Loading...

Table of Content

    15 November 2021, Volume 16 Issue 11 Previous Issue    Next Issue
    For Selected: Toggle Thumbnails
    Mechanisms implicated in the contralateral effect in the central nervous system after unilateral injury: focus on the visual system
    Fernando Lucas-Ruiz, Caridad Galindo-Romero, Virginia Albaladejo-García, Manuel Vidal-Sanz, Marta Agudo-Barriuso
    2021, 16 (11):  2125-2131.  doi: 10.4103/1673-5374.310670
    Abstract ( 162 )   PDF (1259KB) ( 169 )   Save
    The retina, as part of the central nervous system is an ideal model to study the response of neurons to injury and disease and to test new treatments. During the last decade is becoming clear that unilateral lesions in bilateral areas of the central nervous system trigger an inflammatory response in the contralateral uninjured site. This effect has been better studied in the visual system where, as a rule, one retina is used as experimental and the other as control. Contralateral retinas in unilateral models of retinal injury show neuronal degeneration and glial activation. The mechanisms by which this adverse response in the central nervous system occurs are discussed in this review, focusing primarily on the visual system. 
    Related Articles | Metrics
    Toward three-dimensional in vitro models to study neurovascular unit functions in health and disease
    Tara M. Caffrey, #, Emily B. Button, Jerome Robert, #
    2021, 16 (11):  2132-2140.  doi: 10.4103/1673-5374.310671
    Abstract ( 151 )   PDF (2095KB) ( 155 )   Save
    The high metabolic demands of the brain require an efficient vascular system to be coupled with neural activity to supply adequate nutrients and oxygen. This supply is coordinated by the action of neurons, glial and vascular cells, known collectively as the neurovascular unit, which temporally and spatially regulate local cerebral blood flow through a process known as neurovascular coupling. In many neurodegenerative diseases, changes in functions of the neurovascular unit not only impair neurovascular coupling but also permeability of the blood-brain barrier, cerebral blood flow and clearance of waste from the brain. In order to study disease mechanisms, we need improved physiologically-relevant human models of the neurovascular unit. Advances towards modeling the cellular complexity of the neurovascular unit in vitro have been made using stem-cell derived organoids and more recently, vascularized organoids, enabling intricate studies of non-cell autonomous processes. Engineering and design innovations in microfluidic devices and tissue engineering are progressing our ability to interrogate the cerebrovasculature. These advanced models are being used to gain a better understanding of neurodegenerative disease processes and potential therapeutics. Continued innovation is required to build more physiologically-relevant models of the neurovascular unit encompassing both the cellular complexity and designed features to interrogate neurovascular unit functionality. 
    Related Articles | Metrics
    Apolipoprotein A1, the neglected relative of Apolipoprotein E and its potential role in Alzheimer’s disease
    Kristina Endres
    2021, 16 (11):  2141-2148.  doi: 10.4103/1673-5374.310669
    Abstract ( 102 )   PDF (532KB) ( 149 )   Save
    Lipoproteins are multi-molecule assemblies with the primary function of transportation and processing of lipophilic substances within aqueous bodily fluids (blood, cerebrospinal fluid). Nevertheless, they also exert other physiological functions such as immune regulation.  In particular, neurons are both sensitive to uncontrolled responses of the immune system and highly dependent on a controlled and sufficient supply of lipids. For this reason, the role of certain lipoproteins and their protein-component (apolipoproteins, Apo’s) in neurological diseases is perceivable. ApoE, for example, is well-accepted as one of the major risk factors for sporadic Alzheimer’s disease with a protective allele variant (ε2) and a risk-causing allele variant (ε4). ApoA1, the major protein component of high-density lipoproteins, is responsible for transportation of excess cholesterol from peripheral tissues to the liver. The protein is synthesized in the liver and intestine but also can enter the brain via the choroid plexus and thereby might have an impact on brain lipid homeostasis. This review focuses on the role of ApoA1 in Alzheimer’s disease and discusses whether its role within this neurodegenerative disorder is specific or represents a general neuroprotective mechanism.
    Related Articles | Metrics
    Oral frailty and neurodegeneration in Alzheimer’s disease
    Vittorio Dibello, #, Madia Lozupone, #, Daniele Manfredini, Antonio Dibello, Roberta Zupo, Rodolfo Sardone, Antonio Daniele, Frank Lobbezoo, Francesco Panza
    2021, 16 (11):  2149-2153.  doi: 10.4103/1673-5374.310672
    Abstract ( 102 )   PDF (391KB) ( 163 )   Save
    Frailty is a critical intermediate status of the aging process with a multidimensional and multisystem nature and at higher risk for adverse health-related outcomes, including falls, disability, hospitalizations, institutionalization, mortality, dementia, and Alzheimer’s disease. Among different frailty phenotypes, oral frailty has been recently suggested as a novel construct defined as a decrease in oral function with a coexisting decline in cognitive and physical functions. We briefly reviewed existing evidence on operational definitions of oral frailty, assessment and screening tools, and possible relationships among oral frailty, oral microbiota, and Alzheimer’s disease neurodegeneration. Several underlying mechanism may explain the oral health-frailty links including undernutrition, sarcopenia linked to both poor nutrition and frailty, psychosocial factors, and the chronic inflammation typical of oral disease. Oral microbiota may influence Alzheimer’s disease risk through circulatory or neural access to the brain and the interplay with periodontal disease, often causing tooth loss also linked to an increased Alzheimer’s disease risk. On this bases, COR388, a bacterial protease inhibitor targeting Porphyromonas gingivalis implicated in periodontal disease, is now being tested in a double-blind, placebo-controlled Phase II/III study in mild-to-moderate Alzheimer’s disease. Therefore, oral status may be an important contributor to general health, including Alzheimer’s disease and late-life cognitive disorders, suggesting the central role of preventive strategies targeting the novel oral frailty phenotype and including maintenance and improvement of oral function and nutritional status to reduce the burden of both oral dysfunction and frailty.
    Related Articles | Metrics
    Non-coding RNAs and other determinants of neuroinflammation and endothelial dysfunction: regulation of gene expression in the acute phase of ischemic stroke and possible therapeutic applications
    Mario Daidone, Marco Cataldi, Antonio Pinto, Antonino Tuttolomondo
    2021, 16 (11):  2154-2158.  doi: 10.4103/1673-5374.310607
    Abstract ( 103 )   PDF (303KB) ( 167 )   Save
    Ischemic stroke occurs under a variety of clinical conditions and has different pathogeneses, resulting in necrosis of brain parenchyma. Stroke pathogenesis is characterized by neuroinflammation and endothelial dysfunction. Some of the main processes triggered in the early stages of ischemic damage are the rapid activation of resident inflammatory cells (microglia, astrocytes and endothelial cells), inflammatory cytokines, and translocation of intercellular nuclear factors. Inflammation in stroke includes all the processes mentioned above, and it consists of either protective or detrimental effects concerning the “polarization” of these processes. This polarization comes out from the interaction of all the molecular pathways that regulate genome expression: the epigenetic factors. In recent years, new regulation mechanisms have been cleared, and these include non-coding RNAs, adenosine receptors, and the activity of mesenchymal stem/stromal cells and microglia. We reviewed how long non-coding RNA and microRNA have emerged as an essential mediator of some neurological diseases. We also clarified that their roles in cerebral ischemic injury may provide novel targets for the treatment of ischemic stroke. To date, we do not have adequate tools to control pathophysiological processes associated with stroke. Our goal is to review the role of non-coding RNAs and innate immune cells (such as microglia and mesenchymal stem/stromal cells) and the possible therapeutic effects of their modulation in patients with acute ischemic stroke. A better understanding of the mechanisms that influence the “polarization” of the inflammatory response after the acute event seems to be the way to change the natural history of the disease.
    Related Articles | Metrics
    Altered microRNA expression in animal models of Huntington’s disease and potential therapeutic strategies
    Bridget Martinez, Philip V. Peplow,
    2021, 16 (11):  2159-2169.  doi: 10.4103/1673-5374.310673
    Abstract ( 109 )   PDF (487KB) ( 101 )   Save
    A review of recent animal models of Huntington’s disease showed many microRNAs had altered expression levels in the striatum and cerebral cortex, and which were mostly downregulated. Among the altered microRNAs were miR-9/9*, miR-29b, miR-124a, miR-132, miR-128, miR-139, miR-122, miR-138, miR-23b, miR-135b, miR-181 (all downregulated) and miR-448 (upregulated), and similar changes had been previously found in Huntington’s disease patients. In the animal cell studies, the altered microRNAs included miR-9, miR-9*, miR-135b, miR-222 (all downregulated) and miR-214 (upregulated). In the animal models, overexpression of miR-155 and miR-196a caused a decrease in mutant huntingtin mRNA and protein level, lowered the mutant huntingtin aggregates in striatum and cortex, and improved performance in behavioral tests. Improved performance in behavioral tests also occurred with overexpression of miR-132 and miR-124. In the animal cell models, overexpression of miR-22 increased the viability of rat primary cortical and striatal neurons infected with mutant huntingtin and decreased huntingtin -enriched foci of ≥ 2 µm. Also, overexpression of miR-22 enhanced the survival of rat primary striatal neurons treated with 3-nitropropionic acid. Exogenous expression of miR-214, miR-146a, miR-150, and miR-125b decreased endogenous expression of huntingtin mRNA and protein in HdhQ111/HdhQ111 cells. Further studies with animal models of Huntington’s disease are warranted to validate these findings and identify specific microRNAs whose overexpression inhibits the production of mutant huntingtin protein and other harmful processes and may provide a more effective means of treating Huntington’s disease in patients and slowing its progression.
    Related Articles | Metrics
    Mesenchymal stem cell treatment for peripheral nerve injury: a narrative review
    Rui-Cheng Zhang, #, Wen-Qi Du, #, Jing-Yuan Zhang, #, Shao-Xia Yu, Fang-Zhi Lu, Hong-Mei Ding, Yan-Bo Cheng, Chao Ren, De-Qin Geng
    2021, 16 (11):  2170-2176.  doi: 10.4103/1673-5374.310941
    Abstract ( 131 )   PDF (1242KB) ( 272 )   Save

    Peripheral nerve injuries occur as the result of sudden trauma and lead to reduced quality of life. The peripheral nervous system has an inherent capability to regenerate axons. However, peripheral nerve regeneration following injury is generally slow and incomplete that results in poor functional outcomes such as muscle atrophy. Although conventional surgical procedures for peripheral nerve injuries present many benefits, there are still several limitations including scarring, difficult accessibility to donor nerve, neuroma formation and a need to sacrifice the autologous nerve. For many years, other therapeutic approaches for peripheral nerve injuries have been explored, the most notable being the replacement of Schwann cells, the glial cells responsible for clearing out debris from the site of injury. Introducing cultured Schwann cells to the injured sites showed great benefits in promoting axonal regeneration and functional recovery. However, there are limited sources of Schwann cells for extraction and difficulties in culturing Schwann cells in vitro. Therefore, novel therapeutic avenues that offer maximum benefits for the treatment of peripheral nerve injuries should be investigated. This review focused on strategies using mesenchymal stem cells to promote peripheral nerve regeneration including exosomes of mesenchymal stem cells, nerve engineering using the nerve guidance conduits containing mesenchymal stem cells, and genetically engineered mesenchymal stem cells. We present the current progress of mesenchymal stem cell treatment of peripheral nerve injuries.

    Related Articles | Metrics
    Corneal neuromediator profiles following laser refractive surgery
    Lily Wei Yun Yang, Jodhbir S. Mehta, Yu-Chi Liu
    2021, 16 (11):  2177-2183.  doi: 10.4103/1673-5374.308666
    Abstract ( 110 )   PDF (927KB) ( 139 )   Save
    Laser refractive surgery is one of the most commonly performed procedures worldwide. In laser refractive surgery, Femtosecond Laser in Situ Keratomileusis and Refractive Lenticule Extraction have emerged as promising alternatives to microkeratome Laser in Situ Keratomileusis and Photorefractive Keratectomy. Following laser refractive surgery, the corneal nerves, epithelial and stromal cells release neuromediators, including neurotrophins, neuropeptides and neurotransmitters. Notably, nerve growth factor, substance P, calcitonin gene-related peptide and various cytokines are important mediators of neurogenic inflammation and corneal nerve regeneration. Alterations in neuromediator profiles and ocular surface parameters following laser refractive surgery are attributed to the surgical techniques and the severity of tissue insult induced. In this review, we will discuss the (1) Functions of neuromediators and their physiological and clinical significance; (2) Changes in the neuromediators following various laser refractive surgeries; (3) Correlation between neuromediators, ocular surface health and corneal nerve status; and (4) Future directions, including the use of neuromediators as potential biomarkers for ocular surface health following laser refractive surgery, and as adjuncts to aid in corneal regeneration after laser refractive surgery.
    Related Articles | Metrics
    Neuroimmune connections between corticotropin-releasing hormone and mast cells: novel strategies for the treatment of neurodegenerative diseases
    Piplu Bhuiyan, Yi-Wei Wang, Huan-Huan Sha, Hong-Quan Dong, Yan-Ning Qian
    2021, 16 (11):  2184-2197.  doi: 10.4103/1673-5374.310608
    Abstract ( 117 )   PDF (1177KB) ( 173 )   Save
    Corticotropin-releasing hormone is a critical component of the hypothalamic–pituitary–adrenal axis, which plays a major role in the body’s immune response to stress. Mast cells are both sensors and effectors in the interaction between the nervous and immune systems. As first responders to stress, mast cells can initiate, amplify and prolong neuroimmune responses upon activation. Corticotropin-releasing hormone plays a pivotal role in triggering stress responses and related diseases by acting on its receptors in mast cells. Corticotropin-releasing hormone can stimulate mast cell activation, influence the activation of immune cells by peripheral nerves and modulate neuroimmune interactions. The latest evidence shows that the release of corticotropin-releasing hormone induces the degranulation of mast cells under stress conditions, leading to disruption of the blood-brain barrier, which plays an important role in neurological diseases, such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, autism spectrum disorder and amyotrophic lateral sclerosis. Recent studies suggest that stress increases intestinal permeability and disrupts the blood-brain barrier through corticotropin-releasing hormone-mediated activation of mast cells, providing new insight into the complex interplay between the brain and gastrointestinal tract. The neuroimmune target of mast cells is the site at which the corticotropin-releasing hormone directly participates in the inflammatory responses of nerve terminals. In this review, we focus on the neuroimmune connections between corticotropin-releasing hormone and mast cells, with the aim of providing novel potential therapeutic targets for inflammatory, autoimmune and nervous system diseases.
    Related Articles | Metrics
    Searching for alternatives to brain regeneration
    Chiara La Rosa, Luca Bonfanti
    2021, 16 (11):  2198-2200.  doi: 10.4103/1673-5374.310683
    Abstract ( 98 )   PDF (964KB) ( 90 )   Save
    Brain regeneration from an evolutionary perspective: Brain regeneration (the full restoration of tissue after loss from injury or disease) is the most sought after goal for researchers working in developmental neurobiology. It also appears to be the most challenging to achieve when considering the mammalian brain. Whereas remarkable regenerative capacities can be present in the central nervous systems of many non-mammalian vertebrates (e.g., fish, amphibians), these kinds of processes appear to be dramatically reduced in mammals (Bonfanti, 2011). The reasons for such differences across animal classes are not completely understood, yet, some clear aspects have emerged from the study of well-established models like the teleost fish brain (Lange and Brand, 2020), which has: i) multiple, widespread stem cell niches that provide continuous, physiological cell renewal, as well as regeneration after lesioning; ii) additional neural elements that can de-differentiate after injury and re-acquire stem cell properties; iii) the ability to re-activate developmental programs in order to provide regenerative capacity. Studies on regeneration in various tissues and organs across animal species indicate that physiological and lesion-induced regeneration requires the coexistence of some (if not all) of the above-mentioned aspects, which, in the mammalian brain, are either absent or restricted to very small neurogenic niches. The most intuitive explanation for differences in brain regeneration across animal classes, apart from causal reasons, is the need for more neuroanatomical complexity linked to increased computational capabilities that often occurs in parallel with increased brain size. The “complexity” of large brains appears to be incompatible with substantial cell renewal/regeneration, a process that would be biologically expensive and somehow in contrast with the requirement for “stability” of the neural circuits (e.g., to retain long-term memories related to multiple previous experiences in long-living organisms). The current state of knowledge is still a mix of evidence and theories that are blurred by the frequently irregular patterns of evolution, but it does point to an important, underestimated issue: phylogenetic variations in the location, amount, rate, and type of brain plasticity in mammals.
    Related Articles | Metrics
    Mitofusin activation enhances mitochondrial motility and promotes neuroregeneration in CMT2A
    Gerald W. Dorn II
    2021, 16 (11):  2201-2203.  doi: 10.4103/1673-5374.310684
    Abstract ( 164 )   PDF (457KB) ( 109 )   Save
    Human brains represent only 2% of body mass, but their high relative metabolic activity accounts for ~20% of total body adenosine triphosphate (ATP) consumption. ATP generated by neuronal mitochondria fuels nerve signaling and homeostatic repair. In the peripheral nervous system, which has greater capacity for regeneration after physical, toxic or genetic injury than the central nervous system, ATP also powers actin polymerization/depolymerization for growth cone formation and axon extension. Mitochondrial ATP generation is therefore a central component of neuronal functioning in the central and peripheral nervous systems.
    Related Articles | Metrics
    Chitosan-based nanoparticles in Alzheimer’s disease: messenger or message?
    Eniko Manek, Georg A. Petroianu
    2021, 16 (11):  2204-2205.  doi: 10.4103/1673-5374.310685
    Abstract ( 104 )   PDF (264KB) ( 127 )   Save
    Cellulose is the most common natural (plant) polymer while its animal-kingdom close relative chitin comes in second. It is estimated that there are some 10 billion tons of chitin in the world (d’Ayala et al., 2008). Chitin may be described as cellulose with one hydroxyl group on each monomer replaced with an acetyl amine group. Chitin-producing organisms like protozoa, fungi, arthropods, and nematodes are often pathogens in other species. Despite the absence of endogenous chitin, mammals express chitinases (E.C 3.2.2.14) with enzymatic activity. 
    Related Articles | Metrics
    Neutrophil in diabetic stroke: emerging therapeutic strategies
    Rashmi Kumari, Kusum Sinha
    2021, 16 (11):  2206-2208.  doi: 10.4103/1673-5374.310677
    Abstract ( 110 )   PDF (612KB) ( 129 )   Save
    Post-ischemic inflammation is a coordinated process, which lasts from hours to days and involves recruitment of inflammatory cells from blood to the brain endothelial cells. Recently, the adhesion of leukocytes at endothelium, especially neutrophils, and its implication in post-stroke neuronal injury have been extensively explored and reported in both experimental and clinical  settings (Jian et al., 2019). However its role in diabetic patients following stroke is still elusive. Some significant differences such as risk factors, stroke subtypes and clinical outcomes are different between diabetic and non-diabetic. The higher prevalence of lacunar stroke, higher frequency of hypertension and lower neurological deficit at admission were reported earlier in diabetic patients. We found that early increase of neutrophils plays a prominent role in instigating a larger stroke size and worse clinical outcomes as compared to patients that do not have diabetes. After ischemic stroke, neutrophils are recruited to ischemic brain and can enter into the brain following hypoxia-ischemia (HI) through cerebral vessels, choroid plexus, and subarachnoid space. Figure 1A shows the various routes of entry of neutrophils in the db/db mouse brain 24 hours post stroke. Among all  immune cells, neutrophils  are  the first one to appear in the brain at day 1 post HI and remain until 7 days in the perilesional space, and subsequently other cells such as T & B lymphocytes  migrate to the lesion (Chu et al., 2014). These neutrophils remain in the vessel, release matrix metalloproteases and other proteases to damage the blood-brain barrier and the secondary damage starts, when neutrophils penetrate the brain parenchyma (Jickling et al., 2015) Previously, we have seen an increased matrix metalloproteinase-9 with graded infarct size and a direct relationship between matrix metalloproteinase-9 and neutrophils, which confirms the role of neutrophils mediating stroke injury (Kumari et al., 2020). 
    Related Articles | Metrics
    Evaluation of glial cell proliferation with non-invasive molecular imaging methods after stroke
    Ana Joya, Abraham Martín
    2021, 16 (11):  2209-2210.  doi: 10.4103/1673-5374.310681
    Abstract ( 92 )   PDF (631KB) ( 106 )   Save
    Glial proliferation: For the last decades, glial cells have been wrongly believed to have a mere passive supporting role for neurons. Nevertheless, this notion has clearly changed and it is now admitted that these cells are essential for the correct development and regulation of the nervous system. Glia cell population are commonly subdivided in astrocytes, oligodendrocytes and microglia. During the development, neural stem cells (NSCs) (called neuroepithelial progenitor cells or NPCs) transform into radial glia, the primary progenitor cells for neurons, astrocytes and oligodendrocytes (Zuchero and Barres, 2015). Microglial cells, however, derive from a mesenchymal precursor infiltration, meaning that during brain development, precursors generated in the bone narrow invade the nervous parenchyma and differentiate into microglial cells (Zuchero and Barres, 2015). This proliferative capacity is preserved in the adult mammalian brain, and neurogenic NSCs are stored in two restricted regions of the central nervous system (CNS), the forebrain subventricular zone (SVZ) and the hippocampal dentate gyrus (subgranular zone). These cells continue to produce neurons and glial cells during the adulthood, being activated after certain signals and leaving the quiescent state (Urbán et al., 2019). This process, in which glial progenitor cells differentiate into mature glia during development and in the adult brain to maintain and regulate brain function, is called gliogenesis (Ardaya et al., 2020). Besides these two niches, oligodendrocyte progenitor cells (OPCs) are present all around the CNS, both in the white and gray matter. These cells are the major dividing cells in the CNS generating new myelinating oligodendrocytes, or to a lesser extent astrocytes and they are constantly scanning the environment and controlling brain homeostasis. In addition, there is evidence of generation of new astrocytes from proliferating mature astrocytes in the brain parenchyma (Frisén, 2016). In summary, the capacity of generation of new glial cells is preserved not only in the SVZ and subgranular zone niches, but in the parenchymal tissue of the adult brain. In fact, the proliferative capacity of glial cells is increased in the injured CNS following neurological diseases. Adult OPCs play an important role in demyelinating diseases, where they turn to an activated state and start proliferating and migrating to the demyelination areas. Once there, they differentiate into mature oligodendrocytes and renew the destroyed myelin (Kuhn et al., 2019). After brain ischemia, microglia and astrocytes play an important role, representing the primary defense line facing neuroinflammation. Different studies using rodents have tried to disclose how microglia and astrocytes behave in this context and what triggers its activation. There is evidence of formation of a glial scar by reactive astrocytes originated from NSCs in the SVZ niche, but also generated through proliferation of local resident astrocytes (Nakafuku and Del Águila, 2020). Krishnasamy et al. (2017) showed the expression of nestin, a stem cell marker, in both reactive astrocytes and activated microglia after brain injury. These studies confirmed that glial cells differentiate and proliferate in order to restore the injured tissue, concluding that adult mammalian brain has the capacity of tissue repair following neuroinflammation. 
    Related Articles | Metrics
    It takes more than tau to tangle: using proteomics to determine how phosphorylated tau mediates toxicity in neurodegenerative diseases
    Geoffrey Pires, Eleanor Drummond
    2021, 16 (11):  2211-2212.  doi: 10.4103/1673-5374.310680
    Abstract ( 108 )   PDF (227KB) ( 140 )   Save
    Two of the most common causes of dementia are Alzheimer’s disease (AD) and frontotemporal dementia (FTD). AD is an irreversible, progressive neurodegenerative disorder that is clinically characterized by severe memory loss and behavioral impairment that eventually interferes with everyday function. AD is neuropathologically defined by the presence of extracellular β-amyloid plaques and intracellular accumulation of neurofibrillary tangles (NFTs) that primarily contain aggregated, hyperphosphorylated tau (pTau). Intriguingly, pTau is also the central protein in multiple subtypes of FTD (e.g. corticobasal degeneration, progressive supranuclear palsy, Pick’s disease). FTD is an umbrella term for a group of neurological conditions that primarily affect the temporal and frontal regions of the brain. Mutations in the tau gene (MAPT) can cause familial FTD, providing further evidence of the integral role of tau in FTD. Physiologically, tau regulates microtubule structure and dynamics, as well as axonal transport through interaction with tubulin. Tau is also involved in neuronal development and synaptogenesis. In AD and FTD, tau becomes hyperphosphorylated and undergoes major conformational changes, causing it to aggregate into the characteristic neuropathological lesions that define AD and FTD. Despite the known involvement of tau in these diseases, exactly how tau mediates toxicity is still unclear. 
    Related Articles | Metrics
    A biophysical perspective on the unexplored mechanisms driving Parkinson’s disease by amphetamine-like stimulants
    Carla Ferreira, Joana Couceiro, Sandra Tenreiro, Alexandre Quintas
    2021, 16 (11):  2213-2214.  doi: 10.4103/1673-5374.310675
    Abstract ( 98 )   PDF (583KB) ( 104 )   Save
    Epidemiological studies have reported an increased risk of Parkinson’s disease (PD) development in amphetamine-type stimulant users during their lifetime (Garwood et al., 2006; Rumpf et al., 2017). Protein inclusions mainly composed of misfolded and aggregated α-synuclein are the pathological hallmark of PD and other disorders known as synucleinopathies. Molecular studies present evidence that amphetamine upregulates α-synuclein synthesis in substantia nigra. The increment of α-synuclein levels promotes its aggregation and amyloid fibril formation, increasing  reactive oxygen species (ROS), and consequently dopamine oxidation (Wang and Witt, 2014), known to be toxic for dopaminergic neurons involved in motor function and limbic-motor integration. Over the years, these damaged cells lose their functionality and may die precociously, depleting the reserve of neural cells necessary for the normal neurological function which contributes to the onset of PD, when a critical number of cells are lost (Garwood et al., 2006). Therefore, the use of amphetamine-type stimulants may be a trigger event in the development of PD and parkinsonism, in conjugation to other risk factors that a given individual may hold. Despite the evidence, a previous study suggests that there is not enough data to corroborate the loss of dopamine neurons due to human amphetamine-type stimulant exposure, and consequently its implication in the PD development (Kish et al., 2017). Thus, elucidating the mechanisms underlying amphetamine-type stimulant influence on PD may contribute to better knowledge about the risk factors for the onset of this disease by these substances and adopt social policies to prevent future cases. The present perspective highlights the uncharted spots of the molecular mechanisms of α-synuclein aggregation pathways and how additional studies are necessary to understand the role of amphetamine-like stimulants as triggers of PD by changing α-synuclein thermodynamic and kinetic landscape. 
    Related Articles | Metrics
    Glycans to improve efficacy and solubility of protein aggregation inhibitors
    Ashim Paul, Daniel Segal, Elsa Zacco
    2021, 16 (11):  2215-2216.  doi: 10.4103/1673-5374.310688
    Abstract ( 109 )   PDF (217KB) ( 137 )   Save
    Misfolding and subsequent aberrant self-assembly of certain proteins into toxic amyloid deposits are hallmarks of various diseases, most notably neurodegenerative disorders such as Alzheimer’s disease (AD) and Parkinson’s disease (Chiti and Dobson, 2017). Aromatic residues in amyloidogenic proteins have been shown to be key factors in protein oligomerization and fibrilization, mostly driven by π-π interactions. Together with aromaticity, post-translational modifications can greatly affect a protein’s solubility and conformation and, as a consequence, its propensity to aggregate. Among post-translational modifications, this perspective focuses on protein glycosylation, the decoration of a protein with carbohydrate motifs, its effect on amyloid formation and its employment in the inhibition of protein aggregation. 
    Related Articles | Metrics
    Diagnostic and therapeutic potential of exosomal miRNAs in Alzheimer’s disease
    Ida Manna, Selene De Benedittis, Enrico Iaccino, Andrea Quattrone, Aldo Quattrone
    2021, 16 (11):  2217-2218.  doi: 10.4103/1673-5374.310674
    Abstract ( 91 )   PDF (548KB) ( 95 )   Save
    Alzheimer’s disease (AD) is a primary cause of dementia. AD is a neurodegenerative disorder, characterized by synapses loss, extracellular amyloid plaques composed of the amyloid-β peptide (Aβ) and intracellular aggregates of hyperphosphorylated tau protein. AD is a complex disease linked to multiple interacting factors, both environmental and genetic, which can contribute to the onset and severity of the disease. Longitudinal studies have highlighted several cardiovascular risk factors that can increase the risk of AD. The genetic landscape of AD has changed dramatically in recent decades. Early studies identified mutations in the amyloid precursor protein gene (APP) as well as proteins that are involved in the enzymatic cleavage of APP to toxic β-amyloid (Aβ), namely presenilin-1 and presnilin-2. However, these mutations were found in familial cases of early-onset AD, while the causes of sporadic late-onset AD are still unknown. The latest advances in Genome-wide Association Studies (GWAS), sequencing, and bioinformatics have begun to unravel the complex genetic architecture of the sporadic form of AD. GWAS were able to uncover common variants with high frequency in the population that individually carried low risk (Robinson et al., 2017). The advent of next-generation and third-generation sequencing platforms shows great promise in further unravelling the genetics of AD. Exome sequencing has been gradually optimized to identify mutations in protein-coding regions, and genome sequencing detects potential disease-causing mutations in non-coding sections of DNA. It has been suggested that underlying the base of neurodegenerative diseases, there is, also, an involvement of epigenetic mechanisms able to influence the expression of genes without altering the DNA sequence, including methylation, non-coding RNAs such as microRNA, and chromatin remodeling (Fenoglio et al., 2018). All these findings radically changed the understanding of AD pathology. In fact, the understanding of the genetic and epigenetic mechanisms and the biological pathways underlying AD has and will continue to have significant benefits also for the search for new therapeutic targets. Clinical symptoms of AD are assessed by instrumental and cognitive examinations, associated with the patient’s medical history, to establish a “probable AD” diagnosis (McKhann et al., 1984). To complete these assessments, five biomarkers of AD, divided into two categories, were validated in clinical practice. The first category of biomarkers concerns the dosage of the Aβ protein, i.e. the decrease in the concentrations of the Aβ42 protein in the cerebrospinal fluid (CSF). A second biomarker uses positron emission tomography (PET), a neuroimaging technique to measure Aβ deposition by calculating the absorption and retention of a tracer. These techniques are well correlated, and have been validated by post mortem examination. The second category of biomarkers concerns neurodegeneration: a first kind of biomarker is the total tau protein and phosphorylated tau assay in the patient’s CSF, which increases during the course of the disease; a second kind of biomarker is the use of structural magnetic resonance imaging to measure increased atrophy during AD. A third category of biomarkers is hypometabolism in disease as measured by [18F] fluorodeoxyglucose PET imaging. Again these correlate well with post mortem outcomes (Lashley et al., 2018). In summary, definitive diagnosis of AD is only possible with a post mortem examination of brain tissue showing senile plaques and neurofibrillary tangles. Diagnosis, even at an early stage, is now performed by tests on CSF and with PET, but these tests are expensive or invasive. To date, there are no peripheral AD biomarkers used in clinical practice, so, considering the invasive nature of lumbar puncture for CSF sampling and the cost of neuroimaging, there is an absolute need to have specific biomarkers for early diagnosis of AD. In this regard, recent works have shown that high-precision tests for plasma Aβ42/Aβ40 detection are predictive for the accumulation of Aβ in the brain. Therefore, the development of blood-based Aβ biomarkers is of great interest (Schindler et al., 2019). So overall, identification of cost-effective biomarkers and use of more accessible biofluids, such as blood, could represent valid peripheral biomarkers for the AD diagnosis. 
    Related Articles | Metrics
    Alpha-synuclein preformed fibrils: a tool to understand Parkinson’s disease and develop disease modifying therapy
    Piotr Chmielarz, Andrii Domanskyi
    2021, 16 (11):  2219-2221.  doi: 10.4103/1673-5374.310686
    Abstract ( 220 )   PDF (853KB) ( 167 )   Save
    Parkinson’s disease (PD) is the second most common neurodegenerative disorder characterized by multiple motor and non-motor symptoms, which include, among others, constipation, sleep disturbance, bradykinesia, gait and balance abnormalities, muscle stiffness and resting tremor. The motor symptoms are caused by progressive age-related death of dopaminergic neurons and in the vast majority of patients suffering from age-related idiopathic PD the cause of dopaminergic neurodegeneration is unknown. Even in the familial early-onset PD where genetic mutations have been identified, the molecular mechanisms driving degeneration of dopaminergic neurons are far from clear. Consequently, there is no clinically approved disease-modifying therapy capable of stopping or at least slowing down the disease progression.
    Related Articles | Metrics
    Exploring the contribution of the mitochondrial disulfide relay system to Parkinson’s disease: the PINK1/CHCHD4 interplay
    Giuseppe Arena, Nazanine Modjtahedi, Rejko Kruger
    2021, 16 (11):  2222-2224.  doi: 10.4103/1673-5374.310679
    Abstract ( 138 )   PDF (563KB) ( 148 )   Save
    Parkinson’s disease (PD) is a common movement disorder of the elderly caused by the degeneration of dopaminergic neurons in the substantia nigra pars compacta of the brain. 
    Related Articles | Metrics
    Amyloid pores in mitochondrial membranes
    Neville Vassallo
    2021, 16 (11):  2225-2226.  doi: 10.4103/1673-5374.310682
    Abstract ( 98 )   PDF (528KB) ( 107 )   Save
    Neurodegenerative diseases of the amyloid type include common conditions such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and amyotrophic lateral sclerosis. Despite the fact that the phenotypes of these neuropathic maladies differ widely, ranging from cognitive to motor and psychotic disturbances, they are all characterized by the pathological accumulation and deposition in the central nervous system of well-ordered protein aggregates known as amyloid fibrils. Accumulating evidence indicates that rather than the end-stage mature fibrils, however, it is the smaller, metastable intermediate forms (known as oligomers) of the aggregated protein and peptides which represent the most neurotoxic species (Chiti and Dobson, 2017). One suggested mechanism for such toxicity appears to involve the ability of oligomers to interact with plasma membranes whilst inducing cell leakage (Surguchov et al., 2017). However, contemporary work increasingly points to mitochondria, and hence mitochondrial membranes, as preferential targets for the pathogenic action of oligomers in the neuronal cell (Ghio et al., 2016).
    Related Articles | Metrics
    Mitochondrial inorganic polyphosphate (polyP): the missing link of mammalian bioenergetics
    Brendan McIntyre, Maria E. Solesio
    2021, 16 (11):  2227-2228.  doi: 10.4103/1673-5374.310687
    Abstract ( 133 )   PDF (502KB) ( 135 )   Save
    Bioenergetics imbalance is a deleterious feature, which is present in the etiopathology of many human diseases, including in diabetes, cancer, and neurodegeneration. Therefore, targeting the components of mammalian bioenergetics, as well as the mechanisms that regulate the relationship between these components, could be a promising pharmacological strategy against a wide variety of pathologies. While for many years mammalian bioenergetics has been exclusively circumscribed to the mitochondrial oxidative phosphorylation (OXPHOS), which is the main mechanism to obtain adenosine triphosphate (ATP) in mammals, and to the cytoplasmic glycolysis and the closely related pentose phosphate pathway, in the last few decades many authors have advocated for expanding this term to include all the mechanisms that are involved in matching the cellular demands and production of energy to meet the needs of the cell under different states, including physiological and pathological conditions. This broader definition will include other key energy metabolites, such as the ubiquitous inorganic polyphosphate (polyP).
    Related Articles | Metrics
    The neurosphere assay: an effective in vitro technique to study neural stem cells
    Rita Soares, Filipa F. Ribeiro, Diogo M. Lourenço, Rui S. Rodrigues, João B. Moreira, Ana M. Sebastião, Vanessa A. Morais, Sara Xapelli
    2021, 16 (11):  2229-2231.  doi: 10.4103/1673-5374.310678
    Abstract ( 272 )   PDF (911KB) ( 242 )   Save
    Neural stem cells (NSCs) are known to be present in the adult mammalian brain where they constitutively differentiate into the neuronal, astroglial, and oligodendroglial lineages, in defined processes termed neurogenesis, astrogliogenesis and oligodendrogenesis, respectively (reviewed in Braun and Jessberger, 2014). During brain development, NSCs are present throughout the brain, becoming progressively restricted to defined brain regions. In the adult brain, NSCs are mainly present in areas classically known as neurogenic niches, i.e. the subventricular zone (SVZ), along the lateral walls of the lateral ventricles, and the subgranular zone, located in the dentate gyrus (DG) of the hippocampus. These areas are particularly enriched with NSCs, which not only are multipotent cells but also proliferative cells with the ability to self-renew, thus maintaining their own pool of cells. In fact, neurogenesis, astrogliogenesis and oligodendrogenesis are highly intricate processes comprising several steps, including proliferation, differentiation, migration, and functional integration of the newly formed cells in the existing circuitry, which are regulated by a plethora of factors. These newly differentiated adult-born cells have the capacity to continuously modulate brain function and plasticity, by constantly reacting to external or internal stimuli (reviewed in Braun and Jessberger, 2014). 
    Related Articles | Metrics
    Over-specialization versus synergy in neuroscience: professionals’ integration is more than the sum of its parts
    Elisa Cainelli, Luca Vedovelli
    2021, 16 (11):  2232-2233.  doi: 10.4103/1673-5374.310676
    Abstract ( 125 )   PDF (385KB) ( 133 )   Save
    Health care systems in industrialized countries are built around acute health problems (Etzwiler, 1997). This approach is successful when the goal is to improve or overcome pathologies that result in severe health issues or death. With the recent tremendous improvement of the treatments of life-threatening conditions, the clinical focus has moved from short term results to the overall expected quality of life and long-term outcomes. Unfortunately, the new objectives imply to consider a multitude of interacting processes and variables. In fact, a complex interplay of biological systems (nervous, immune, and endocrine) works together in an integrated and synergistic way to deal with a constantly evolving environment, determining our physical, behavioral, and psychological/subjective conditions. This interplay is the mechanism that allows us to adapt promptly and appropriately to circumstances, the key feature for successful living. 
    Related Articles | Metrics
    Low-dose metformin treatment in the subacute phase improves the locomotor function of a mouse model of spinal cord injury
    Wen-Ye Song, #, Han Ding, #, Tiffany Dunn, #, Jun-Ling Gao, Javier Allende Labastida, Caitlin Schlagal, Guang-Zhi Ning, Shi-Qing Feng, Ping Wu
    2021, 16 (11):  2234-2242.  doi: 10.4103/1673-5374.310695
    Abstract ( 145 )   PDF (4688KB) ( 144 )   Save
    Metformin, a first-line drug for type-2 diabetes, has been shown to improve locomotor recovery after spinal cord injury. However, there are studies reporting no beneficial effect. Recently, we found that high dose of metformin (200 mg/kg, intraperitoneal) and acute phase administration (immediately after injury) led to increased mortality and limited locomotor function recovery. Consequently, we used a lower dose (100 mg/kg, i.p.) metformin in mice, and compared the effect of immediate administration after spinal cord injury (acute phase) with that of administration at 3 days post-injury (subacute phase). Our data showed that metformin treatment starting at the subacute phase significantly improved mouse locomotor function evaluated by Basso Mouse Scale (BMS) scoring. Immunohistochemical studies also revealed significant inhibitions of microglia/macrophage activation and astrogliosis at the lesion site. Furthermore, metformin treatment at the subacute phase reduced neutrophil infiltration. These changes were in parallel with the increased survival rate of spinal neurons in animals treated with metformin. These findings suggest that low-dose metformin treatment for subacute spinal cord injury can effectively improve the functional recovery possibly through anti-inflammation and neuroprotection. This study was approved by the Institute Animal Care and Use Committee at the University of Texas Medical Branch (approval No. 1008041C) in 2010.
    Related Articles | Metrics
    Teriflunomide provides protective properties after oxygen-glucose-deprivation in hippocampal and cerebellar slice cultures
    Anna Wolters, #, Judith Reuther, #, Philipp Gude, Thomas Weber, Carsten Theiss, Heike Vogelsang, #, Veronika Matschke, #
    2021, 16 (11):  2243-2249.  doi: 10.4103/1673-5374.310689
    Abstract ( 125 )   PDF (3117KB) ( 130 )   Save
    One of the major challenges in emergency medicine is out-of-hospital cardiac arrest (OHCA). Every year, about 53–62/100 000 people worldwide suffer an out-of-hospital cardiac arrest with serious consequences, whereas persistent brain injury is a major cause of morbidity and mortality of those surviving a cardiac arrest. Today, only few and insufficient strategies are known to limit neurological damage of ischemia and reperfusion injury. The aim of the present study was to investigate whether teriflunomide, an approved drug for treatment of relapsing-remitting-multiple-sclerosis, exerts a protective effect on brain cells in an in vitro model of ischemia. Therefore, organotypic slice cultures from rat hippocampus and cerebellum were exposed to oxygen-glucose-deprivation and subsequently treated with teriflunomide. The administration of teriflunomide in the reperfusion time on both hippocampal and cerebellar slice cultures significantly decreased the amount of detectable propidium iodide signal compared with an untreated culture, indicating that more cells survive after oxygen-glucose-deprivation. However, hippocampal slice cultures showed a higher vulnerability to ischemic conditions and a more sensitive response to teriflunomide compared with cerebellar slice cultures. Our study suggests that teriflunomide, applied as a post-treatment after an oxygen-glucose-deprivation, has a protective effect on hippocampal and cerebellar cells in organotypic slice cultures of rats. All procedures were conducted under established standards of the German federal state of North Rhine Westphalia, in accordance with the European Communities Council Directive 2010/63/EU on the protection of animals used for scientific purposes.
    Related Articles | Metrics
    Role of the caudate-putamen nucleus in sensory gating in induced tinnitus in rats
    Meng-Lin Wang, Yu Song, Jun-Xiu Liu, Ya-Li Du, Shan Xiong, Xin Fan, Jiang Wang, Zhi-Di Zhang, Lan-Qun Mao, Fu-Rong Ma,
    2021, 16 (11):  2250-2256.  doi: 10.4103/1673-5374.310692
    Abstract ( 110 )   PDF (1546KB) ( 124 )   Save
    Tinnitus can be described as the conscious perception of sound without external stimulation, and it is often accompanied by anxiety, depression, and insomnia. Current clinical treatments for tinnitus are ineffective. Although recent studies have indicated that the caudate-putamen nucleus may be a sensory gating area involved in noise elimination in tinnitus, the underlying mechanisms of this disorder are yet to be determined. To investigate the potential role of the caudate-putamen nucleus in experimentally induced tinnitus, we created a rat model of tinnitus induced by intraperitoneal administration of 350 mg/kg sodium salicylate. Our results revealed that the mean spontaneous firing rate of the caudate-putamen nucleus was increased by sodium salicylate treatment, while dopamine levels were decreased. In addition, electrical stimulation of the caudate-putamen nucleus markedly reduced the spontaneous firing rate of neurons in the primary auditory cortex. These findings suggest that the caudate-putamen nucleus plays a sensory gating role in sodium salicylate-induced tinnitus. This study was approved by the Institutional Animal Care and Use Committee of Peking University Health Science Center (approval No. A2010031) on December 6, 2017.
    Related Articles | Metrics

    Identification of potential candidate proteins for reprogramming spinal cord-derived astrocytes into neurons: a proteomic analysis

    Wen-Hao Chen, #, Yu-Xiang Lin, #, Ling Lin, Bao-Quan Zhang, Shu-Xia Xu, Wei Wang
    2021, 16 (11):  2257-2263.  doi: 10.4103/1673-5374.310697
    Abstract ( 130 )   PDF (2668KB) ( 326 )   Save
    Our previous study has confirmed that astrocytes overexpressing neurogenic differentiation factor 1 (NEUROD1) in the spinal cord can be reprogrammed into neurons under in vivo conditions. However, whether they can also be reprogrammed into neurons under in vitro conditions remains unclear, and the mechanisms of programmed conversion from astrocytes to neurons have not yet been clarified. In the present study, we prepared reactive astrocytes from newborn rat spinal cord astrocytes using the scratch method and infected them with lentivirus carrying NEUROD1. The results showed that NEUROD1 overexpression reprogrammed the cultured reactive astrocytes into neurons in vitro with an efficiency of 13.4%. Using proteomic and bioinformatic analyses, 1952 proteins were identified, of which 92 were differentially expressed. Among these proteins, 11 were identified as candidate proteins in the process of reprogramming based on their biological functions and fold-changes in the bioinformatic analysis. Furthermore, western blot assay revealed that casein kinase II subunit alpha (CSNK2A2) and pinin (PNN) expression in NEUROD1-overexpressing reactive astrocytes was significantly increased, suggesting that NEUROD1 can directly reprogram spinal cord-derived reactive astrocytes into neurons in vitro, and that the NEUROD1-CSNK2A2-PNN pathway is involved in this process. This study was approved by the Animal Ethics Committee of Fujian Medical University, China (approval No. 2016-05) on April 18, 2016. 
    Related Articles | Metrics
    Apelin-13 regulates electrical activity in the globus pallidus and induces postural changes in rats
    Ying Wang, Yan Xue, Cui Liu, Lei Chen
    2021, 16 (11):  2264-2268.  doi: 10.4103/1673-5374.310694
    Abstract ( 105 )   PDF (1006KB) ( 116 )   Save
    The globus pallidus is the relay nucleus of the basal ganglia, and changes in its electrical activity can cause motor impairment. Apelin-13 is widely distributed in the central and peripheral nervous systems. It has been demonstrated that apelin-13 plays important roles in the regulation of blood pressure and other non-motor functions. However, its role in motor function has rarely been reported. In the present study, apelin-13 (10 μM/100 μM) was injected into the globus pallidus of rats. The results showed that apelin-13 increased the spontaneous discharges in the majority of pallidal neurons. However, an apelin-13-induced inhibitory effect on the firing rate was also observed in a few pallidal neurons. In postural tests, after the systemic administration of haloperidol, unilateral pallidal injection of apelin-13 caused a contralateral deflection. Together, these findings suggest that apelin-13 regulates the electrical activity of pallidal neurons and thus participates in central motor control in rats. The study was approved by the Animal Ethics Committee of Qingdao University (approval No. 20200615Wistar0451003020) on June 15, 2020.
    Related Articles | Metrics
    A translational study of somatosensory evoked potential time–frequency components in rats, goats, and humans
    Hong-Yan Cui, #, Yi-Xin Wu, #, Rong Li, Guang-Sheng Li, Yong Hu
    2021, 16 (11):  2269-2275.  doi: 10.4103/1673-5374.310693
    Abstract ( 123 )   PDF (1327KB) ( 133 )   Save
    Somatosensory evoked potentials (SEPs) have been widely used to assess neurological function in clinical practice. A good understanding of the association between SEP signals and neurological function is helpful for precise diagnosis of impairment location. Previous studies on SEPs have been reported in animal models. However, few studies have reported the relationships between SEP waveforms in animals and those in humans. In this study, we collected normal SEP waveforms and decomposed them into specific time–frequency components (TFCs). Our results showed three stable TFC distribution regions in intact goats and rats and in humans. After we induced spinal cord injury in the animal models, a greater number of small TFC distribution regions were observed in the injured goat and rat groups than in the normal group. Moreover, there were significant correlations (P < 0.05) and linear relationships between the main SEP TFCs of the human group and those of the goat and rat groups. A stable TFC distribution of SEP components was observed in the human, goat and rat groups, and the TFC distribution modes were similar between the three groups. Results in various animal models in this study could be translated to future clinical studies based on SEP TFC analysis. Human studies were approved by the Institutional Review Board of the University of Hong Kong/Hospital Authority Hong Kong West Cluster (approval No. UM 05-312 T/975) on December 5, 2005. Rat experiments were approved by the Committee on the Use of Live Animals in Teaching and Research of Li Ka Shing Faculty of Medicine of the University of Hong Kong (approval No. CULART 2912-12) on January 28, 2013. Goat experiments were approved by the Animal Ethics Committee of Affiliated Hospital of Guangdong Medical University (approval No. GDY2002132) on March 5, 2018.
    Related Articles | Metrics
    Decellularized optic nerve functional scaffold transplant facilitates directional axon regeneration and remyelination in the injured white matter of the rat spinal cord
    Yu-Rong Bai, #, Bi-Qin Lai, #, Wei-Tao Han, Jia-Hui Sun, Ge Li, Ying Ding, Xiang Zeng, Yuan-Huan Ma, Yuan-Shan Zeng
    2021, 16 (11):  2276-2283.  doi: 10.4103/1673-5374.310696
    Abstract ( 158 )   PDF (7097KB) ( 82 )   Save
    Axon regeneration and remyelination of the damaged region is the most common repair strategy for spinal cord injury. However, achieving good outcome remains difficult. Our previous study showed that porcine decellularized optic nerve better mimics the extracellular matrix of the embryonic porcine optic nerve and promotes the directional growth of dorsal root ganglion neurites. However, it has not been reported whether this material promotes axonal regeneration in vivo. In the present study, a porcine decellularized optic nerve was seeded with neurotrophin-3-overexpressing Schwann cells. This functional scaffold promoted the directional growth and remyelination of regenerating axons. In vitro, the porcine decellularized optic nerve contained many straight, longitudinal channels with a uniform distribution, and microscopic pores were present in the channel wall. The spatial micro topological structure and extracellular matrix were conducive to the adhesion, survival and migration of neural stem cells. The scaffold promoted the directional growth of dorsal root ganglion neurites, and showed strong potential for myelin regeneration. Furthermore, we transplanted the porcine decellularized optic nerve containing neurotrophin-3-overexpressing Schwann cells in a rat model of T10 spinal cord defect in vivo. Four weeks later, the regenerating axons grew straight, the myelin sheath in the injured/transplanted area recovered its structure, and simultaneously, the number of inflammatory cells and the expression of chondroitin sulfate proteoglycans were reduced. Together, these findings suggest that porcine decellularized optic nerve loaded with Schwann cells overexpressing neurotrophin-3 promotes the directional growth of regenerating spinal cord axons as well as myelin regeneration. All procedures involving animals were conducted in accordance with the ethical standards of the Institutional Animal Care and Use Committee of Sun Yat-sen University (approval No. SYSU-IACUC-2019-B034) on February 28, 2019.
    Related Articles | Metrics
    A multi-channel collagen scaffold loaded with neural stem cells for the repair of spinal cord injury
    Shuo Liu, #, Yuan-Yuan Xie, #, Liu-Di Wang, Chen-Xu Tai, Dong Chen, Dan Mu, Yan-Yan Cui, Bin Wang
    2021, 16 (11):  2284-2292.  doi: 10.4103/1673-5374.310698
    Abstract ( 157 )   PDF (5849KB) ( 174 )   Save
    Collagen scaffolds possess a three-dimensional porous structure that provides sufficient space for cell growth and proliferation, the passage of nutrients and oxygen, and the discharge of metabolites. In this study, a porous collagen scaffold with axially-aligned luminal conduits was prepared. In vitro biocompatibility analysis of the collagen scaffold revealed that it enhances the activity of neural stem cells and promotes cell extension, without affecting cell differentiation. The collagen scaffold loaded with neural stem cells improved the hindlimb motor function in the rat model of T8 complete transection and promoted nerve regeneration. The collagen scaffold was completely degraded in vivo within 5 weeks of implantation, exhibiting good biodegradability. Rectal temperature, C-reactive protein expression and CD68 staining demonstrated that rats with spinal cord injury that underwent implantation of the collagen scaffold had no notable inflammatory reaction. These findings suggest that this novel collagen scaffold is a good carrier for neural stem cell transplantation, thereby enhancing spinal cord repair following injury. This study was approved by the Animal Ethics Committee of Nanjing Drum Tower Hospital (the Affiliated Hospital of Nanjing University Medical School), China (approval No. 2019AE02005) on June 15, 2019.
    Related Articles | Metrics
    Micro-computed tomography utility for estimation of intraparenchymal spinal cord cystic lesions in small animals
    Mahmoud Farrag, Dipak D. Pukale, Nic D. Leipzig,
    2021, 16 (11):  2293-2298.  doi: 10.4103/1673-5374.310690
    Abstract ( 141 )   PDF (2783KB) ( 160 )   Save
    Precise assessment of spinal cord cystic lesions is crucial to formulate effective therapeutic strategies, yet histological assessment of the lesion remains the primary method despite numerous studies showing inconsistent results regarding estimation of lesion size via histology. On the other hand, despite numerous advances in micro-computed tomography (micro-CT) imaging and analysis that have allowed precise measurements of lesion size, there is not enough published data on its application to estimate intraspinal lesion size in laboratory animal models. This work attempts to show that micro-CT can be valuable for spinal cord injury research by demonstrating accurate estimation of syrinx size and compares between micro-CT and traditional histological analysis. We used a post-traumatic syringomyelia rat model to compare micro-CT analysis to conventional histological analysis. The study showed that micro-CT can detect lesions within the spinal cord very similar to histology. Importantly, micro-CT appears to provide more accurate estimates of the lesions with more measures (e.g., surface area), can detect compounds within the cord, and can be done with the tissue of interest (spinal cord) intact. In summary, the experimental work presented here provides one of the first investigations of the use of micro-CT for estimating the size of intraparenchymal cysts and detecting materials within the spinal cord. All animal procedures were approved by the University of Akron Institutional Animal Care and Use Committee (IACUC) (protocol # LRE 16-05-09 approved on May 14, 2016). 
    Related Articles | Metrics
    Evaluation of the combined activity of benzimidazole arylhydrazones as new anti-Parkinsonian agents: monoamine oxidase-B inhibition, neuroprotection and oxidative stress modulation
    Neda Anastassova, Denitsa Aluani, Anton Kostadinov, Miroslav Rangelov, Nadezhda Todorova, Nadya Hristova-Avakumova, Maria Argirova, Nikolay Lumov, Magdalena Kondeva-Burdina, Virginia Tzankova, Denitsa Yancheva
    2021, 16 (11):  2299-2309.  doi: 10.4103/1673-5374.309843
    Abstract ( 183 )   PDF (3318KB) ( 153 )   Save
    Neuroprotective drugs and selective monoamine oxidase inhibitors can slow down the progression and improve symptoms of Parkinson’s disease (PD). Since there is an implication of oxidative stress in the pathophysiological mechanisms of the disease, the compounds possessing an ability to reduce the oxidative stress are prime candidates for neuroprotection. Thereby our current study is focused on the development of new multi-target PD drugs capable of inhibiting the activity of monoamine oxidase-B while exerting neuroprotective and antioxidant properties. A small series of benzimidazole derivatives containing hydroxy and methoxy arylhydrazone fragments has been synthesized and the neurotoxicity of the compounds has been evaluated in vitro on neuroblastoma SH-SY5Y cells and on isolated rat brain synaptosomes by measuring the cell viability and the levels of reduced glutathione and a good safety profile has been shown. The 2-hydroxy-4-methoxy substituted arylhydrazone 7 was the least toxic on neuronal SH-SY5Y cells and showed the lowest neurotoxicity in rat brain synaptosomes. The neuroprotective properties of the test compounds were further assessed using two models: H2O2-induced oxidative stress on SH-SY5Y cells and 6-hydroxydopamine-induced neurotoxicity in rat brain synaptosomes. Compound 7 showed more pronounced neuroprotective activity on SH-SY5Y cells, compared to the referent melatonin and rasagiline. It also preserved the synaptosomal viability and the reduced glutathione levels; the effects were stronger than those of rasagiline and comparable to melatonin. All the tested compounds were capable to inhibit human monoamine oxidase-B enzyme to a significant extent, however, compound 7 exerted the most prominent inhibitory activity, similar to selegiline and rasagiline. The carried out molecular docking studies revealed that the activity is related to the appropriate molecular structure enabling the ligand to enter deeper in the narrow and highly lipophylic active site pocket of the human monoamine oxidase-B and has a favoring interaction with the key amino acid residues Tyr326 and Cys172. Since much scientific evidence points out the implication of iron dyshomeostasis in PD, the compounds were tested to reduce the ferrous iron induced oxidative molecular damage on biologically important molecules in an in vitro lecithin containing model system. All the investigated compounds denoted protection effect, stronger than the one of the referent melatonin. In order to support the assignments of the significant neuroprotective and antioxidant pharmacological activities, the radical-scavenging mechanisms of the most promising compound 7 were evaluated using DFT methods. It was found that the most probable free radicals scavenging mechanism in nonpolar phase is the hydrogen atom transfer from the amide group of compound 7, while in polar medium the process is expected to occur by a proton transfer. The current study outlines a perspective leading structure, bearing the potential for a new anti-PD drug. All performed procedures were approved by the Institutional Animal Care Committee of  the Medical University of Sofia (Bulgarian Agency for Food Safety with Permission № 190, approved on February 6, 2020). 
    Related Articles | Metrics
    Phenotypic and functional comparison of rat enteric neural crest-derived cells during fetal and early-postnatal stages
    Dong-Hao Tian, #, Chuan-Hui Qin, #, Wen-Yao Xu, #, Wei-Kang Pan, Yu-Ying Zhao, Bai-Jun Zheng, Xin-Lin Chen, Yong Liu, Ya Gao, Hui Yu
    2021, 16 (11):  2310-2315.  doi: 10.4103/1673-5374.310701
    Abstract ( 142 )   PDF (2588KB) ( 131 )   Save
    In our previous study, we showed that with increasing time in culture, the growth characteristics of enteric neural crest-derived cells (ENCCs) change, and that the proliferation, migration and neural differentiation potential of these cells in vitro notably diminish. However, there are no studies on the developmental differences in these characteristics between fetal and early-postnatal stages in vitro or in vivo. In this study, we isolated fetal (embryonic day 14.5) and postnatal (postnatal day 2) ENCCs from the intestines of rats. Fetal ENCCs had greater maximum cross-sectional area of the neurospheres, stronger migration ability, and reduced apoptosis, compared with postnatal ENCCs. However, fetal and postnatal ENCCs had a similar differentiation ability. Fetal and postnatal ENCCs both survived after transplant into a rat model of Hirschsprung’s disease. In these rats with Hirschsprung’s disease, the number of ganglionic cells in the myenteric plexus was higher and the distal intestinal pressure change was greater in animals treated with fetal ENCCs compared with those treated with postnatal ENCCs. These findings suggest that, compared with postnatal ENCCs, fetal ENCCs exhibit higher survival and proliferation and migration abilities, and are therefore a more appropriate seed cell for the treatment of Hirschsprung’s disease. This study was approved by the Animal Ethics Committee of the Second Affiliated Hospital of Xi’an Jiaotong University (approval No. 2016086) on March 3, 2016.
    Related Articles | Metrics
    Transcriptome analysis of molecular mechanisms underlying facial nerve injury repair in rats
    Qian-Qian Cao, #, Shuo Li, #, Yan Lu, Di Wu, Wei Feng, Yong Shi, Lu-Ping Zhang
    2021, 16 (11):  2316-2323.  doi: 10.4103/1673-5374.310700
    Abstract ( 187 )   PDF (2482KB) ( 261 )   Save
    Although the transcriptional alterations inside the facial nucleus after facial nerve injury have been well studied, the gene expression changes in the facial nerve trunk after injury are still unknown. In this study, we established an adult rat model of facial nerve crush injury by compressing the right lateral extracranial nerve trunk. Transcriptome sequencing, differential gene expression analysis, and cluster analysis of the injured facial nerve trunk were performed, and 39 intersecting genes with significant variance in expression were identified. Gene Ontology annotation and Kyoto Encyclopedia of Genes and Genomes pathway analyses of the 39 intersecting genes revealed that these genes are mostly involved in leukocyte cell-cell adhesion and phagocytosis and have essential roles in regulating nerve repair. Quantitative real-time polymerase chain reaction assays were used to validate the expression of pivotal genes. Finally, nine pivotal genes that contribute to facial nerve recovery were identified, including Arhgap30, Akr1b8, C5ar1, Csf2ra, Dock2, Hcls1, Inpp5d, Sla, and Spi1. Primary Schwann cells were isolated from the sciatic nerve of neonatal rats. After knocking down Akr1b8 in Schwann cells with an Akr1b8-specific small interfering RNA plasmid, expression levels of monocyte chemoattractant protein-1 and interleukin-6 were decreased, while cell proliferation and migration were not obviously altered. These findings suggest that Akr1b8 likely regulates the interaction between Schwann cells and macrophages through regulation of cytokine expression to promote facial nerve regeneration. This study is the first to reveal a transcriptome change in the facial nerve trunk after facial nerve injury, thereby revealing the potential mechanism underlying repair of facial nerve injury. This study was approved by the Animal Ethics Committee of Nantong University, China in 2018 (approval No. S20180923-007). 
    Related Articles | Metrics
    Magnet-targeted delivery of bone marrow-derived mesenchymal stem cells improves therapeutic efficacy following hypoxic-ischemic brain injury
    Chuang Sun, #, Ao-Dan Zhang, #, Hong-Hai Chen, Jie Bian, Zheng-Juan Liu
    2021, 16 (11):  2324-2329.  doi: 10.4103/1673-5374.310942
    Abstract ( 110 )   PDF (1993KB) ( 123 )   Save
    Stem cell transplantation may represent a feasible therapeutic option for the recovery of neurological function in children with hypoxic-ischemic brain injury; however, the therapeutic efficacy of bone marrow-derived mesenchymal stem cells largely depends on the number of cells that are successfully transferred to the target. Magnet-targeted drug delivery systems can use a specific magnetic field to attract the drug to the target site, increasing the drug concentration. In this study, we found that the double-labeling using superparamagnetic iron oxide nanoparticle and poly-L-lysine (SPIO-PLL) of bone marrow-derived mesenchymal stem cells had no effect on cell survival but decreased cell proliferation 48 hours after labeling. Rat models of hypoxic-ischemic brain injury were established by ligating the left common carotid artery. One day after modeling, intraventricular and caudal vein injections of 1 × 105 SPIO-PLL-labeled bone marrow-derived mesenchymal stem cells were performed. Twenty-four hours after the intraventricular injection, magnets were fixed to the left side of the rats’ heads for 2 hours. Intravoxel incoherent motion magnetic resonance imaging revealed that the perfusion fraction and the diffusion coefficient of rat brain tissue were significantly increased in rats treated with SPIO-PLL-labeled cells through intraventricular injection combined with magnetic guidance, compared with those treated with SPIO-PLL-labeled cells through intraventricular or tail vein injections without magnetic guidance. Hematoxylin-eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining revealed that in rats treated with SPIO-PLL-labeled cells through intraventricular injection under magnetic guidance, cerebral edema was alleviated, and apoptosis was decreased. These findings suggest that targeted magnetic guidance can be used to improve the therapeutic efficacy of bone marrow-derived mesenchymal stem cell transplantation for hypoxic-ischemic brain injury. This study was approved by the Animal Care and Use Committee of The Second Hospital of Dalian Medical University, China (approval No. 2016-060) on March 2, 2016.
    Related Articles | Metrics
    Magnesium acetyltaurate prevents retinal damage and visual impairment in rats through suppression of NMDA-induced upregulation of NF-κB, p53 and AP-1 (c-Jun/c-Fos)
    Lidawani Lambuk, Igor Iezhitsa, , Renu Agarwal, Puneet Agarwal, Anna Peresypkina, Anna Pobeda, Nafeeza Mohd Ismail
    2021, 16 (11):  2330-2344.  doi: 10.4103/1673-5374.310691
    Abstract ( 152 )   PDF (5768KB) ( 277 )   Save
    Magnesium acetyltaurate (MgAT) has been shown to have a protective effect against N-methyl-D-aspartate (NMDA)-induced retinal cell apoptosis. The current study investigated the involvement of nuclear factor kappa-B (NF-κB), p53 and AP-1 family members (c-Jun/c-Fos) in neuroprotection by MgAT against NMDA-induced retinal damage. In this study, Sprague-Dawley rats were randomized to undergo intravitreal injection of vehicle, NMDA or MgAT as pre-treatment to NMDA. Seven days after injections, retinal ganglion cells survival was detected using retrograde labelling with fluorogold and BRN3A immunostaining. Functional outcome of retinal damage was assessed using electroretinography, and the mechanisms underlying antiapoptotic effect of MgAT were investigated through assessment of retinal gene expression of NF-κB, p53 and AP-1 family members (c-Jun/c-Fos) using reverse transcription-polymerase chain reaction. Retinal phospho-NF-κB, phospho-p53 and AP-1 levels were evaluated using western blot assay. Rat visual functions were evaluated using visual object recognition tests. Both retrograde labelling and BRN3A immunostaining revealed a significant increase in the number of retinal ganglion cells in rats receiving intravitreal injection of MgAT compared with the rats receiving intravitreal injection of NMDA. Electroretinography indicated that pre-treatment with MgAT partially preserved the functional activity of NMDA-exposed retinas. MgAT abolished NMDA-induced increase of retinal phospho-NF-κB, phospho-p53 and AP-1 expression and suppressed NMDA-induced transcriptional activity of NF-κB, p53 and AP-1 family members (c-Jun/c-Fos). Visual object recognition tests showed that MgAT reduced difficulties in recognizing the visual cues (i.e. objects with different shapes) after NMDA exposure, suggesting that visual functions of rats were relatively preserved by pre-treatment with MgAT. In conclusion, pre-treatment with MgAT prevents NMDA induced retinal injury by inhibiting NMDA-induced neuronal apoptosis via downregulation of transcriptional activity of NF-κB, p53 and AP-1-mediated c-Jun/c-Fos. The experiments were approved by the Animal Ethics Committee of Universiti Teknologi MARA (UiTM), Malaysia, UiTM CARE No 118/2015 on December 4, 2015 and UiTM CARE No 220/7/2017 on December 8, 2017 and Ethics Committee of Belgorod State National Research University, Russia, No 02/20 on January 10, 2020.
    Related Articles | Metrics