the exact mechanisms of action remain elusive to this day. Recent evidence suggests that lithium is critically involved in a variety of signaling pathways affecting apoptosis, inflammation, and neurogenesis, all of which contributing to the complex pathophysiology of various neurological diseases. As a matter of fact, preclinical work reports both acute and long-term neuroprotection in distinct neurological disease models such as Parkinson’s disease, traumatic brain injury, Alzheimer’s disease, and ischemic stroke. Lithium treatment reduces cell injury, decreases α‑synuclein aggregation and Tau protein phosphorylation, modulates inflammation and even stimulates neuroregeneration under experimental conditions of Parkinson’s disease, traumatic brain injury, and Alzheimer’s disease. The therapeutic impact of lithium under conditions of ischemic stroke was also studied in numerous preclinical in vitro and in vivo studies, giving rise to a randomized double-blind clinical stroke trial. The preclinic data revealed a lithium-induced upregulation of anti-apoptotic proteins such as B-cell lymphoma 2, heat shock protein 70, and activated protein 1, resulting in decreased neuronal cell loss. Lithium, however, does not only yield postischemic neuroprotection but also enhances endogenous neuroregeneration by stimulating neural stem cell proliferation and by regulating distinct signaling pathways such as the RE1-silencing transcription factor. In line with this, lithium treatment has been shown to modulate postischemic cytokine secretion patterns, diminishing microglial activation and stabilizing blood-brain barrier integrity yielding reduced levels of neuroinflammation. The aforementioned observations culminated in a first clinical trial, which revealed an improved motor recovery in patients with cortical stroke after lithium treatment. Beside its well-known psychiatric indications, lithium is thus a promising neuroprotective candidate for the aforementioned neurological diseases. A detailed understanding of the lithium-induced mechanisms, however, is important for prospective clinical trials which may pave the way for a successful bench-to-bedside translation in the future. In this review, we will give an overview of lithium-induced neuroprotective mechanisms under various pathological conditions, with special emphasis on ischemic stroke. 
    Related Articles | Metrics
    Abnormal metal homeostasis as a common drug target to combat neurodegenerative diseases
    Dagmar Pretsch
    2021, 16 (12):  2388-2389.  doi: 10.4103/1673-5374.313039
    Abstract ( 127 )   PDF (236KB) ( 103 )   Save
    As life expectancy increases, the prevalence of age-related diseases will also increase. There has been an estimation that 50 million individuals are living with dementia in 2019. This number will increase to 75 million in 2030 and 131.5 million by 2050 (McGill-Carter, 2020). Neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson´s disease (PD) and Huntington disease (HD) disproportionately affect older adult populations, inflicting a considerable physical, emotional, and economic burden.
    Related Articles | Metrics
    Caspase-1: an important player and possible target for repair of the blood-brain barrier underlying neurodegeneration
    Daniel Rand, Itzik Cooper
    2021, 16 (12):  2390-2392.  doi: 10.4103/1673-5374.313031
    Abstract ( 189 )   PDF (678KB) ( 194 )   Save
    Our ageing population is bearing an increasing burden on society with an unprecedented rise in brain disease, especially neurodegeneration. Treatment for neurodegeneration is practically non-existent, one major reason being the blood-brain barrier (BBB). The BBB is a crucial component in maintaining a healthy brain as it protects the brain from the entrance of toxins and removes them from the brain’s vicinity, providing proper clearance. Therefore, BBB malfunction is the basis for numerous brain diseases making it a major target for potential new treatments. A major common underlying mechanism for BBB damage is excessive central nervous system (CNS) inflammation. In turn, BBB disruption has been linked to the initiation of neuroinflammation, forming a vicious cycle. The release of cytokines and destruction of cells are associated with BBB breakdown through multiple molecular mechanisms leading to the chemical and hormonal dysregulation of the brain, increased immune cell infiltration, decreased removal of waste, and degeneration of CNS cells that depending on the affected brain region—ultimately lead to cognitive decline and other CNS disorders (Van Dyken and Lacoste, 2018; Figure 1). 
    Related Articles | Metrics
    Microglia activation: a role for mitochondrial DNA?
    Marcello Pinti, Diana Ferraro, Milena Nasi
    2021, 16 (12):  2393-2394.  doi: 10.4103/1673-5374.313034
    Abstract ( 432 )   PDF (399KB) ( 190 )   Save
    Microglial cells and perivascular macrophages are the only resident immune cells of the brain parenchyma and act as innate immune sentinels in the central nervous system (CNS). Microglial cells are vital for the maintenance of CNS homeostasis thanks to their strict interaction with neurons. When the homeostasis of the microenvironment is disrupted, microglia can alter their phenotype acquiring pro- or anti-inflammatory function to defend the brain. On the other hand, the excessive activation of proinflammatory microglia in response to primary neurodegeneration, axonal degeneration, and additional peripheral activation processes linked to systemic inflammation can trigger or maintain chronic inflammation. Therefore, under such conditions, the proinflammatory phenotype of microglia could be harmful and associated with the pathogenesis of neurological disease characterized by inflammation, such as neurodegenerative diseases, demyelinating diseases, CNS trauma, and epilepsy. Despite the numerous studies on that field, the primary stimuli that provoke and maintain such inflammation, as well as the biological pathways and mechanisms that cause detrimental actions of microglia are still a subject of debate. Microglia can sense cellular damage and stress by recognizing the damage-associated molecular patterns (DAMPs) through the pattern recognition receptors (PRRs). Several lines of evidence, obtained from studies in humans and animal models, suggest that DAMPs could play a relevant role in the pathogenesis of several neurodegenerative diseases (Gong et al., 2020). The category of DAMPs includes several molecules, some of them can be released from damaged mitochondria (the so-called mitochondrial DAMPs, mtDAMPs), such as N-formyl peptides, cardiolipin, the mitochondrial transcription factor A (TFAM), succinate, adenosine triphosphate, and mitochondrial DNA (mtDNA). Damaged cells accumulate dysfunctional mitochondria that trigger processes such as cell senescence, apoptosis, or necrosis. In all of these cases, mtDAMPs can be released in the extracellular space and could be recognized through different pattern recognition receptors by innate immune cells recruited to remove cellular debris of dying cells. Recently, increasing attention has been paid to mtDNA, as DAMP able to strongly stimulate cells through Toll-like receptor (TLR) 9 contributing to inflammation even in the absence of infection (sterile inflammation) (Riley et al., 2020). After an extensive cell injury, several mitochondrial products, including mtDNA, can enter the bloodstream or cerebrospinal fluid (CSF), where they are recognized by the innate immune system and evoke a local or systemic response. The cell-free mtDNA is stable and resistant to nuclease digestion, more than genomic DNA, and could be detected in blood or CSF. Even in healthy people, mtDNA is present at relatively high levels in the blood and easily measurable. Over the past few years, there has been a growing interest in mtDNA as a potential biomarker as its levels are increased in several physio-pathological conditions characterized by chronic inflammation (Cossarizza et al., 2011; Pinti et al., 2014; Nasi et al., 2016), including neurodegenerative diseases such as multiple sclerosis (MS) (Nasi et al., 2020a). Interestingly, mtDNA levels were found higher also in CSF from people with MS but not in people affected by Parkinson’s disease or Alzheimer’s disease (Gambardella et al., 2019). Parkinson’s disease and Alzheimer’s disease are characterized by a loss of neuronal mitochondria (where probably the low levels of mtDNA in the CSF come from) followed by neuronal death, while MS is characterized by a strong inflammatory response in which mtDNA could be released into the CSF. Thus, MS represents a valuable model of neuro-inflammation, in which mtDAMPs could have a prominent role. On the other hand, the neuro-inflammation itself is strictly associated with mitochondrial dysfunction that could trigger a vicious circle: dysfunctional mitochondria can induce inflammation and inflammation induces mitochondrial dysfunction followed by the further release of mtDAMPs. However, the triggers by which mtDAMPs are released are still unknown, as well as the precise role of mtDNA and mtDAMPs in patients with MS has poorly been investigated.
    Related Articles | Metrics
    Light of the stars: calcium signals light up astrocytes in the presence of autoreactive CD4+ T cells
    Jelena Bogdanović Pristov, Ljiljana Nikolić
    2021, 16 (12):  2395-2396.  doi: 10.4103/1673-5374.313036
    Abstract ( 141 )   PDF (404KB) ( 122 )   Save
    Astrocytes play important roles in the central nervous system (CNS) to support and regulate CNS function. They are abundant type of glia that form a meshwork of interconnected cells almost completely tiling the CNS. Each astrocyte extends densely ramified processes that establish close contacts and interaction with other astrocytes, neurons and blood vessels, placing these cells in an ideal position to control extracellular milieu and to exert global effects on CNS physiology. Astrocytes are dynamic cells that continuously sense and respond to the physiological and pathological cues within their local environment. This responsiveness of astrocytes and their intercellular communication with neighboring cells in the CNS principally relies on increase in intracellular Ca2+ and release of neuroactive compounds such as adenosine 5′-triphosphate (ATP), that are critical for maintaining proper CNS function (Butt, 2011; Bazargani and Attwell, 2016). Intracellular Ca2+ level in astrocytes is under the control of diverse ionotropic and metabotropic receptors that upon activation promote Ca2+ entry into the cell or its liberation from cellular Ca2+ stores. This promotes rise of Ca2+ signals in astrocytes that spread information between astrocytes themselves and toward neurons to regulate CNS function. When CNS environment becomes disturbed following injury or in disease, astrocytes generally respond with an augmentation of Ca2+ signaling within their network that has deleterious effects and drive pathological processes (Shigetomi et al., 2019). Thus, Ca2+ signaling acts as a powerful and highly adaptable system for astrocyte communication with neighboring cells in the CNS and enables astrocytes to detect and respond to changes in their local environment in both, physiological and pathological conditions. In our recent paper (Bijelić et al., 2020) we show that astrocyte Ca2+ signals controlled by purinergic P2X7 ionotropic receptors are also important for astrocytic communication with specific immune cells that infiltrate into the CNS in experimental autoimmune encephalomyelitis, a commonly used animal model of multiple sclerosis. We show that astrocytes respond to the nearby autoreactive CD4+ T cells with an increase in their intracellular Ca2+ (Figure 1), hinting at the previously unrecognized involvement of astrocytic Ca2+ signaling in the nervous system-immune system communication in an autoimmune disease of CNS.  

    Related Articles | Metrics
    Exploring genes that control microglial heterogeneity and transition
    Tomomi Okajima, Fuminori Tsuruta
    2021, 16 (12):  2397-2398.  doi: 10.4103/1673-5374.313035
    Abstract ( 114 )   PDF (763KB) ( 134 )   Save
    Microglia, which comprise approximately 10% of total cells in the brain, are the resident immune cells in the central nervous system and contribute to maintaining the brain homeostasis through monitoring their microenvironment (Kettenmann et al., 2011). Recent studies have reported that microglia also regulate neural circuit formation after birth. The functional transition in microglia has been considered to correlate with their morphological changes over time. Although microglial morphologies were found to be modulated by expressions of particular genes, the mechanisms that underlie morphological changes in microglia have not been fully elucidated. Recently, we have reported that FAT atypical cadherin family protein 3 (FAT3) is a novel factor that stabilizes microglial processes regulated by hypoxanthine (Okajima et al., 2020). Since the timing of FAT3 expression coincides with that of microglial morphological changes during the postnatal stage, it is plausible that FAT3 regulates the microglial transition. In this review, we discuss the biological relevance among microglial heterogeneity and transitions. Moreover, we introduce our recent findings that link FAT3 to the aspect of microglial transition and functions during the postnatal stage.
    Related Articles | Metrics
    Netrin-1 marshals mitochondrial movement, morphology, and metabolism in myelin
    Diane S. Nakamura, Timothy E. Kennedy
    2021, 16 (12):  2399-2400.  doi: 10.4103/1673-5374.313028
    Abstract ( 133 )   PDF (480KB) ( 155 )   Save
    Oligodendrocytes are the myelinating cells of the central nervous system (CNS) that ensheath nearby axons to support action potential propagation and axon metabolism. Myelination involves the rapid production of lipid-rich membrane, compaction of the multilamellar myelin sheath, and the resultant restriction of cytoplasm to non-compact compartments. During myelination, septate-like junctions form between the axon and lateral cytoplasmic endings of the myelin sheath at a specialized domain called the paranode (Figure 1A). Cytoplasm-filled loops at the paranode (hereon referred to as paranodal loops) host a network of organelles, but their regulation and function are poorly understood. Until recently, it was not at all clear how molecules from the oligodendrocyte cell body were transported across the compacted layers of the myelin sheath to access the cytoplasmic growing edge. In 2014, cytoplasmic channels that traverse the compact myelin sheath were identified in the CNS (Snaidero et al., 2014), opening the door to investigate the function and regulation of the organelles contained within these specialized cytoplasmic compartments. Further, it was shown that active mitochondria in non-compact myelin contribute to the generation of calcium transients that appear to regulate internode remodeling (Battefeld et al., 2019), supporting a functional link between mitochondria and myelin remodeling.
    Related Articles | Metrics
    A 3R-Tau-mediated mechanism in oligodendrocytes: could it be the key for neuroprotection after stroke?
    Mario Villa Gonzalez, Maria José Pérez-Álvarez
    2021, 16 (12):  2401-2402.  doi: 10.4103/1673-5374.313027
    Abstract ( 154 )   PDF (374KB) ( 139 )   Save
    Cerebrovascular accident or stroke have a high global incidence. The most common types of stroke are ischemic, accounting for 87% of the total number, and they are triggered by a reduction or interruption of blood flow to the central nervous system, usually caused by a thrombus, embolus or atherosclerotic plaque. The severity of brain damage caused by this kind of stroke is directly related to the size of the vessel that is occluded and the duration of occlusion. Stroke is currently the leading cause of long-term disability worldwide and as such it has an enormous socioeconomic impact (Benjamin et al., 2019). In fact, between 30–50% of stroke patients do not recover functional independence and need personal assistance to carry out normal everyday activities. Also, according to statistics issued by the World Health Organization (WHO), ischemic stroke is the second cause of death worldwide. This year (2020) has witnessed a new cause of stroke related to coronavirus disease 2019 (COVID-19), with devastating consequences for the prognosis of this disease across all ages (García-Moncó et al., 2020). Given that the incidence of stroke increases with age and life expectancy is rising worldwide, the WHO predicts an increase in the prevalence and incidence of this condition in the coming years (Benjamin et al., 2019). The only therapeutic strategy currently used to reduce ischemic brain damage is early reperfusion using surgical methods (mechanical thrombectomy), or more frequently, the administration of a thrombolytic agent, namely the recombinant tissue plasminogen activator. However, the last approach has a therapeutic window limited to 4.5 hours after the first symptoms appear (Biggs et al., 2019). This temporal restriction implies that reperfusion is not a suitable strategy for a significant number of stroke patients due to the high risk of cerebral hemorrhage. Statistics in different countries reflect that only 10–20% of patients in acute phase of stroke, receive thrombolysis treatment (Lees et al., 2010). The remaining patients do not receive any pharmacological therapy, thus precluding the opportunity to reduce the disability and cognitive dysfunction caused by ischemia. An extension of the time window up to 24 hours has been reported for mechanical thrombectomy in certain patients (Dmytriw et al., 2019). Although reperfusion enhances the prognosis of stroke, it does not prevent the neurodegenerative processes that occur after damage. Therefore, to improve the beneficial effects of reperfusion and to enable pharmacological intervention in patients for whom this approach is not suitable, it is critical to explore alternative therapeutic strategies.

    Related Articles | Metrics
    Combinatorial genetics methods for discovering high-order regulatory combinations and engineering genetic drivers for neural differentiation
    Dawn G. L. Thean, Alan S. L. Wong
    2021, 16 (12):  2403-2404.  doi: 10.4103/1673-5374.313038
    Abstract ( 110 )   PDF (568KB) ( 97 )   Save
    Researchers are still striving to find better therapeutics to revert or slow down the progression of neurological disorders such as Parkinson’s disease, Alzheimer’s disease, and Huntington’s disease. These disorders are the result of neuronal cell death in different parts of the brain. Medications or treatments used to relieve the symptoms in patients have not been well established. They do not recover the damaged neural tissues and can result in unwanted side effects. Therefore, an increasing number of studies look to stem cells as a promising therapeutic, because of their self-renewal capabilities and flexibility of differentiation into desired cell lineages for engraftment into the patient to recover the lost neural tissues. However, before stem cells can be clinically used in treating neurological disorders, there are still areas that require a better understanding to unlock their full potential.
    Related Articles | Metrics
    DNMT1-dependent regulation of cortical interneuron function and survival
    Daniel Pensold, Geraldine Zimmer-Bensch
    2021, 16 (12):  2405-2406.  doi: 10.4103/1673-5374.313037
    Abstract ( 130 )   PDF (567KB) ( 144 )   Save
    Increased occurrence of age-associated disabilities and neurodegenerative diseases is the price we pay for the tremendous elevation in life expectancy in our modern society. Aging comes along with structural, neurochemical and physiological alterations in the brain that cause memory decline and cognitive impairments (Rozycka and Liguz-Lecznar, 2017). Numerous factors contribute to cognitive aging including hormonal, metabolic, and immune dysregulation, elevated oxidative stress and inflammation, changes in neurotransmission, and diminished neurotrophic support of neurons (Rozycka and Liguz-Lecznar, 2017). Thereby, different brain regions and neuronal cell types are distinctively affected by the process of aging. Apart from reduced excitability and plasticity, the decline in inhibitory function represents a prominent feature of aged brains (Zimmer-Bensch, 2019a). A selective vulnerability of inhibitory interneurons and GABAergic (gamma-aminobutyric acid) synapses is reported for diverse regions of the aged brain across different species. This is reflected by reduced numbers of inhibitory cortical interneuron subtypes, as well as by functional and structural changes of GABAergic synapses (Rozycka and Liguz-Lecznar, 2017; Zimmer-Bensch, 2019a).
    Related Articles | Metrics
    BACE1 role in Alzheimer’s disease and other dementias: from the theory to the practice
    Carlo Cervellati, Giuseppe Valacchi, Giovanni Zuliani
    2021, 16 (12):  2407-2408.  doi: 10.4103/1673-5374.313041
    Abstract ( 216 )   PDF (474KB) ( 151 )   Save
    AD is a multifactorial and multifaceted disease, with a complex and still not completely understood pathogenesis (Cervellati et al., 2016; Iturria-Medina et al., 2016). Multiple hypotheses have been proposed to explain the pathobiology of the disease. In this plethora of mechanistic models, the central role amyloid beta (Aβ) forming neuritic plaques remains undiscussed. However, the adjective “central” does not mean that Aβ alone can trigger and fuel all the AD neurodegenerative process. Indeed, it is now amply recognized that a combination of multiple biochemical (especially redox), immune system and vascular abnormalities must also occur to define the direction of disease trajectory (Iturria-Medina et al., 2016). This complexity is probably one of the main reasons for the current unavailability of disease-modifying therapies that may prevent or slow the rate of disease progression. 
    Related Articles | Metrics
    Open late: neuronal membrane disruption late in traumatic brain injury
    Martina L. Hernandez, MiJin Cho, Audrey D. Lafrenaye
    2021, 16 (12):  2409-2410.  doi: 10.4103/1673-5374.313029
    Abstract ( 152 )   PDF (279KB) ( 187 )   Save
    Membrane disruption is a pathology in which the cellular phospholipid bilayer, which is the cell’s primary defense from the extracellular environment, is compromised. Some neurons that are impacted by membrane disruption experience cell death, and others demonstrate delayed disruption, providing an opportunity for therapeutic intervention. This pathology is visualized using infusions of cell impermeable tracers, such as dextran, calcein and propidium iodide followed by microscopic analysis (Geddes et al., 2003a, b; Singleton and Povlishock, 2004; Farkas et al., 2006; Whalen et al., 2008; Cullen et al., 2011; Lafrenaye et al., 2014; Levine et al., 2016; Hernandez et al., 2019; LaPlaca et al., 2019; Prado and LaPlaca, 2020), and therefore little is known about membrane disruption clinically. However, membrane disruption has been well documented to occur in various pre-clinical models of traumatic brain injury (TBI).
    Related Articles | Metrics
    Neurovascular ageing: transcriptomic readout and implications on therapeutic targeting in Alzheimer’s disease
    Zhongqi Li, Bonaventure Ip, Vincent C. T. Mok, Ho Ko
    2021, 16 (12):  2411-2412.  doi: 10.4103/1673-5374.313042
    Abstract ( 136 )   PDF (438KB) ( 143 )   Save
    Related Articles | Metrics
    The antiaging role of oxytocin
    Tarek Benameur, Maria A. Panaro, Chiara Porro
    2021, 16 (12):  2413-2414.  doi: 10.4103/1673-5374.313030
    Abstract ( 276 )   PDF (526KB) ( 213 )   Save
    Related Articles | Metrics
    Rogue gene networks gone awry in Alzheimer’s disease
    Emily Miyoshi, Vivek Swarup
    2021, 16 (12):  2415-2416.  doi: 10.4103/1673-5374.313032
    Abstract ( 122 )   PDF (240KB) ( 156 )   Save
    The human brain consists of billions of cells encompassing hundreds of distinct cell-types, each with unique functions and properties. Identification of the molecular architecture of the brain has been revolutionized by next-generation sequencing (NGS), as evident by recent transcriptomic and genetic/epigenetic studies. NGS paved the way to perform large-scale, genome-wide sequencing studies on human postmortem brain tissue, and this offered an unprecedented opportunity to elucidate the genetic bases of polygenic neurological disorders, like Alzheimer’s disease (AD).
    Related Articles | Metrics
    A long story for a short peptide: therapeutic efficacy of a cleavage-specific tau antibody
    Giuseppina Amadoro, Valentina Latina, Pietro Calissano
    2021, 16 (12):  2417-2419.  doi: 10.4103/1673-5374.313043
    Abstract ( 142 )   PDF (590KB) ( 141 )   Save
    AD, the main cause of dementia in elderly people, is a multifactorial neurodegenerative disorder characterized by a long prodromal phase (starting more than two decades before clinical symptoms appear) with brain accumulation/misfolding of amyloid β (Aβ) in insoluble amyloid plaques and of tau protein in neurofibrillary tangles. Even though the slow-progressing clinical development of the disease opens important diagnostic and therapeutic perspectives for the preventive medicine, there’s a general consensus that the amyloid deposition reaches early a plateau and does not change over time. On the contrary, the tau pathology is tightly linked with synaptic deterioration and neuronal death which eventually lead to the manifestation of classical symptomatology (Jack et al., 2018). Indeed, several lines of evidence support the notion that alterations of tau homeostasis actually drive the neurodegeneration in human tauopathies, including the most common AD where no genetic mutation in microtubule associated protein tau (MAPT) has been reported up to now. To this regard genetic, clinical and histopathological studies have undoutebly shown that abnormalities in tau protein(s) are sufficient to cause, both in vitro and in vivo, synaptic dysfunction, motor/sensorimotor and cognitive deficits indicative of loss of selective vulnerable neuronal populations (Spillantini and Goedert, 2013). Secondly, compelling evidence have also demonstrated that tau proteins have necessary, not-dispensable role in Aβ-dependent neurodegeneration, both in cellular and animal AD models and in elderly individuals (Iqbal and Gong, 2016). Moreover, both the “amyloid-cascade hypothesis” (i.e. Aβ is the initial insult driving tau pathology) and the “dual hit-model” (i.e. tau pathology is independent of Aβ which just provokes the tau spread to neocortex), which have been proposed to explain the etiopathogenesis of AD, posit a crucial place (i.e. induttive and/or permissive) of tau pathobiology in the chains of events ending in synaptic derangement and irreversible loss of neuronal viability (Small and Duff, 2008). Consistently, tau pathology correlates much more strongly than Aβ pathology with neurodegeneration and cognitive impairment, both spatially and temporally. In fact, the amyloid deposition does not cause the pronounced synaptic and neuronal loss which typically characterize the progressive clinical course of AD. Finally, tau is a common downstream effector both in Aβ-dependent and -independent pathogenic mechanisms such as increased amyloid precursor protein (APP) gene dosage/APP-derived-C-Terminal fragment (CTF,C99), cholesterol metabolism/endocytic, trafficking microglial immune activation, apoliprotein E allele epsilon 4.
    Related Articles | Metrics
    A pilot exploration with Posiphen to normalize amyloid precursor protein in Down syndrome
    Xu-Qiao Chen
    2021, 16 (12):  2420-2421.  doi: 10.4103/1673-5374.313044
    Abstract ( 152 )   PDF (501KB) ( 868 )   Save
    DS is the most common cause of Alzheimer’s disease (AD) and has a known AD-causing genetic variation which is trisomy of the whole or part of Homo sapiens chromosome 21 (HSA21) (Chen and Mobley, 2019a). Compared with AD, DS usually present with various symptoms and manifestations which are related to dysfunction of multiple body systems; the extra copies of the many genes present in HSA21 in DS can cause various developmental problems including neurodevelopmental deficits, which underlie the physical features in DS and may also contribute to the AD-related neurological symptoms of DS like cognition decline (Antonarakis et al., 2020). However, DS and AD share many pathological hallmarks including amyloid plaques and neurofibrillary tangles, synaptic and neuronal loss, dysregulation of endosomal pathway and others (Chen and Mobley, 2019a). The plaques and tangles have been studied extensively, while the dysfunction of early endosome has been found to be impacted long before appearance of amyloid deposits and neurofibrillary tangles with amyloid beta (Aβ) of varing length and phosphorylated tau as the main components, respectively (Nixon, 2017; Chen and Mobley, 2019a). Deficient endosome-mediated retrograde axonal transport of neurotrophic signals plays an important role in the neuropathogenesis in both DS and AD as continuing neurotrophic support is required for maintenance of mature neurons including the basal forebrain cholinergic neurons (BFCNs) whose degeneration has been linked to age-related cognitive dysfunction in DS and AD (Chen and Mobley, 2019b). HSA21 contains about 233 protein-coding genes with several encoding protein products demonstrated to contribute to different phenotypes in DS (Antonarakis et al., 2020) from the extensive researches in the different mouse models of DS, including Ts65Dn mouse which are segmentally trisomic for orthologs of about half of the protein coding genes located on HSA21 (Antonarakis et al., 2020). Accumulated evidence have attributed AD pathogenesis to toxic oligomeric Aβ and tau (Chen and Mobley, 2019a), however, recent clinical trials intending to target them in AD have yet to demonstrate considerable efficacy. Importantly, the 99 amino acid C-terminal fragment (β-CTF) of amyloid precursor protein (APP) was recently found to dysregulate endosomal and lysosomal systems as well as induce cholinergic neurodegeneration in an Aβ-independent manner (Xu et al., 2016; Nixon, 2017; Chen and Mobley, 2019b). Thus, reviewing the hypotheses for AD and discovering novel targets will be beneficial to combat AD and AD in DS.

    Related Articles | Metrics
    Multiple sclerosis: why we should focus on both sides of the (auto)antibody
    Jeroen den Dunnen, Lynn Mes, Willianne Hoepel, Joost Smolders
    2021, 16 (12):  2422-2424.  doi: 10.4103/1673-5374.313045
    Abstract ( 177 )   PDF (587KB) ( 150 )   Save
    Various clinical and experimental findings suggest a pathogenic role of antibodies in multiple sclerosis (MS). Yet, whether antibodies contribute to the pathogenesis or progression of MS is still a subject of intense debate. This controversy particularly results from unclarity regarding the target antigens of the antibodies that are found in the central nervous system (CNS) of MS patients. The identification of such target antigen(s) at disease onset remains an important topic of investigation, but these antigens may be heterogeneous and not the decisive factor for the initiation of MS development. In addition to antigen-specific binding of IgG, IgG may also promote pathology in MS patients by binding in an antigen non-specific manner. Therefore, we propose that we should not only focus on the antigen-binding part of MS antibodies, but also should pay attention to the other side of the antibodies in the CNS of MS patients, i.e. the fragment crystallizable (Fc) tail (Figure 1A). The characteristics of the Fc tail, particularly the (combination of) IgG subclass, allotype, and glycosylation determine the pathogenicity of IgG, but these characteristics are still poorly defined in MS. Unraveling these characteristics may not only lead to better understanding of MS pathogenesis, but may also yield new strategies for therapy.
    Related Articles | Metrics
    Advance in the use of gold nanoparticles in the treatment of neurodegenerative diseases: new perspectives
    Gustavo de Bem Silveira, Alexandre Pastoris Muller, Ricardo Andrez Machado-de-Ávila, Paulo Cesar Lock Silveira
    2021, 16 (12):  2425-2426.  doi: 10.4103/1673-5374.313040
    Abstract ( 310 )   PDF (528KB) ( 187 )   Save
    Various conditions affecting nerve cells and the nervous system due to the loss of neurons and their connecting networks are described under the superordinate phrase “Neurodegenerative diseases”. Such diseases lead to disability due to gradual neuronal death in both the central nervous system and the peripheral nervous system. While many of these diseases have unknown causes, sometimes these are due to medical conditions such as alcoholism, a tumor, or a stroke, or other causes which may include genetic mutations, toxins, chemicals, and viruses. Neurodegenerative diseases can be induced by various neurotoxic events, such as excessive inflammation, reactive oxygen species (ROS) production, and mitochondrial dysfunctions. The main symptoms associated with these disorders are related to movement (ataxia), mental functioning (dementia), or both, causing morbidity and death, thus having social and economic implications. The available treatments for these disorders provide only symptomatic relief, such as extending the lifespan to a few years. Still, a lot of research is in progress to find therapeutic markers for such diseases, as the complexity of the pathophysiology of neurodegenerative diseases and the underlying cell interactions is often imperfectly understood, limiting the development of therapeutic approaches (Khan et al., 2020). These physical constraints and the lack of specificity of current pharmacological approaches explain why most drugs and neurosurgical procedures have not been effective in the treatment of central nervous system disorders. As examples, pharmaceutical drugs used as treatment for Alzheimer’s disease have low effects and those for Parkinson’s disease loses their activity in a few years.
    Related Articles | Metrics
    Multiple hurdle mechanism and blood-brain barrier in epilepsy: glucocorticoid receptor-heat shock proteins on drug regulation
    Aneesha Achar, Chaitali Ghosh
    2021, 16 (12):  2427-2428.  doi: 10.4103/1673-5374.313046
    Abstract ( 361 )   PDF (428KB) ( 178 )   Save
    Epilepsy is a complex neurologic condition which affects over 50 million people worldwide. Pharmacotherapy, primarily involving the use of anti-seizure drugs (ASDs), is an essential part of controlling seizures. However, nearly 30% of patients develop drug-resistant epilepsy, clinically defined as the persistence of seizure following trials of two ASDs (Kwan et al., 2010). Although several hypotheses have been proposed to explain this phenomenon, the mechanism of drug-resistant epilepsy still remains unclear. However, a growing body of evidence has demonstrated that blood-brain barrier (BBB) dysfunction represents an important hallmark of the epileptic brain (Salar et al., 2014; Gorter et al., 2019). As previously reported, initial brain injury or seizure may trigger disruption of the BBB, resulting in the immediate release of glutamate. Excess glutamate results in cell stress, inflammatory and cell adhesion molecule activation, and leukocyte infiltration into the brain. Finally, neuronal death, rewiring, gliosis, neurogenesis and angiogenesis, and upregulation/downregulation of receptors, transporters, and ion channels may take place within weeks to months of initial injury. These structural and functional BBB changes may trigger further risk of future seizures and anomalies (Gorter et al., 2019). Evidently, the association between drug-resistant epilepsy and impairment of the BBB function cannot be ignored.
    Related Articles | Metrics
    Spinal cord injury: can we repair spinal cord non-invasively by using magnetic stimulation?
    Quentin Delarue, Chaima Chalfouh, Nicolas Guérout
    2021, 16 (12):  2429-2430.  doi: 10.4103/1673-5374.313033
    Abstract ( 163 )   PDF (696KB) ( 121 )   Save
    Spinal cord injury (SCI) is currently an incurable condition which induces sensorimotor impairments below the injury level. Mainly, SCI are the consequence of physical damages which occur on spinal cord due to traffic accidents or sports and recreation injuries. To date, nor treatment of therapy could be proposed to patients with SCI (Wilson et al., 2012).
    Related Articles | Metrics
    A method for using video presentation to increase the vividness and activity of cortical regions during motor imagery tasks
    Kengo Fujiwara, Masatomo Shibata, Yoshinaga Awano, Koji Shibayama, Naoki Iso, Moemi Matsuo, Akira Nakashima, Takefumi Moriuchi, Wataru Mitsunaga, Toshio Higashi
    2021, 16 (12):  2431-2437.  doi: 10.4103/1673-5374.313058
    Abstract ( 147 )   PDF (1341KB) ( 222 )   Save
    In recent years, mental practice (MP) using laterally inverted video of a subject’s non-paralyzed upper limb to improve the vividness of presented motor imagery (MI) has been shown to be effective for improving the function of a paralyzed upper limb. However, no studies have yet assessed the activity of cortical regions engaged during MI task performance using inverse video presentations and neurophysiological indicators. This study sought to investigate changes in MI vividness and hemodynamic changes in the cerebral cortex during MI performance under the following three conditions in near-infrared spectroscopy: MI-only without inverse video presentation (MI-only), MI with action observation (AO) of an inverse video presentation of another person’s hand (AO + MI (other hand)), and MI with AO of an inverse video presentation of a participant’s own hand (AO + MI (own hand)). Participants included 66 healthy right-handed adults (41 men and 25 women; mean age: 26.3 ± 4.3 years). There were 23 patients in the MI-only group (mean age: 26.4 ± 4.1 years), 20 in the AO + MI (other hand) group (mean age: 25.9 ± 5.0 years), and 23 in the AO + MI (own hand) group (mean age: 26.9 ± 4.1 years). The MI task involved transferring 1 cm × 1 cm blocks from one plate to another, once per second, using chopsticks held in the non-dominant hand. Based on a visual analog scale (VAS), MI vividness was significantly higher in the AO + MI (own hand) group than in the MI-only group and the AO + MI (other hand) group. A main effect of condition was revealed in terms of MI vividness, as well as regions of interest (ROIs) in certain brain areas associated with motor processing. The data suggest that inverse video presentation of a person’s own hand enhances the MI vividness and increases the activity of motor-related cortical areas during MI. This study was approved by the Institutional Ethics Committee of Nagasaki University Graduate School of Biomedical and Health Sciences (approval No. 18121303) on January 18, 2019.
    Related Articles | Metrics
    Protein profiling identified mitochondrial dysfunction and synaptic abnormalities after dexamethasone intervention in rats with traumatic brain injury
    Fei Niu, Bin Zhang, Jie Feng, Xiang Mao, Xiao-Jian Xu, Jin-Qian Dong, Bai-Yun Liu
    2021, 16 (12):  2438-2445.  doi: 10.4103/1673-5374.313047
    Abstract ( 185 )   PDF (1505KB) ( 133 )   Save
    Dexamethasone has been widely used after various neurosurgical procedures due to its anti-inflammatory property and the abilities to restore vascular permeability, inhibit free radicals, and reduce cerebrospinal fluid production. According to the latest guidelines for the treatment of traumatic brain injury in the United States, high-dose glucocorticoids cause neurological damage. To investigate the reason why high-dose glucocorticoids after traumatic brain injury exhibit harmful effect, rat controlled cortical impact models of traumatic brain injury were established. At 1 hour and 2 days after surgery, rat models were intraperitoneally administered dexamethasone 10 mg/kg. The results revealed that 31 proteins were significantly upregulated and 12 proteins were significantly downregulated in rat models of traumatic brain injury after dexamethasone treatment. The Ingenuity Pathway Analysis results showed that differentially expressed proteins were enriched in the mitochondrial dysfunction pathway and synaptogenesis signaling pathway. Western blot analysis and immunohistochemistry results showed that Ndufv2, Maob and Gria3 expression and positive cell count in the dexamethasone-treated group were significantly greater than those in the model group. These findings suggest that dexamethasone may promote a compensatory increase in complex I subunits (Ndufs2 and Ndufv2), increase the expression of mitochondrial enzyme Maob, and upregulate synaptic-transmission-related protein Gria3. These changes may be caused by nerve injury after traumatic brain injury treatment by dexamethasone. The study was approved by Institutional Ethics Committee of Beijing Neurosurgical Institute (approval No. 201802001) on June 6, 2018.
    Related Articles | Metrics
    Differential development and electrophysiological activity in cultured cortical neurons from the mouse and cynomolgus monkey
    Xue-Yan Zhang, Jun Li, Cai-Juan Li, Ying-Qi Lin, Chun-Hui Huang, Xiao Zheng, Xi-Chen Song, Zhu-Chi Tu, Xiao-Jiang Li, Sen Yan
    2021, 16 (12):  2446-2452.  doi: 10.4103/1673-5374.313056
    Abstract ( 163 )   PDF (1754KB) ( 206 )   Save
    In vitro cultures of primary cortical neurons are widely used to investigate neuronal function. However, it has yet to be fully investigated whether there are significant differences in development and function between cultured rodent and primate cortical neurons, and whether these differences influence the utilization of cultured cortical neurons to model pathological conditions. Using in vitro culture techniques combined with immunofluorescence and electrophysiological methods, our study found that the development and maturation of primary cerebral cortical neurons from cynomolgus monkeys were slower than those from mice. We used a microelectrode array technique to compare the electrophysiological differences in cortical neurons, and found that primary cortical neurons from the mouse brain began to show electrical activity earlier than those from the cynomolgus monkey. Although cultured monkey cortical neurons developed slowly in vitro, they exhibited typical pathological features-revealed by immunofluorescent staining-when infected with adeno-associated viral vectors expressing mutant huntingtin (HTT), the Huntington’s disease protein. A quantitative analysis of the cultured monkey cortical neurons also confirmed that mutant HTT significantly reduced the length of neurites. Therefore, compared with the primary cortical neurons of mice, cultured monkey cortical neurons have longer developmental and survival times and greater sustained physiological activity, such as electrophysiological activity. Our findings also suggest that primary cynomolgus monkey neurons cultured in vitro can simulate a cell model of human neurodegenerative disease, and may be useful for investigating time-dependent neuronal death as well as treatment via neuronal regeneration. All mouse experiments and protocols were approved by the Animal Care and Use Committee of Jinan University of China (IACUC Approval No. 20200512-04) on May 12, 2020. All monkey experiments were approved by the IACUC protocol (IACUC Approval No. LDACU 20190820-01) on August 23, 2019 for animal management and use.
    Related Articles | Metrics
    SOCS1/JAK2/STAT3 axis regulates early brain injury induced by subarachnoid hemorrhage via inflammatory responses
    Yang Wang, Xiang-Qian Kong, Fei Wu, Bin Xu, De-Jun Bao, Chuan-Dong Cheng, Xiang-Ping Wei, Yong-Fei Dong, Chao-Shi Niu
    2021, 16 (12):  2453-2464.  doi: 10.4103/1673-5374.313049
    Abstract ( 205 )   PDF (9276KB) ( 91 )   Save
    The SOCS1/JAK2/STAT3 axis is strongly associated with tumor growth and progression, and participates in cytokine secretion in many diseases. However, the effects of the SOCS1/JAK2/STAT3 axis in experimental subarachnoid hemorrhage remain to be studied. A subarachnoid hemorrhage model was established in rats by infusing autologous blood into the optic chiasm pool. Some rats were first treated with JAK2/STAT3 small interfering RNA (Si-JAK2/Si-STAT3) or overexpression plasmids of JAK2/STAT3. In the brains of subarachnoid hemorrhage model rats, the expression levels of both JAK2 and STAT3 were upregulated and the expression of SOCS1 was downregulated, reaching a peak at 48 hours after injury. Simultaneously, the interactions between JAK2 and SOCS1 were reduced. In contrast, the interactions between JAK2 and STAT3 were markedly enhanced. Si-JAK2 and Si-STAT3 treatment alleviated cortical neuronal cell apoptosis and necrosis, destruction of the blood–brain barrier, brain edema, and cognitive functional impairment after subarachnoid hemorrhage. This was accompanied by decreased phosphorylation of JAK2 and STAT3 protein, decreased total levels of JAK2 and STAT3 protein, and increased SOCS1 protein expression. However, overexpression of JAK2 and STAT3 exerted opposite effects, aggravating subarachnoid hemorrhage-induced early brain injury. Si-JAK2 and Si-STAT3 inhibited M1-type microglial conversion and the release of pro-inflammatory factors (inducible nitric oxide synthase, interleukin-1β, and tumor necrosis factor-α) and increased the release of anti-inflammatory factors (arginase-1, interleukin-10, and interleukin-4). Furthermore, primary neurons stimulated with oxyhemoglobin were used to simulate subarachnoid hemorrhage in vitro, and the JAK2 inhibitor AG490 was used as an intervention. The in vitro results also suggested that neuronal protection is mediated by the inhibition of JAK2 and STAT3 expression. Together, our findings indicate that the SOCS1/JAK2/STAT3 axis contributes to early brain injury after subarachnoid hemorrhage both in vitro and in vivo by inducing inflammatory responses. This study was approved by the Animal Ethics Committee of Anhui Medical University and the First Affiliated Hospital of University of Science and Technology of China (approval No. LLSC-20180202) on March 1, 2018.
    Related Articles | Metrics
    Hippocampal insulin resistance and the Sirtuin 1 signaling pathway in diabetes-induced cognitive dysfunction
    Hui Yang, Lin Tang, Zhan Qu, Shi-Hui Lei, Wei Li, Yu-Hong Wang
    2021, 16 (12):  2465-2474.  doi: 10.4103/1673-5374.313051
    Abstract ( 144 )   PDF (3751KB) ( 458 )   Save
    In the peripheral nervous system, the activation of Sirtuin 1 can improve insulin resistance; however, the role played by Sirtuin 1 in the central nervous system remains unknown. In this study, rat models of diabetes mellitus were generated by a single injection of streptozotocin. At 8 weeks after streptozotocin injection, the Morris water maze test and western blot assays confirmed that the diabetic model rats had learning and memory deficits, insulin resistance, and Sirtuin 1 expression could be detected in the hippocampus. Insulin and the insulin receptor inhibitor S961 were intranasally administered to investigate the regulatory effects of insulin signaling on Sirtuin 1. The results showed that insulin administration improved the impaired cognitive function of diabetic model rats and increased the expression levels of phosphorylated insulin receptor, phosphorylated insulin receptor substrate 1, and Sirtuin 1 in the hippocampus. Conversely, S961 administration resulted in more severe cognitive dysfunction and reduced the expression levels of phosphorylated insulin receptor, phosphorylated insulin receptor substrate 1, and Sirtuin 1. The Sirtuin 1 activator SRT2104 and the inhibitor Sirtinol were injected into the lateral ventricle, which revealed that the activation of Sirtuin 1 increased the expression levels of target of rapamycin complex 1, phosphorylated cAMP-response element-binding protein, and brain-derived neurotrophic factor. Hippocampal dendritic length and spine density also increased in response to Sirtuin 1 activation. In contrast, Sirtinol decreased the expression levels of target of rapamycin complex 1, phosphorylated cAMP-response element-binding protein, and brain-derived neurotrophic factor and damaged the dendritic structure. These findings suggest that the Sirtuin 1 signaling pathway plays an important role in the development of insulin resistance-related cognitive deficits in diabetic rats. This study was approved by the Animal Ethics Welfare Committee of the First Affiliated Hospital of Hunan University of Chinese Medicine (approval No. ZYFY201811207) in November 2018. 
    Related Articles | Metrics
    Potential protective role of ACE-inhibitors and AT1 receptor blockers against levodopa-induced dyskinesias: a retrospective case-control study
    Elena Contaldi, Luca Magistrelli, Anna V. Milner, Marco Cosentino, Franca Marino, Cristoforo Comi
    2021, 16 (12):  2475-2478.  doi: 10.4103/1673-5374.313061
    Abstract ( 129 )   PDF (424KB) ( 184 )   Save
    Growing evidence has highlighted that angiotensin-converting enzyme (ACE)-inhibitors (ACEi)/AT1 receptor blockers (ARBs) may influence the complex interplay between dopamine and the renin-angiotensin system in the nigrostriatal pathway, thus affecting the development of levodopa-induced dyskinesia in Parkinson’s disease (PD). In the present study, we analyzed whether the use of this class of medication was associated with a reduced occurrence of levodopa-induced dyskinesia, using electronically-stored information of idiopathic PD patients enrolled at Novara University Hospital “Maggiore della Carità”. We conducted a retrospective case-control study identifying PD patients with dyskinesias (PwD; n = 47) as cases. For each PwD we selected a non-dyskinetic control (NoD), nearly perfectly matched according to sex, Unified Parkinson’s Disease Rating Scale (UPDRS) part III score, and duration of antiparkinsonian treatment. Binary logistic regression was used to evaluate whether dyskinesias were associated with ACEi/ARBs use. Ninety-four PD patients were included, aged 72.18 ± 9 years, with an average disease duration of 10.20 ± 4.8 years and 9.04 ± 4.9 years of antiparkinsonian treatment. The mean UPDRS part III score was 18.87 ± 7.6 and the median HY stage was 2. In the NoD group, 25 (53.2%) were users and 22 (46.8%) non-users of ACEi/ARBs. Conversely, in the PwD group, 11 (23.4%) were users and 36 non-users (76.6%) of this drug class (Pearson chi-square = 8.824, P = 0.003). Concerning general medication, there were no other statistically significant differences between groups. After controlling for tremor dominant phenotype, levodopa equivalent daily dose, HY 3-4, and disease duration, ACEi/ARBs use was a significant predictor of a lower occurrence of dyskinesia (OR = 0.226, 95% CI: 0.080–0.636, P = 0.005). Therefore, our study suggests that ACEi/ARBs may reduce levodopa-induced dyskinesia occurrence and, thanks to good tolerability and easy management, represent a feasible choice when dealing with the treatment of hypertension in PD patients. The study was approved by the Ethics Committee of Novara University Hospital “Maggiore della Carità” (CE 65/16) on July 27, 2016.
    Related Articles | Metrics
    Effects of a multimodal exercise protocol on functional outcomes, epigenetic modulation and brain-derived neurotrophic factor levels in institutionalized older adults: a quasi-experimental pilot study
    Iasmin Fraga, Camila Weber, Wériton Baldo Galiano, Lucio Iraci, Mariana Wohlgemuth, Gabriela Morales, Camila Cercato, Juliana Rodriguez, Daniela Pochmann, Caroline Dani, Pérsia Menz, Adriane Dal Bosco, Viviane Rostirola Elsner
    2021, 16 (12):  2479-2485.  doi: 10.4103/1673-5374.313067
    Abstract ( 108 )   PDF (522KB) ( 158 )   Save
    Epigenetic changes have been shown to be associated with both aging process and aging-related diseases. There is evidence regarding the benefits of physical activity on the functionality, cognition, and quality of life of institutionalized older adults, however, the molecular mechanisms involved are not elucidated. The purpose of this pilot study was to investigate the effects of a multimodal exercise intervention on functional outcomes, cognitive performance, quality of life (QOL), epigenetic markers and brain-derived neurotrophic factor (BDNF) levels among institutionalized older adult individuals. Participants (n = 8) without dementia who were aged 73.38 ± 11.28 years and predominantly female (87.5%) were included in this quasi-experimental pilot study. A multimodal exercise protocol (cardiovascular capacity, strength, balance/agility and flexibility, perception and cognition) consisted of twice weekly sessions (60 minutes each) over 8 weeks. Balance (Berg Scale), mobility (Timed Up and Go test), functional capacity (Six-Minute Walk test), cognitive function (Mini-Mental State Examination) and QOL (the World Health Organization Quality of Life-BREF Scale questionnaire) were evaluated before and after the intervention. Blood sample (15 mL) was also collected before and after intervention for analysis of biomarkers global histone H3 acetylation and brain-derived neurotrophic factor levels. Significant improvements were observed in cognitive function, balance, mobility, functional capacity and QOL after the intervention. In addition, a tendency toward an increase in global histone H3 acetylation levels was observed, while brain-derived neurotrophic factor level remained unchanged. This study provided evidence that an 8-week multimodal exercise protocol has a significant effect on ameliorating functional outcomes and QOL in institutionalized older adult individuals. In addition, it was also able to promote cognitive improvement, which seems to be partially related to histone hyperacetylation status. The Ethics Research Committee of Centro Universitário Metodista-IPA, Brazil approved the current study on June 6, 2019 (approval No. 3.376.078). 
    Related Articles | Metrics
    Skeletal muscle stiffness as measured by magnetic resonance elastography after chronic spinal cord injury: a cross-sectional pilot study
    Mina P. Ghatas, M. Rehan Khan, Ashraf S. Gorgey
    2021, 16 (12):  2486-2493.  doi: 10.4103/1673-5374.313060
    Abstract ( 141 )   PDF (669KB) ( 145 )   Save
    Skeletal muscle stiffness is altered after spinal cord injury (SCI). Assessing muscle stiffness is essential for rehabilitation and pharmaceutical interventions design after SCI. The study used magnetic resonance elastography to assess the changes in stiffness after chronic SCI compared to matched able-bodied controls and determine its association with muscle size, spasticity, and peak torque in persons with SCI. Previous studies examined the association between muscle stiffness and spasticity, however, we are unaware of other studies that examined the effects of muscle composition on stiffness after SCI. Ten participants (one female) with chronic SCI and eight (one female) matched able-bodied controls participated in this cross-sectional study. Magnetic resonance elastography was utilized to monitor stiffness derived from shear waves propagation. Modified Ashworth scale was used to evaluate spasticity scores in a blinded fashion. Peak isometric and isokinetic torques were measured using a biodex dynamometer. Stiffness values were non-significantly lower (12.5%; P = 0.3) in the SCI group compared to able-bodied controls.  Moreover, stiffness was positively related to vastus lateralis whole muscle cross-sectional area (CSA) (r2 = 0.64, P < 0.005) and vastus lateralis absolute muscle CSA after accounting for intramuscular fat (r2 = 0.78, P < 0.0007). Stiffness was also positively correlated to both isometric (r2= 0.55–0.57, P < 0.05) and isokinetic peak (r2= 0.46–0.48, P < 0.05) torques. Our results suggest that larger clinical trial is warranted to confirm the preliminary findings that muscle stiffness is altered after SCI compared to healthy controls. Stiffness appeared to be influenced by infiltration of intramuscular fat and modestly by the spasticity of the paralyzed muscles. The preliminary data indicated that the relationship between muscle stiffness and peak torque is not altered with changing the frequency of pulses or angular velocities. All study procedures were approved by the Institutional Review Board at the Hunter Holmes McGuire VA Medical Center, USA (IRB #: 02314) on May 3, 2017.

    Related Articles | Metrics
    11C-PK11195 plasma metabolization has the same rate in multiple sclerosis patients and healthy controls: a cross-sectional study
    Aline Morais de Souza, Milena Sales Pitombeira, Larissa Estessi de Souza, Fabio Luiz Navarro Marques, Carlos Alberto Buchpiguel, Caroline Cristiano Real, Daniele de Paula Faria
    2021, 16 (12):  2494-2498.  doi: 10.4103/1673-5374.313062
    Abstract ( 145 )   PDF (747KB) ( 138 )   Save
    11C-PK11195 is a positron emitter tracer used for Positron Emission Tomography (PET) imaging of innate immune cell activation in studies of neuroinflammatory diseases. For the image quantitative analysis, it is necessary to quantify the intact fraction of this tracer in the arterial plasma during imaging acquisition (plasma intact fraction). Due to the complexity and costs involved in this analysis it is important to evaluate the real necessity of individual analysis in each 11C-PK11195 PET imaging acquisition. The purpose of this study is to compare 11C-PK11195 plasma metabolization rate between healthy controls and multiple sclerosis (MS) patients and evaluate the interference of sex, age, treatment, and disease phenotype in the tracer intact fraction measured in arterial plasma samples. 11C-PK11195 metabolization rate in arterial plasma was quantified by high performance liquid chromatography in samples from MS patients (n = 50) and healthy controls (n = 23) at 20, 45, and 60 minutes after 11C-PK11195 injection. Analyses were also stratified by sex, age, treatment type, and MS phenotype. The results showed no significant differences in the metabolization rate of healthy controls and MS patients, or in the stratified samples. In conclusion, 11C-PK11195 metabolization has the same rate in patients with MS and healthy controls, which is not affected by sex, age, treatment, and disease phenotype. Thus, these findings could contribute to exempting the necessity for tracer metabolization determination in all 11C-PK11195 PET imaging acquisition, by using a population metabolization rate average. The study procedures were approved by the Ethics Committee for Research Projects Analysis of the Hospital das Clinicas of the University of Sao Paulo Medical School (approval No. 624.065) on April 23, 2014.
    Related Articles | Metrics
    Rutin pretreatment promotes microglial M1 to M2 phenotype polarization
    Guang-Ping Lang, Can Li, Ying-Ying Han
    2021, 16 (12):  2499-2504.  doi: 10.4103/1673-5374.313050
    Abstract ( 579 )   PDF (1814KB) ( 318 )   Save
    Microglial cells are important resident innate immune components in the central nervous system that are often activated during neuroinflammation. Activated microglia can display one of two phenotypes, M1 or M2, which each play distinct roles in neuroinflammation. Rutin, a dietary flavonoid, exhibits protective effects against neuroinflammation. However, whether rutin is able to influence the M1/M2 polarization of microglia remains unclear. In this study, in vitro BV-2 cell models of neuroinflammation were established using 100 ng/mL lipopolysaccharide to investigate the effects of 1-hour rutin pretreatment on microglial polarization. The results revealed that rutin pretreatment reduced the expression of the proinflammatory cytokines tumor necrosis factor-α, interleukin-1β, and interleukin-6 and increased the secretion of interleukin-10. Rutin pretreatment also downregulated the expression of the M1 microglial markers CD86 and inducible nitric oxide synthase and upregulated the expression of the M2 microglial markers arginase 1 and CD206. Rutin pretreatment inhibited the expression of Toll-like receptor 4 and myeloid differentiation factor 88 and blocked the phosphorylation of I kappa B kinase and nuclear factor-kappa B. These results showed that rutin pretreatment may promote the phenotypic switch of microglia M1 to M2 by inhibiting the Toll-like receptor 4/nuclear factor-kappa B signaling pathway to alleviate lipopolysaccharide-induced neuroinflammation. 
    Related Articles | Metrics
    P2X4 receptor participates in autophagy regulation in Parkinson’s disease
    Xue Zhang, Jing Wang, Jin-Zhao Gao, Xiao-Na Zhang, Kai-Xin Dou, Wan-Da Shi, An-Mu Xie
    2021, 16 (12):  2505-2511.  doi: 10.4103/1673-5374.313053
    Abstract ( 161 )   PDF (1323KB) ( 138 )   Save
    Dysfunctional autophagy often occurs during the development of neurodegenerative diseases, such as Parkinson’s disease, Huntington’s disease, and Alzheimer’s disease. The purinergic P2X4 receptor is an ATP-gated ion channel that is widely expressed in the microglia, astrocytes, and neurons of the central and peripheral nervous systems. P2X4R is involved in the regulation of cellular excitability, synaptic transmission, and neuroinflammation. However, the role played by P2X4R in Parkinson’s disease remains poorly understood. Rat models of Parkinson’s disease were established by injecting 6-hydroxydopamine into the substantia nigra pars compacta. P2X4R-targeted small interfering RNA (siRNA) was injected into the same area 1 week before injury induction to inhibit the expression of the P2X4 receptor. The results showed that the inhibition of P2X4 receptor expression in Parkinson’s disease model rats reduced the rotation behavior induced by apomorphine treatment, increased the latency on the rotarod test, and upregulated the expression of tyrosine hydroxylase, brain-derived neurotrophic factor, LC3-II/LC3-I, Beclin-1, and phosphorylated tropomyosin receptor kinase B (TrkB) in brain tissue, while simultaneously reducing p62 levels. These findings suggest that P2X4 receptor activation might inhibit neuronal autophagy through the regulation of the brain-derived neurotrophic factor/TrkB signaling pathway, leading to dopaminergic neuron damage in the substantia nigra and the further inhibition of P2X4 receptor-mediated autophagy. These results indicate that P2X4 receptor might serve as a potential novel target for the treatment of Parkinson’s disease. This study was approved by the Animal Ethics Committee of Affiliated Hospital of Qingdao University (approval No. QYFYWZLL26119) on April 12, 2016.

    Related Articles | Metrics
    Environmental enrichment combined with fasudil promotes motor function recovery and axonal regeneration after stroke
    Yi-Tong Zhu, Qun Zhang, Hong-Yu Xie, Ke-Wei Yu, Gao-Jing Xu, Si-Yue Li, Yi Wu
    2021, 16 (12):  2512-2520.  doi: 10.4103/1673-5374.313048
    Abstract ( 149 )   PDF (2365KB) ( 193 )   Save
    Fasudil, a Rho-associated protein kinase (ROCK) inhibitor, has a protective effect on the central nervous system. In addition, environmental enrichment is a promising technique for inducing the recovery of motor impairments in ischemic stroke models. The present study aimed to explore whether environmental enrichment combined with fasudil can facilitate motor function recovery and induce cortical axonal regeneration after stroke. First, a mouse model of ischemic cerebral stroke was established by photochemical embolization of the left sensorimotor cortex. Fasudil solution (10 mg/kg per day) was injected intraperitoneally for 21 days after the photothrombotic stroke. An environmental enrichment intervention was performed on days 7–21 after the photothrombotic stroke. The results revealed that environmental enrichment combined with fasudil improved motor function, increased growth-associated protein 43 expression in the infarcted cerebral cortex, promoted axonal regeneration on the contralateral side, and downregulated ROCK, p-LIM domain kinase (LIMK)1, and p-cofilin expression. The combined intervention was superior to monotherapy. These findings suggest that environmental enrichment combined with fasudil treatment promotes motor recovery after stroke, at least partly by stimulating axonal regeneration. The underlying mechanism might involve ROCK/LIMK1/cofilin pathway regulation. This study was approved by the Institutional Animal Care and Use Committee of Fudan University, China (approval No. 20160858A232) on February 24, 2016. 
    Related Articles | Metrics
    Stress increases MHC-I expression in dopaminergic neurons and induces autoimmune activation in Parkinson’s disease
    Bao-Yan Wang, Yong-Yi Ye, Chen Qian, Hong-Bo Zhang, Heng-Xu Mao, Long-Ping Yao, Xiang Sun, Guo-Hui Lu, Shi-Zhong Zhang
    2021, 16 (12):  2521-2527.  doi: 10.4103/1673-5374.313057
    Abstract ( 291 )   PDF (1301KB) ( 161 )   Save
    The expression of major histocompatibility complex class I (MHC-I), a key antigen-presenting protein, can be induced in dopaminergic neurons in the substantia nigra, thus indicating its possible involvement in the occurrence and development of Parkinson’s disease. However, it remains unclear whether oxidative stress induces Parkinson’s disease through the MHC-I pathway. In the present study, polymerase chain reaction and western blot assays were used to determine the expression of MHC-I in 1-methyl-4-phenylpyridinium (MPP+)-treated SH-SY5Y cells and a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson’s disease mouse model. The findings revealed that MHC-I was expressed in both models. To detect whether the expression of MHC-I was able to trigger the infiltration of cytotoxic T cells, immunofluorescence staining was used to detect cytotoxic cluster of differentiation 8 (CD8)+ T cell infiltration in the substantia nigra of MPTP-treated mice. The results indicated that the presentation of MHC-I in dopaminergic neurons was indeed accompanied by an increase in the number of CD8+ T cells. Moreover, in MPTP-induced Parkinson’s disease model mice, the genetic knockdown of endogenous MHC-I, which was caused by injecting specific adenovirus into the substantia nigra, led to a significant reduction in CD8+ T cell infiltration and alleviated dopaminergic neuronal death. To further investigate the molecular mechanisms of oxidative stress-induced MHC-I presentation, the expression of PTEN-induced kinase 1 (PINK1) was silenced in MPP+-treated SH-SY5Y cells using specific small interfering RNA (siRNA), and there was more presentation of MHC-I in these cells compared with control siRNA-treated cells. Taken together, MPP+-/MPTP-induced oxidative stress can trigger MHC-I presentation and autoimmune activation, thus rendering dopaminergic neurons susceptible to immune cells and degeneration. This may be one of the mechanisms of oxidative stress-induced Parkinson’s disease, and implies the potential neuroprotective role of PINK1 in oxidative stress-induced MHC-I presentation. All animal experiments were approved by the Southern Medical University Ethics Committee (No. 81802040, approved on February 25, 2018).
    Related Articles | Metrics
    Structural remodeling in related brain regions in patients with facial synkinesis
    Jia-Jia Wu, Ye-Chen Lu, Mou-Xiong Zheng, Xu-Yun Hua, Chun-Lei Shan, Wei Ding, Jian-Guang Xu
    2021, 16 (12):  2528-2533.  doi: 10.4103/1673-5374.313055
    Abstract ( 163 )   PDF (1084KB) ( 173 )   Save
    Facial synkinesis is a troublesome sequelae of facial nerve malfunction. It is difficult to recover from synkinesis, despite improved surgical techniques for isolating the peripheral facial nerve branches. Furthermore, it remains unclear whether long-term dysfunction of motor control can lead to irreversible plasticity-induced structural brain changes. This case-control study thus investigated the structural brain alterations associated with facial synkinesis. The study was conducted at Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, China. Twenty patients with facial synkinesis (2 male and 18 female, aged 33.35 ± 6.97 years) and 19 healthy volunteers (2 male and 17 female, aged 33.21 ± 6.75 years) underwent magnetic resonance imaging, and voxel-based and surface-based morphometry techniques were used to analyze data. There was no significant difference in brain volume between patients with facial synkinesis and healthy volunteers. Patients with facial synkinesis exhibited a significantly reduced cortical thickness in the contralateral superior and inferior temporal gyri and a reduced sulcal depth of the ipsilateral precuneus compared with healthy volunteers. In addition, sulcal depth of the ipsilateral precuneus was negatively correlated with the severity of depression. These findings suggest that there is a structural remodeling of gray matter in patients with facial synkinesis after facial nerve malfunction. This study was approved by the Ethics Review Committee of the Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, China (approval No. 2017-365-T267) on September 13, 2017, and was registered with the Chinese Clinical Trial Registry (registration number: ChiCTR1800014630) on January 25, 2018.
    Related Articles | Metrics
    Gene therapy with caspase-3 small interfering RNA-nanoparticles is neuroprotective after optic nerve damage
    Mohamed Tawfik, Xiwei Zhang, Lisa Grigartzik, Peter Heiduschka, Werner Hintz, Petra Henrich-Noack, Berend van Wachem, Bernhard A. Sabel
    2021, 16 (12):  2534-2541.  doi: 10.4103/1673-5374.313068
    Abstract ( 154 )   PDF (1226KB) ( 183 )   Save
    Apoptosis, a key mechanism of programmed cell death, is triggered by caspase-3 protein and lowering its levels with gene therapy may rescue cell death after central nervous system damage. We developed a novel, non-viral gene therapy to block caspase-3 gene expression using small interfering RNA (siRNA) delivered by polybutylcyanoacrylate nanoparticles (CaspNPs). In vitro CaspNPs significantly blocked caspase-3 protein expression in C6 cells, and when injected intraocularly in vivo, CaspNPs lowered retinal capsase-3 immunofluorescence by 57.9% in rats with optic nerve crush. Longitudinal, repeated retinal ganglion cell counts using confocal neuroimaging showed that post-traumatic cell loss after intraocular CaspNPs injection was only 36.1% versus 63.4% in lesioned controls. Because non-viral gene therapy with siRNA-nanoparticles can selectively silence caspace-3 gene expression and block apoptosis in post-mitotic neurons, siRNA delivery with nanoparticles may be promising for neuroprotection or restoration of central visual system damage and other neurological disorders. The animal study procedures were approved by the German National Act on the use of experimental animals (Ethic Committee Referat Verbraucherschutz, Veterinärangelegenheiten; Landesverwaltungsamt Sachsen-Anhalt, Halle, Germany, # IMP/G/01-1150/12 and # IMP/G/01-1469/17).
    Related Articles | Metrics
    Inhibitor of DNA binding 2 accelerates nerve regeneration after sciatic nerve injury in mice
    Zhong-Hai Huang, Ai-Ying Feng, Jing Liu, Libing Zhou, Bing Zhou, Panpan Yu
    2021, 16 (12):  2542-2548.  doi: 10.4103/1673-5374.313054
    Abstract ( 142 )   PDF (3376KB) ( 118 )   Save
    Inhibitor of DNA binding 2 (Id2) can promote axonal regeneration after injury of the central nervous system. However, whether Id2 can promote axonal regeneration and functional recovery after peripheral nerve injury is currently unknown. In this study, we established a mouse model of bilateral sciatic nerve crush injury. Two weeks before injury, AAV9-Id2-3×Flag-GFP was injected stereotaxically into the bilateral ventral horn of lumbar spinal cord. Our results showed that Id2 was successfully delivered into spinal cord motor neurons projecting to the sciatic nerve, and the number of regenerated motor axons in the sciatic nerve distal to the crush site was increased at 2 weeks after injury, arriving at the tibial nerve and reinnervating a few endplates in the gastrocnemius muscle. By 1 month after injury, extensive neuromuscular reinnervation occurred. In addition, the amplitude of compound muscle action potentials of the gastrocnemius muscle was markedly recovered, and their latency was shortened. These findings suggest that Id2 can accelerate axonal regeneration, promote neuromuscular reinnervation, and enhance functional improvement following sciatic nerve injury. Therefore, elevating the level of Id2 in adult neurons may present a promising strategy for peripheral nerve repair following injury. The study was approved by the Experimental Animal Ethics Committee of Jinan University (approval No. 20160302003) on March 2, 2016.
    Related Articles | Metrics
    Spastin interacts with collapsin response mediator protein 3 to regulate neurite growth and branching
    Zhi-Sheng Ji, Jian-Ping Li, Chao-Hua Fu, Jian-Xian Luo, Hua Yang, Guo-Wei Zhang, Wutian Wu, Hong-Sheng Lin
    2021, 16 (12):  2549-2556.  doi: 10.4103/1673-5374.313052
    Abstract ( 132 )   PDF (2660KB) ( 160 )   Save
    Cytoskeletal microtubule rearrangement and movement are crucial in the repair of spinal cord injury. Spastin plays an important role in the regulation of microtubule severing. Both spastin and collapsin response mediator proteins can regulate neurite growth and branching; however, whether spastin interacts with collapsin response mediator protein 3 (CRMP3) during this process remains unclear, as is the mechanism by which CRMP3 participates in the repair of spinal cord injury. In this study, we used a proteomics approach to identify key proteins associated with spinal cord injury repair. We then employed liquid chromatography-mass spectrometry to identify proteins that were able to interact with glutathione S-transferase-spastin. Then, co-immunoprecipitation and staining approaches were used to evaluate potential interactions between spastin and CRMP3. Finally, we co-transfected primary hippocampal neurons with CRMP3 and spastin to evaluate their role in neurite outgrowth. Mass spectrometry identified the role of CRMP3 in the spinal cord injury repair process. Liquid chromatography-mass spectrometry pulldown assays identified three CRMP3 peptides that were able to interact with spastin. CRMP3 and spastin were co-expressed in the spinal cord and were able to interact with one another in vitro and in vivo. Lastly, CRMP3 overexpression was able to enhance the ability of spastin to promote neurite growth and branching. Therefore, our results confirm that spastin and CRMP3 play roles in spinal cord injury repair by regulating neurite growth and branching. These proteins may therefore be novel targets for spinal cord injury repair. The Institutional Animal Care and Use Committee of Jinan University, China approved this study (approval No. IACUS-20181008-03) on October 8, 2018. 
    Related Articles | Metrics
    Tensile mechanical analysis of anisotropy and velocity dependence of the spinal cord white matter: a biomechanical study
    Norihiro Nishida, Itsuo Sakuramoto, Yoshihiro Fujii, Rudolf Yoga Hutama, Fei Jiang, Junji Ohgi, Yasuaki Imajo, Hidenori Suzuki, Masahiro Funaba, Xian Chen, Takashi Sakai
    2021, 16 (12):  2557-2562.  doi: 10.4103/1673-5374.313059
    Abstract ( 119 )   PDF (2056KB) ( 111 )   Save
    In spinal cord injuries, external forces from various directions occur at various velocities. Therefore, it is important to physically evaluate whether the spinal cord is susceptible to damage and an increase in internal stress for external forces. We hypothesized that the spinal cord has mechanical features that vary under stress depending on the direction and velocity of injury. However, it is difficult to perform experiment because the spinal cord is very soft. There are no reports on the effects of multiple external forces. In this study, we used bovine spinal cord white matter to test and analyze the anisotropy and velocity dependence of the spinal cord. Tensile-vertical, tensile-parallel, shear-vertical, and shear-parallel tests were performed on the white matter in the fibrous direction (cranial to caudal). Strain rate in the experiment was 0.1, 1, 10, and 100/s. We calculated the Young’s modulus of the spinal cord. Results of the tensile and shear tests revealed that stress tended to increase when external forces were applied parallel to the direction of axon fibers, such as in tensile-vertical and shear-vertical tests. However, external forces those tear against the fibrous direction and vertically, such as in tensile-parallel and shear-parallel tests, were less likely to increase stress even with increased velocity. We found that the spinal cord was prone to external forces, especially in the direction of the fibers, and to be under increased stress levels when the velocity of external forces increased. From these results, we confirmed that the spinal cord has velocity dependence and anisotropy. The Institutional Animal Care and Use Committee of Yamaguchi University waived the requirement for ethical approval.
    Related Articles | Metrics