Loading...

Table of Content

    15 October 2022, Volume 17 Issue 10 Previous Issue   
    For Selected: Toggle Thumbnails
    Polyphenols as potential enhancers of stem cell therapy against neurodegeneration
    Diana Rodríguez-Vera, Antonio Abad-García, Nancy Vargas-Mendoza, Rodolfo Pinto-Almazán, Eunice D. Farfán-García, José A. Morales-González, Marvin A. Soriano-Ursúa
    2022, 17 (10):  2093-2101.  doi: 10.4103/1673-5374.335826
    Abstract ( 390 )   PDF (2384KB) ( 145 )   Save
    The potential of polyphenols for treating chronic-degenerative diseases (particularly neurodegenerative diseases) is attractive. However, the selection of the best polyphenol for each treatment, the mechanisms by which they act, and their efficacy are frequently discussed. In this review, the basics and the advances in the field, as well as suggestions for using natural and synthetic polyphenols alone or in a combinatorial strategy with stem cell assays, are compiled and discussed. Thus, stem cells exhibit several responses when polyphenols are added to their environment, which could provide us with knowledge for advancing the elucidation of the origin of neurodegeneration. But also, polyphenols are being included in the innovative strategies of novel therapies for treating neurodegenerative diseases as well as metabolic diseases related to neurodegeneration. In this regard, flavonoid compounds are suggested as the best natural polyphenols due to their several mechanisms for acting in ameliorative effects; but increasing reports are involving other polyphenols. Even if some facts limiting bioactivity prevent them from conventional use, some natural polyphenols and derivatives hold the promise for being improved compounds, judged by their induced effects. The current results suggest polyphenols as enhancers of stem cell therapy against the targeted diseases.
    Related Articles | Metrics
    Role of platelet-derived extracellular vesicles in traumatic brain injury-induced coagulopathy and inflammation
    Liang Liu, Quan-Jun Deng
    2022, 17 (10):  2102-2107.  doi: 10.4103/1673-5374.335825
    Abstract ( 234 )   PDF (1031KB) ( 82 )   Save
    Extracellular vesicles are composed of fragments of exfoliated plasma membrane, organelles or nuclei and are released after cell activation, apoptosis or destruction. Platelet-derived extracellular vesicles are the most abundant type of extracellular vesicle in the blood of patients with traumatic brain injury. Accumulated laboratory and clinical evidence shows that platelet-derived extracellular vesicles play an important role in coagulopathy and inflammation after traumatic brain injury. This review discusses the recent progress of research on platelet-derived extracellular vesicles in coagulopathy and inflammation and the potential of platelet-derived extracellular vesicles as therapeutic targets for traumatic brain injury.
    Related Articles | Metrics
    Biomaterial and tissue-engineering strategies for the treatment of brain neurodegeneration
    Bridget Martinez, Philip V. Peplow
    2022, 17 (10):  2108-2116.  doi: 10.4103/1673-5374.336132
    Abstract ( 168 )   PDF (611KB) ( 79 )   Save
    The incidence of neurodegenerative diseases is increasing due to changing age demographics and the incidence of sports-related traumatic brain injury is tending to increase over time. Currently approved medicines for neurodegenerative diseases only temporarily reduce the symptoms but cannot cure or delay disease progression. Cell transplantation strategies offer an alternative approach to facilitating central nervous system repair, but efficacy is limited by low in vivo survival rates of cells that are injected in suspension. Transplanting cells that are attached to or encapsulated within a suitable biomaterial construct has the advantage of enhancing cell survival in vivo. A variety of biomaterials have been used to make constructs in different types that included nanoparticles, nanotubes, microspheres, microscale fibrous scaffolds, as well as scaffolds made of gels and in the form of micro-columns. Among these, Tween 80-methoxy poly(ethylene glycol)-poly(lactic-co-glycolic acid) nanoparticles loaded with rhynchophylline had higher transport across a blood-brain barrier model and decreased cell death in an in vitro model of Alzheimer’s disease than rhynchophylline or untreated nanoparticles with rhynchophylline. In an in vitro model of Parkinson’s disease, trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin had a similar protective ability as free non-Fe hemin. A positive effect on neuron survival in several in vivo models of Parkinson’s disease was associated with the use of biomaterial constructs such as trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin, carbon nanotubes with olfactory bulb stem cells, poly(lactic-co-glycolic acid) microspheres with attached DI-MIAMI cells, ventral midbrain neurons mixed with short fibers of poly-(L-lactic acid) scaffolds and reacted with xyloglucan with/without glial-derived neurotrophic factor, ventral midbrain neurons mixed with Fmoc-DIKVAV hydrogel with/without glial-derived neurotrophic factor. Further studies with in vivo models of Alzheimer’s disease and Parkinson’s disease are warranted especially using transplantation of cells in agarose micro-columns with an inner lumen filled with an appropriate extracellular matrix material.
    Related Articles | Metrics
    Graphene and graphene-based materials in axonal repair of spinal cord injury
    Shi-Xin Wang, Yu-Bao Lu, Xue-Xi Wang, Yan Wang, Yu-Jun Song, Xiao Wang, Munkhtuya Nyamgerelt
    2022, 17 (10):  2117-2125.  doi: 10.4103/1673-5374.335822
    Abstract ( 172 )   PDF (580KB) ( 550 )   Save
    Graphene and graphene-based materials have the ability to induce stem cells to differentiate into neurons, which is necessary to overcome the current problems faced in the clinical treatment of spinal cord injury. This review summarizes the advantages of graphene and graphene-based materials (in particular, composite materials) in axonal repair after spinal cord injury. These materials have good histocompatibility, and mechanical and adsorption properties that can be targeted to improve the environment of axonal regeneration. They also have good conductivity, which allows them to make full use of electrical nerve signal stimulation in spinal cord tissue to promote axonal regeneration. Furthermore, they can be used as carriers of seed cells, trophic factors, and drugs in nerve tissue engineering scaffolds to provide a basis for constructing a local microenvironment after spinal cord injury. However, to achieve clinical adoption of graphene and graphene-based materials for the repair of spinal cord injury, further research is needed to reduce their toxicity.
    Related Articles | Metrics
    WNT/β-catenin pathway and circadian rhythms in obsessive-compulsive disorder
    Alexandre Vallée, Yves Lecarpentier, Jean-Noël Vallée
    2022, 17 (10):  2126-2130.  doi: 10.4103/1673-5374.332133
    Abstract ( 267 )   PDF (458KB) ( 109 )   Save
    The neuropsychiatric disease named obsessive-compulsive disorder is composed by obsessions and/or compulsions. Obsessive-compulsive disorder etiologies are undefined. However, numerous mechanisms in several localizations are implicated. Some studies showed that both glutamate, inflammatory factors and oxidative stress could have main functions in obsessive-compulsive disorder. Glycogen synthase kinase-3β, the major negative controller of the WNT/β-catenin pathway is upregulated in obsessive-compulsive disorder. In obsessive-compulsive disorder, some studies presented the actions of the different circadian clock genes. WNT/β-catenin pathway and circadian clock genes appear to be intricate. Thus, this review focuses on the interaction between circadian clock genes and the WNT/β-catenin pathway in obsessive-compulsive disorder.
    Related Articles | Metrics
    Unraveling pathological mechanisms in neurological disorders: the impact of cell-based and organoid models
    Jake Langlie, Rahul Mittal, Ariel Finberg, Nathalie B. Bencie, Jeenu Mittal, Hossein Omidian, Yadollah Omidi, Adrien A. Eshraghi
    2022, 17 (10):  2131-2140.  doi: 10.4103/1673-5374.335836
    Abstract ( 150 )   PDF (12653KB) ( 52 )   Save
    Cell-based models are a promising tool in deciphering the molecular mechanisms underlying the pathogenesis of neurological disorders as well as aiding in the discovery and development of future drug therapies. The greatest challenge is creating cell-based models that encapsulate the vast phenotypic presentations as well as the underlying genotypic etiology of these conditions. In this article, we discuss the recent advancements in cell-based models for understanding the pathophysiology of neurological disorders. We reviewed studies discussing the progression of cell-based models to the advancement of three-dimensional models and organoids that provide a more accurate model of the pathophysiology of neurological disorders in vivo. The better we understand how to create more precise models of the neurological system, the sooner we will be able to create patient-specific models and large libraries of these neurological disorders. While three-dimensional models can be used to discover the linking factors to connect the varying phenotypes, such models will also help to understand the early pathophysiology of these neurological disorders and how they are affected by their environment. The three-dimensional cell models will allow us to create more specific treatments and uncover potentially preventative measures in neurological disorders such as autism spectrum disorder, Parkinson’s disease, Alzheimer’s disease, and amyotrophic lateral sclerosis.
    Related Articles | Metrics
    Respiratory plasticity following spinal cord injury: perspectives from mouse to man
    Katherine C. Locke, Margo L. Randelman, Daniel J. Hoh, Lyandysha V. Zholudeva, Michael A. Lane
    2022, 17 (10):  2141-2148.  doi: 10.4103/1673-5374.335839
    Abstract ( 203 )   PDF (5075KB) ( 76 )   Save
    The study of respiratory plasticity in animal models spans decades. At the bench, researchers use an array of techniques aimed at harnessing the power of plasticity within the central nervous system to restore respiration following spinal cord injury. This field of research is highly clinically relevant. People living with cervical spinal cord injury at or above the level of the phrenic motoneuron pool at spinal levels C3–C5 typically have significant impairments in breathing which may require assisted ventilation. Those who are ventilator dependent are at an increased risk of ventilator-associated co-morbidities and have a drastically reduced life expectancy. Pre-clinical research examining respiratory plasticity in animal models has laid the groundwork for clinical trials. Despite how widely researched this injury is in animal models, relatively few treatments have broken through the preclinical barrier. The three goals of this present review are to define plasticity as it pertains to respiratory function post-spinal cord injury, discuss plasticity models of spinal cord injury used in research, and explore the shift from preclinical to clinical research. By investigating current targets of respiratory plasticity research, we hope to illuminate preclinical work that can influence future clinical investigations and the advancement of treatments for spinal cord injury.
    Related Articles | Metrics
    Exploring the role of interleukin-27 as a regulator of neuronal survival in central nervous system diseases
    Andrea N. Nortey, Kimberly N. Garces, Abigail S. Hackam
    2022, 17 (10):  2149-2152.  doi: 10.4103/1673-5374.336134
    Abstract ( 170 )   PDF (485KB) ( 49 )   Save
    Interleukin-27 is a pleiotropic cytokine that is involved in tissue responses to infection, cell stress, neuronal disease, and tumors. Recent studies in various tissues indicate that interleukin-27 has complex activating and inhibitory properties in innate and acquired immunity. The availability of recombinant interleukin-27 protein and mice with genetic deletions of interleukin-27, its receptors and signaling mediators have helped define the role of interleukin-27 in neurodegenerative diseases. Interleukin-27 has been well-characterized as an important regulator of T cell activation and differentiation that enhances or suppresses T cell responses in autoimmune conditions in the central nervous system. Evidence is also accumulating that interleukin-27 has neuroprotective activities in the retina and brain. Interleukin-27 is secreted from and binds to infiltrating microglia, macrophage, astrocytes, and even neurons and it promotes neuronal survival by regulating pro- and anti-inflammatory cytokines, neuroinflammatory pathways, oxidative stress, apoptosis, autophagy, and epigenetic modifications. However, interleukin-27 can have the opposite effect and induce inflammation and cell death in certain situations. In this review, we describe the current understanding of regulatory activities of interleukin-27 on cell survival and inflammation and discuss its mechanisms of action in the brain, spinal cord, and retina. We also review evidence for and against the therapeutic potential of interleukin-27 for dampening harmful neuroinflammatory responses in central nervous system diseases.
    Related Articles | Metrics
    Potential significance of CX3CR1 dynamics in stress resilience against neuronal disorders
    Koichi Inoue
    2022, 17 (10):  2153-2156.  doi: 10.4103/1673-5374.335831
    Abstract ( 381 )   PDF (413KB) ( 109 )   Save
    Recent findings have implicated inflammatory responses in the central nervous system in a variety of neuropsychiatric and neurodegenerative diseases, and the understanding and control of immunological responses could be a major factor of future therapeutic strategies for neurological disorders. Microglia, derived from myelogenous cells, respond to a number of stimuli and make immune responses, resulting in a prominent role as cells that act on inflammation in the central nervous system. Fractalkine (FKN or CX3CL1) signaling is an important factor that influences the inflammatory response of microglia. The receptor for FKN, CX3CR1, is usually expressed in microglia in the brain, and therefore the inflammatory response of microglia is modified by FKN. Reportedly, FKN often suppresses inflammatory responses in microglia and activation of its receptor may be effective in the treatment of inflammatory neurological disorders. However, it has also been suggested that inflammatory responses facilitated by FKN signaling aggravate neurological disorders. Thus, further studies are still required to resolve the conflicting interpretation of the protective or deleterious contribution of microglial FKN signaling. Yet notably, regulation of FKN signaling has recently been shown to be beneficial in the treatment of human diseases, although not neurological diseases. In addition, a CX3CR1 inhibitor has been developed and successfully tested in animal models, and it is expected to be in human clinical trials in the future. In this review, I describe the potential therapeutic consideration of microglial CX3CR1 dynamics through altered FKN signaling.
    Related Articles | Metrics
    Brain delivering RNA-based therapeutic strategies by targeting mTOR pathway for axon regeneration after central nervous system injury
    Ming-Xi Li, Jing-Wen Weng, Eric S. Ho, Shing Fung Chow, Chi Kwan Tsang
    2022, 17 (10):  2157-2165.  doi: 10.4103/1673-5374.335830
    Abstract ( 252 )   PDF (967KB) ( 112 )   Save
    Injuries to the central nervous system (CNS) such as stroke, brain, and spinal cord trauma often result in permanent disabilities because adult CNS neurons only exhibit limited axon regeneration. The brain has a surprising intrinsic capability of recovering itself after injury. However, the hostile extrinsic microenvironment significantly hinders axon regeneration. Recent advances have indicated that the inactivation of intrinsic regenerative pathways plays a pivotal role in the failure of most adult CNS neuronal regeneration. Particularly, substantial evidence has convincingly demonstrated that the mechanistic target of rapamycin (mTOR) signaling is one of the most crucial intrinsic regenerative pathways that drive axonal regeneration and sprouting in various CNS injuries. In this review, we will discuss the recent findings and highlight the critical roles of mTOR pathway in axon regeneration in different types of CNS injury. Importantly, we will demonstrate that the reactivation of this regenerative pathway can be achieved by blocking the key mTOR signaling components such as phosphatase and tensin homolog (PTEN). Given that multiple mTOR signaling components are endogenous inhibitory factors of this pathway, we will discuss the promising potential of RNA-based therapeutics which are particularly suitable for this purpose, and the fact that they have attracted substantial attention recently after the success of coronavirus disease 2019 vaccination. To specifically tackle the blood-brain barrier issue, we will review the current technology to deliver these RNA therapeutics into the brain with a focus on nanoparticle technology. We will propose the clinical application of these RNA-mediated therapies in combination with the brain-targeted drug delivery approach against mTOR signaling components as an effective and feasible therapeutic strategy aiming to enhance axonal regeneration for functional recovery after CNS injury.
    Related Articles | Metrics
    Status of precision medicine approaches to traumatic brain injury
    Sahithi Reddi, Smita Thakker-Varia, Janet Alder, Anna O. Giarratana
    2022, 17 (10):  2166-2171.  doi: 10.4103/1673-5374.335824
    Abstract ( 142 )   PDF (1669KB) ( 62 )   Save
    Traumatic brain injury (TBI) is a serious condition in which trauma to the head causes damage to the brain, leading to a disruption in brain function. This is a significant health issue worldwide, with around 69 million people suffering from TBI each year. Immediately following the trauma, damage occurs in the acute phase of injury that leads to the primary outcomes of the TBI. In the hours-to-days that follow, secondary damage can also occur, leading to chronic outcomes. TBIs can range in severity from mild to severe, and can be complicated by the fact that some individuals sustain multiple TBIs, a risk factor for worse long-term outcomes. Although our knowledge about the pathophysiology of TBI has increased in recent years, unfortunately this has not been translated into effective clinical therapies. The U.S. Food and Drug Administration has yet to approve any drugs for the treatment of TBI; current clinical treatment guidelines merely offer supportive care. Outcomes between individuals greatly vary, which makes the treatment for TBI so challenging. A blow of similar force can have only mild, primary outcomes in one individual and yet cause severe, chronic outcomes in another. One of the reasons that have been proposed for this differential response to TBI is the underlying genetic differences across the population. Due to this, many researchers have begun to investigate the possibility of using precision medicine techniques to address TBI treatment. In this review, we will discuss the research detailing the identification of genetic risk factors for worse outcomes after TBI, and the work investigating personalized treatments for these higher-risk individuals. We highlight the need for further research into the identification of higher-risk individuals and the development of personalized therapies for TBI.
    Related Articles | Metrics
    Diabetic corneal neuropathy as a surrogate marker for diabetic peripheral neuropathy
    Wei Zheng So, Natalie Shi Qi Wong, Hong Chang Tan, Molly Tzu Yu Lin, Isabelle Xin Yu Lee, Jodhbir S. Mehta, Yu-Chi Liu
    2022, 17 (10):  2172-2178.  doi: 10.4103/1673-5374.327364
    Abstract ( 208 )   PDF (1239KB) ( 73 )   Save
    Diabetic neuropathy is a prevalent microvascular complication of diabetes mellitus, affecting nerves in all parts of the body including corneal nerves and peripheral nervous system, leading to diabetic corneal neuropathy and diabetic peripheral neuropathy, respectively. Diabetic peripheral neuropathy is diagnosed in clinical practice using electrophysiological nerve conduction studies, clinical scoring, and skin biopsies. However, these diagnostic methods have limited sensitivity in detecting small-fiber disease, hence they do not accurately reflect the status of diabetic neuropathy. More recently, analysis of alterations in the corneal nerves has emerged as a promising surrogate marker for diabetic peripheral neuropathy. In this review, we will discuss the relationship between diabetic corneal neuropathy and diabetic peripheral neuropathy, elaborating on the foundational aspects of each: pathogenesis, clinical presentation, evaluation, and management. We will further discuss the relevance of diabetic corneal neuropathy in detecting the presence of diabetic peripheral neuropathy, particularly early diabetic peripheral neuropathy; the correlation between the severity of diabetic corneal neuropathy and that of diabetic peripheral neuropathy; and the role of diabetic corneal neuropathy in the stratification of complications of diabetic peripheral neuropathy.
    Related Articles | Metrics
    Role of adipose tissue grafting and adipose-derived stem cells in peripheral nerve surgery
    Tiam M. Saffari, Sara Saffari, Krishna S. Vyas, Samir Mardini, Alexander Y. Shin
    2022, 17 (10):  2179-2184.  doi: 10.4103/1673-5374.336870
    Abstract ( 176 )   PDF (3264KB) ( 288 )   Save
    The application of autologous fat grafting in reconstructive surgery is commonly used to improve functional form. This review aims to provide an overview of the scientific evidence on the biology of adipose tissue, the role of adipose-derived stem cells, and the indications of adipose tissue grafting in peripheral nerve surgery. Adipose tissue is easily accessible through the lower abdomen and inner thighs. Non-vascularized adipose tissue grafting does not support oxidative and ischemic stress, resulting in variable survival of adipocytes within the first 24 hours. Enrichment of adipose tissue with a stromal vascular fraction is purported to increase the number of adipose-derived stem cells and is postulated to augment the long-term stability of adipose tissue grafts. Basic science nerve research suggests an increase in nerve regeneration and nerve revascularization, and a decrease in nerve fibrosis after the addition of adipose-derived stem cells or adipose tissue. In clinical studies, the use of autologous lipofilling is mostly applied to secondary carpal tunnel release revisions with promising results. Since the use of adipose-derived stem cells in peripheral nerve reconstruction is relatively new, more studies are needed to explore safety and long-term effects on peripheral nerve regeneration. The Food and Drug Administration stipulates that adipose-derived stem cell transplantation should be minimally manipulated, enzyme-free, and used in the same surgical procedure, e.g. adipose tissue grafts that contain native adipose-derived stem cells or stromal vascular fraction. Future research may be shifted towards the use of tissue-engineered adipose tissue to create a supportive microenvironment for autologous graft survival. Shelf-ready alternatives could be enhanced with adipose-derived stem cells or growth factors and eliminate the need for adipose tissue harvest.
    Related Articles | Metrics
    Basic mechanisms of peripheral nerve injury and treatment via electrical stimulation
    Xiao-Lei Chu, Xi-Zi Song, Qi Li, Yu-Ru Li, Feng He, Xiao-Song Gu, Dong Ming
    2022, 17 (10):  2185-2193.  doi: 10.4103/1673-5374.335823
    Abstract ( 621 )   PDF (1382KB) ( 261 )   Save
    Previous studies on the mechanisms of peripheral nerve injury (PNI) have mainly focused on the pathophysiological changes within a single injury site. However, recent studies have indicated that within the central nervous system, PNI can lead to changes in both injury sites and target organs at the cellular and molecular levels. Therefore, the basic mechanisms of PNI have not been comprehensively understood. Although electrical stimulation was found to promote axonal regeneration and functional rehabilitation after PNI, as well as to alleviate neuropathic pain, the specific mechanisms of successful PNI treatment are unclear. We summarize and discuss the basic mechanisms of PNI and of treatment via electrical stimulation. After PNI, activity in the central nervous system (spinal cord) is altered, which can limit regeneration of the damaged nerve. For example, cell apoptosis and synaptic stripping in the anterior horn of the spinal cord can reduce the speed of nerve regeneration. The pathological changes in the posterior horn of the spinal cord can modulate sensory abnormalities after PNI. This can be observed in cases of ectopic discharge of the dorsal root ganglion leading to increased pain signal transmission. The injured site of the peripheral nerve is also an important factor affecting post-PNI repair. After PNI, the proximal end of the injured site sends out axial buds to innervate both the skin and muscle at the injury site. A slow speed of axon regeneration leads to low nerve regeneration. Therefore, it can take a long time for the proximal nerve to reinnervate the skin and muscle at the injured site. From the perspective of target organs, long-term denervation can cause atrophy of the corresponding skeletal muscle, which leads to abnormal sensory perception and hyperalgesia, and finally, the loss of target organ function. The mechanisms underlying the use of electrical stimulation to treat PNI include the inhibition of synaptic stripping, addressing the excessive excitability of the dorsal root ganglion, alleviating neuropathic pain, improving neurological function, and accelerating nerve regeneration. Electrical stimulation of target organs can reduce the atrophy of denervated skeletal muscle and promote the recovery of sensory function. Findings from the included studies confirm that after PNI, a series of physiological and pathological changes occur in the spinal cord, injury site, and target organs, leading to dysfunction. Electrical stimulation may address the pathophysiological changes mentioned above, thus promoting nerve regeneration and ameliorating dysfunction.
    Related Articles | Metrics
    Functional in vivo assessment of stem cell-secreted pro-oligodendroglial factors
    Jessica Schira-Heinen, Iria Samper Agrelo, Veronica Estrada, Patrick Küry
    2022, 17 (10):  2194-2196.  doi: 10.4103/1673-5374.335800
    Abstract ( 152 )   PDF (781KB) ( 60 )   Save
    The role of adult neural stem cells (NSCs) in demyelinating diseases of the central nervous system (CNS): Multipotent NSCs hold great potential for cell replacement in diseases and upon injury of the CNS. Originating from radial glial cells during nervous system development, adult NSCs are localized in the subgranular zone of the hippocampus and the subventricular zone (SVZ) of the lateral brain ventricles, the main neurogenic zones of the adult brain. Hippocampal precursor cells (type 1 cells) exhibiting properties of radial glial cells give rise to granule neurons through distinct intermediate precursor cells, and integrate into the hippocampal circuitry [reviewed by Kempermann et al. (2015)]. Likewise, under physiological conditions, neuron generation by mouse SVZ-derived NSCs (also known as type B cells) is the predominant cell fate, which thereby results in large numbers of transient amplifying precursor cells (also known as type C cells) which in turn differentiate into neuroblasts (type A cells). These cells migrate along the rostral migratory stream into the olfactory bulb where they undergo maturation into local interneurons. The structure of the rodent SVZ differs from that of the human SVZ since the proliferative capacity is reduced, and migration of neuroblasts is a rare event in adult humans [reviewed by Lim and Alvarez-Buylla (2016)].
    Related Articles | Metrics
    iGluR expression in the hippocampal formation, entorhinal cortex, and superior temporal gyrus in Alzheimer’s disease
    Jason HY Yeung, Henry J Waldvogel, Richard LM Faull, Andrea Kwakowsky
    2022, 17 (10):  2197-2199.  doi: 10.4103/1673-5374.335804
    Abstract ( 276 )   PDF (571KB) ( 113 )   Save
    Alzheimer’s disease (AD) constitutes the largest proportion of dementia worldwide, with a significant associated medical burden. The major pathological hallmarks of AD include the gradual accumulation and deposition of amyloid-beta (Aβ) plaques and hyperphosphorylated tau protein (Revett et al., 2013). Whilst investigations centered around the tau and Aβ hypothesis have been the main focus for the previous decades, the lack of therapeutic solutions has pushed for research into other potential therapeutic targets. Pathological alterations in the glutamatergic system have been postulated to play a central role in the pathogenesis of AD, with intimate downstream and upstream interactions with both Aβ and tau protein (Revett et al., 2013; Yeung et al., 2020a, b; Kwakowsky et al., 2021). Aβ1–42 toxicity is mediated in part by N-methyl-D-aspartate receptor (NMDAR) overactivation resulting in elevated intracellular calcium (Ca2+) and subsequent enzyme-induced neuronal death, whilst NMDAR activation has been shown to increase hyper-phosphorylation of neurofibrillary tau (Revett et al., 2013). The complex interrelationship between Aβ1–42, neurofibrillary tau, and the glutamatergic system is still an area of continuing development. A significant disruption of glutamatergic neurons has been well documented in AD, and the subsequent excitatory-inhibitory balance disturbance can potentially contribute to the memory and learning deficits that are characteristic of this condition (Revett et al., 2013; Jurado, 2018; Yeung et al., 2020a, b, 2021; Babaei, 2021; Kwakowsky et al., 2021).
    Related Articles | Metrics
    Synergy of human platelet lysate and laminin to enhance the neurotrophic effect of human adipose-derived stem cells
    Pietro G di Summa, Srinivas Madduri
    2022, 17 (10):  2200-2202.  doi: 10.4103/1673-5374.335797
    Abstract ( 173 )   PDF (2020KB) ( 60 )   Save
    Despite the spontaneous regenerative capacity of the peripheral nervous system, clinical nerve repair often results in poor functional recovery with the high socio-economic burden. In presence of peripheral nerve injuries and when distal nerve transfers are not possible, nerve autografts, are the solution of choice to cross a critical nerve gap. However, these are associated with donor site morbidity and offer only suboptimal functional recovery. Thus, the need to improve the rate of effective regeneration has directed research attention towards stem cell therapy and nerve tissue engineering.
    Related Articles | Metrics
    Exploiting Caenorhabditis elegans to discover human gut microbiota-mediated intervention strategies in protein conformational diseases
    Daniel M. Czyż
    2022, 17 (10):  2203-2204.  doi: 10.4103/1673-5374.335788
    Abstract ( 173 )   PDF (508KB) ( 55 )   Save
    Age-dependent protein-conformational diseases (PCDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), or amyotrophic lateral sclerosis (ALS), are characterized by misfolding and aggregation of metastable proteins present within the proteome of the affected individual. Recent evidence supports the notion that bacteria and bacterial products may be affecting the stability of these culprit host proteins and therefore influence disease progression and perhaps even its onset. Although specific culprit proteins are associated with each disease (e.g., Aβ in AD, α-synuclein in PD, and TDP-43 in ALS), bacteria found to affect these diseases do not seem to differentiate between these specific proteins but likely affect host proteostasis in general, leading to misfolding of metastable proteins encoded within the proteome. The evidence that supports this hypothesis comes from studies where a single bacterial genus was linked to multiple PCDs. For example, a decrease in Prevotella spp. is linked to constipation, a condition that precedes PD motor symptoms by as much as 20 or more years (Savica et al., 2009; Zhu et al., 2014). Additionally, the abundance of Prevotella found in PD patients negatively correlates with the severity of the disease (Jin et al., 2019). Prevotella abundance was also lower in an AD mouse model (Shen et al., 2017) and ALS patients (Hertzberg et al., 2021), suggesting that this genus may provide protection against proteotoxicity. However, more research has to concentrate on Prevotella to understand its role in PCDs, as other studies have seen increased abundance in affected patients (Guo et al., 2021). Such discrepancy could be attributed to the large number of Prevotella species that may play diverse roles in disease pathogenesis, and since most studies looked at the genus level, the effect of individual species may provide additional clues. A recent study found two Prevotella species (i.e., disiens and corporis) that significantly suppressed polyglutamine protein aggregation in the Caenorhabditis elegans (C. elegans) model (Walker et al., 2021), suggesting that these specific species may enhance host proteostasis and protect the host against protein misfolding and aggregation. What is most promising is that the effect of Prevotella spp. on host proteostasis is observed across organisms, including worms, mice, and humans; therefore, less expensive, and manageable models, such as C. elegans, can be used as a discovery tool. The example of Prevotella and its potential role in the suppression of various PCDs demonstrates that bacteria affect host proteostasis and may be capable of influencing disease pathogenicity mediated by aggregation-prone proteins.
    Related Articles | Metrics
    How useful are biomarkers for the diagnosis of Alzheimer’s disease and especially for its therapy?
    Marta Valenza, Caterina Scuderi
    2022, 17 (10):  2205-2207.  doi: 10.4103/1673-5374.335791
    Abstract ( 155 )   PDF (987KB) ( 58 )   Save
    Alzheimer’s disease key facts: Alzheimer’s disease (AD) is a slowly progressive neurodegenerative disease with no available effective treatment. It is possible to distinguish an early-onset AD that affects a limited number of subjects of young age, and a sporadic or late-onset form of the disease that affects the vast majority of subjects who are diagnosed with AD. As life expectancy has increased considerably over the past century, the number of people diagnosed with AD has grown exponentially. So, AD and AD-related pathologies represent a huge social and economic burden. The number of individuals waiting for effective disease-modifying therapy is impressive. It is estimated that 50 million people worldwide live with dementia, the majority of these cases are caused by AD (World Health Organization, 2021). In the US about 6 million individuals are living with AD, and more than 9 million people are in EU member states (OECD and European Union, 2020). The costs of health care and long-term care are substantial. Given this massive societal impact, enormous efforts have been made to understand the pathogenetic mechanisms of the disease with the hope of identifying new targets and, therefore, developing effective drugs. However, despite huge preclinical and clinical scientific efforts, therapeutic advances are truly modest, and the clinical practice is still anchored to the use of drugs modulating the cholinergic and glutamatergic systems.
    Related Articles | Metrics
    N-methyl-D-aspartate receptor functions altered by neuronal PTP1B activation in Alzheimer’s disease and schizophrenia models
    Alexandre F. R. Stewart, Hsiao-Huei Chen
    2022, 17 (10):  2208-2210.  doi: 10.4103/1673-5374.335793
    Abstract ( 154 )   PDF (815KB) ( 58 )   Save
    Glutamate is the main excitatory neurotransmitter in the brain and binds to two major classes of receptors, the α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) and the N-methyl-D-aspartate (NMDA) receptors. Unlike AMPA receptors that are immediately activated by glutamate release, NMDA receptors are blocked by magnesium and can only be activated by glutamate after membrane depolarization. Thus, NMDA receptors are only activated after repeated AMPA receptor activation by glutamate. NMDA receptors are, for the most part, calcium-permeable channels. Calcium influx through NMDA receptors modulates synaptic transmission in neurons based on prior history of excitation, and provides a means of scaling the strength of synapses required for Hebbian plasticity. NMDA receptors were first characterized in the post-synaptic membrane, where calcium influx controls AMPA receptor levels and activity-dependent gene expression. Tyrosine phosphorylation of postsynaptic NMDA receptors promotes calcium influx, whereas dephosphorylation of NMDA receptors causes their internalization and reduces calcium influx through NMDA receptors (Wang and Salter, 1994). More than 10 years ago, the striatal-enriched tyrosine phosphatase Step61 was tied to NMDA receptor dephosphorylation in the context of Alzheimer’s disease and amyloid-beta (Kurup et al., 2010).
    Related Articles | Metrics
    The integrative role of G protein-coupled receptor heterocomplexes in Parkinson’s disease
    Dasiel O. Borroto-Escuela, Kjell Fuxe
    2022, 17 (10):  2211-2212.  doi: 10.4103/1673-5374.335792
    Abstract ( 145 )   PDF (1570KB) ( 51 )   Save
    In the 1950s to 1970s, the research on Parkinson’s disease (PD) and its treatment had mainly been focused on the Nigro-striatal dopamine (DA) neurons as the major site of degeneration in this disease. It contributed to the search for drugs that restored DA transmission in this pathway and contributed to the introduction of L-DOPA and DA receptor agonists in its treatment (Borroto-Escuela and Fuxe, 2019).
    Related Articles | Metrics
    Brain-derived cell-free DNA
    Dean Southwood, Sanyukta Singh, Zac Chatterton
    2022, 17 (10):  2213-2214.  doi: 10.4103/1673-5374.335794
    Abstract ( 151 )   PDF (1317KB) ( 55 )   Save
    Following cell death, DNA can be released into the blood plasma and other body fluids in the form of cell-free DNA (cfDNA). These DNA fragments are typically ~167 bp in length, corresponding to the length of DNA wrapped around one nucleosome core (147 bp), plus DNA tails that survive endogenous DNase digestion (~10 bp). CfDNA is derived from a variety of sources, each having unique diagnostic applications. During pregnancy, fetal-derived cfDNA within an expectant mother’s blood plasma can diagnose fetal genetic abnormalities and is a well-established method for non-invasive prenatal testing. Cancer-derived cfDNA is also detectable within blood plasma by high sensitivity methods (e.g., NGS, ddPCR) and can accurately diagnose cancers by a minimally invasive blood test (Diehl et al., 2008). The short half-life of cfDNA (< 2 hours) also facilitates real-time measurement of disease burden, such as monitoring cancer relapse. There is now growing evidence of brain-derived cfDNA (bd-cfDNA) within the cerebrospinal fluid (CSF) and blood plasma. In this perspective, we review the present understanding, current challenges, and potential utility of bd-cfDNA.
    Related Articles | Metrics
    Aminopeptidase A and dipeptidyl peptidase 4: a pathogenic duo in Alzheimer’s disease?
    Frédéric Checler, Audrey Valverde
    2022, 17 (10):  2215-2217.  doi: 10.4103/1673-5374.335818
    Abstract ( 159 )   PDF (679KB) ( 42 )   Save
    The etiology of Alzheimer’s disease is far from being completely understood. Genetic approaches have helped in this matter and have greatly supported the view that the β-amyloid precursor protein (βAPP) could be at the center of gravity of the pathology. Thus, mutations responsible for autosomal dominant aggressive forms of Alzheimer’s disease (AD) are all harbored by either βAPP itself or by its cleaving enzyme presenilins 1/2 referred to as γ-secretase. It was therefore convincing to note that fully independent gene products harboring AD-linked mutations, all concur to modulate βAPP proteolytic processing. These genetic clues combined with a bulk of anatomical observations and cellular manipulations pointed to the role of amyloid-β (Aβ), the main biochemical component of senile plaques that accumulate at late stages of AD. Unfortunately, a series of clinical trials designed to either abolish Aβ production or neutralize it once produced have consistently failed (Checler et al., 2021). It remains that the genetic arguments are strong and that a key role of βAPP proteolytic maturation remains of actuality. One way to reconcile genetic evidence and clinical trials failure could be to envision that additional βAPP-derived products could contribute to AD etiology. A close evaluation of biogenesis and toxicity of such pathogenic βAPP-derived products, distinct from genuine Aβ could help to better understand AD etiology.
    Related Articles | Metrics
    Ubiquitin homeostasis disruption, a common cause of proteostasis collapse in amyotrophic lateral sclerosis?
    Christen G. Chisholm, Jeremy S. Lum, Natalie E. Farrawell, Justin J. Yerbury
    2022, 17 (10):  2218-2220.  doi: 10.4103/1673-5374.335786
    Abstract ( 160 )   PDF (826KB) ( 55 )   Save
    Amyotrophic lateral sclerosis (ALS) is associated with proteostasis collapse: ALS is an unrelenting neurodegenerative disease that is characterized by the loss of motor neurons in the brain and spinal cord, resulting in the progressive atrophy, and eventual paralysis, of skeletal muscles. Death due to respiratory failure usually occurs within 2–5 years from symptom onset. Approximately 90% of ALS cases are of unknown etiology and are termed sporadic ALS (sALS). The remaining 10% of ALS cases present with a family history (familial ALS; fALS) and are associated with genetic mutations in a range of over 20 functionally heterogeneous genes. Regardless of disease origin, the pathological hallmark of ALS is the accumulation of ubiquitylated protein inclusions in motor neurons and surrounding glial cells. The presence of these inclusions, compromised largely of misfolded and aggregated proteins, implies a collapse in proteostasis. Proteostasis refers to the maintenance of the proteome in a state of balance or equilibrium so that a cell can perform its proper function. Central to the maintenance of proteostasis are the predominant protein degradation pathways, the ubiquitin-proteasome system (UPS) and the autophagy system. In recent years, the identification of numerous ALS associated genes involved in protein degradation systems, including VCP, SQSTM1, UBQLN2, OPTN, TBK1, CCNF, DNAJC7 and CYLD have strengthened the association between proteostasis failure, in particular protein degradation, and ALS. The accumulating evidence implicating protein degradation pathways as a cause of ALS raises two critical questions; are mutations in genes within functional pathways distinct from protein degradation such as C9orf72, TARDBP, FUS and SOD1 in any way related to the UPS or autophagy dysfunction that causes ALS and why are motor neurons selectively affected? Understanding the unique vulnerability of motor neurons may provide additional insight into the pathogenesis of this currently incurable disease.
    Related Articles | Metrics
    Transcranial electrical stimulation in neurological disease
    Gregory L. Brown, Michael T. Brown
    2022, 17 (10):  2221-2222.  doi: 10.4103/1673-5374.335796
    Abstract ( 218 )   PDF (4426KB) ( 103 )   Save
    Increased lifespan is one of society’s greatest achievements, but this longevity increases the prevalence of diseases of aging, such as neurological disorders. Globally, neurological disorders are the leading cause of disability and the second leading cause of deaths (Feigin et al., 2019). Furthermore, these diseases affect people in low-, medium-, and high-income countries (Feigin et al., 2019). Current technology to modify neurological burden is scarce, which poses numerous challenges for healthcare, global policy, and economic stability (Feigin et al., 2019). To face these challenges, brain stimulation technology, such as transcranial electrical stimulation (TES), has displayed exciting potential. Antal et al. (2017) provide a detailed overview of the safety and application of TES. Commonly, electrodes are attached to the head and a weak current (e.g., 1–2 mA) is applied through the scalp, skull, and into the brain for 10–30 minutes to activate neurons (Antal et al., 2017). The technique is extremely safe with no serious adverse effects reported from thousands of sessions (Antal et al., 2017). The most common side effects are a tingling/itching sensation or redness at the stimulation site (Antal et al., 2017). However, these side effects can be minimized by reducing the electrode-skin impedance, slowly ramping up and ramping down TES, or using topical analgesics (Antal et al., 2017). The development of TES may repair neural dysfunction and stem the oncoming incidence of neurodegenerative disease.
    Related Articles | Metrics
    Novel insights into the pathogenesis of tendon injury: mechanotransduction and neuroplasticity
    Suellen Alessandra Soares de Moraes
    2022, 17 (10):  2223-2224.  doi: 10.4103/1673-5374.335802
    Abstract ( 220 )   PDF (528KB) ( 56 )   Save
    Tendon pathology is characterized by damage to the tendon structural integrity with disruption of collagen fibers (Nourissat et al., 2015). Acute tendon injuries show a macroscopic discontinuity, ranging from partial to complete tendon rupture. They involve inflammation and lead to loss of motion. In chronic conditions (or tendinopathy), symptoms include changes in both locomotor and sensorial functions of the tendon (Nourissat et al., 2015; Scott et al., 2020). Inconsistency in terminology for cases of painful tendon disorders is found, but recently the term tendinopathy was established in consensus as preferable for cases with persistent tendon pain and loss of function related to mechanical loading. This term excludes a problem in clinical practice – i.e., specification of the presence of a particular pathological or biochemical process (Scott et al., 2020).
    Related Articles | Metrics
    Zinc finger protein ZPR1: promising survival motor neuron protein-dependent modifier for the rescue of spinal muscular atrophy
    Juliana Cuartas, Laxman Gangwani
    2022, 17 (10):  2225-2227.  doi: 10.4103/1673-5374.335798
    Abstract ( 186 )   PDF (710KB) ( 56 )   Save
    Spinal muscular atrophy (SMA) is a neuromuscular disease caused by the homozygous mutation or deletion of the survival motor neuron 1 (SMN1) gene. A second copy, SMN2, is similar to SMN1, but produces only ~10% SMN protein because of a single-point mutation (C > T) in coding exon 7 causing a splicing defect which leads to the exclusion of exon 7, resulting in a majority (~90%) of transcripts lacking exon 7 that translate into mutant SMN (SMNΔ7) protein. SMA is caused by chronic low levels of SMN and is characterized by the degeneration of the spinal cord motor neurons leading to symmetrical skeletal muscle atrophy, respiratory failure, and death (Ahmad et al., 2012). Chronic low levels of SMN cause the accumulation of pathogenic R-loops and double-stranded breaks (DSBs) in DNA, leading to genomic instability and neurodegeneration in SMA (Kannan et al., 2018). The severity of SMA disease correlates inversely with SMN levels. The SMN2 gene is a promising target to produce higher levels of SMN by enhancing its expression. Cellular and molecular factors that may regulate the expression of SMN genes are slowly emerging, but precise molecular mechanisms which directly influence SMN expression are unclear. This perspective is focused on the potential role of the zinc finger protein ZPR1 as a molecular factor that may regulate the levels of mammalian SMN genes expression in vivo under the normal and disease conditions through a novel mechanism involving the resolution of R-loops that are formed during transcription (Kannan et al., 2020). The potential of ZPR1 as a therapeutic target for developing a new treatment is discussed in the context of currently available treatments for SMA.
    Related Articles | Metrics
    Complement activation kindles the transition of acute post-traumatic brain injury to a chronic inflammatory disease
    Andrew Erwood, Ali M. Alawieh
    2022, 17 (10):  2228-2229.  doi: 10.4103/1673-5374.335799
    Abstract ( 163 )   PDF (273KB) ( 69 )   Save
    Traumatic brain injury (TBI) remains a major cause of disability among young adults in both civilian and military settings contributing to a high burden on healthcare systems (Badhiwala et al., 2019). Sequel of TBI, even mild injuries, include motor and sensory dysfunction, neurocognitive decline, neuropsychiatric complications, as well as increased risk of neurodegenerative and neurovascular events such as Alzheimer’s disease and stroke (Breunig et al., 2013; Burke et al., 2013; Li et al., 2017). Despite the acute nature of the insult in TBI, pathological changes in the traumatized brain are better recognized as a chronic rather than an acute neurological disease, a phenomenon that remains under-investigated. Robust clinical data support the role of neuroinflammation in propagating neurodegenerative changes following TBI with a pivotal role of the complement system as an early trigger and chronic propagator of this response (Alawieh et al., 2018, 2021; Mallah et al., 2021). Hereby, we discuss how the role of complement pathways in different phases of injury after TBI was investigated using clinically relevant targeted complement inhibitors (Alawieh and Tomlinson, 2016; Alawieh et al., 2018, 2021; Mallah et al., 2021).
    Related Articles | Metrics
    Degeneration of retinal ganglion cells in hypoxic responses: hypoxia-inducible factor inhibition, a new therapeutic insight
    Deokho Lee, Hiromitsu Kunimi, Kazuno Negishi, Toshihide Kurihara
    2022, 17 (10):  2230-2231.  doi: 10.4103/1673-5374.335801
    Abstract ( 222 )   PDF (392KB) ( 64 )   Save
    Degeneration of retinal ganglion cells (RGCs) is one of the hallmarks of common optic neuropathies (Weinreb et al., 2014). Glaucoma, the most common optic neuropathy, is characterized by degeneration of RGCs. Acute angle-closure glaucoma is a serious ocular condition caused by a rapid increase in intraocular pressure (IOP) (Emanuel et al., 2014). One of the experimental models which could mimic this condition is a murine model of retinal ischemia/reperfusion (I/R) injury (Johnson and Tomarev, 2010). Retinal I/R injury can induce a rapid and transient elevation of IOP, which contributes to the degeneration of RGCs. Although understanding the pathophysiology of the degeneration of RGCs was considerably attempted, the major contributing pathways have not been yet elucidated (Calkins and Horner, 2012).
    Related Articles | Metrics
    Bradykinin postconditioning protects rat hippocampal neurons after restoration of spontaneous circulation following cardiac arrest via activation of the AMPK/mTOR signaling pathway
    Shi-Rong Lin, Qing-Ming Lin, Yu-Jia Lin, Xin Qian, Xiao-Ping Wang, Zheng Gong, Feng Chen, Bin Song
    2022, 17 (10):  2232-2237.  doi: 10.4103/1673-5374.337049
    Abstract ( 204 )   PDF (6075KB) ( 83 )   Save
    Bradykinin (BK) is an active component of the kallikrein-kinin system that has been shown to have cardioprotective and neuroprotective effects. We previously showed that BK postconditioning strongly protects rat hippocampal neurons upon restoration of spontaneous circulation (ROSC) after cardiac arrest. However, the precise mechanism underlying this process remains poorly understood. In this study, we treated a rat model of ROSC after cardiac arrest (induced by asphyxiation) with 150 μg/kg BK via intraperitoneal injection 48 hours after ROSC following cardiac arrest. We found that BK postconditioning effectively promoted the recovery of rat neurological function after ROSC following cardiac arrest, increased the amount of autophagosomes in the hippocampal tissue, inhibited neuronal cell apoptosis, up-regulated the expression of autophagy-related proteins LC3 and NBR1 and down-regulated p62, inhibited the expression of the brain injury marker S100β and apoptosis-related protein caspase-3, and affected the expression of adenosine monophosphate-activated protein kinase/mechanistic target of rapamycin pathway-related proteins. Adenosine monophosphate-activated protein kinase inhibitor compound C clearly inhibited BK-mediated activation of autophagy in rats after ROSC following cardiac arrest, which aggravated the injury caused by ROSC. The mechanistic target of rapamycin inhibitor rapamycin enhanced the protective effects of BK by stimulating autophagy. Our findings suggest that BK postconditioning protects against injury caused by ROSC through activating the adenosine monophosphate-activated protein kinase/mechanistic target of the rapamycin pathway.
    Related Articles | Metrics
    The delivery of miR-21a-5p by extracellular vesicles induces microglial polarization via the STAT3 pathway following hypoxia-ischemia in neonatal mice
    Dan-Qing Xin, Yi-Jing Zhao, Ting-Ting Li, Hong-Fei Ke, Cheng-Cheng Gai, Xiao-Fan Guo, Wen-Qiang Chen, De-Xiang Liu, Zhen Wang
    2022, 17 (10):  2238-2246.  doi: 10.4103/1673-5374.336871
    Abstract ( 172 )   PDF (6727KB) ( 153 )   Save
    Extracellular vesicles (EVs) from mesenchymal stromal cells (MSCs) have previously been shown to protect against brain injury caused by hypoxia-ischemia (HI). The neuroprotective effects have been found to relate to the anti-inflammatory effects of EVs. However, the underlying mechanisms have not previously been determined. In this study, we induced oxygen-glucose deprivation in BV-2 cells (a microglia cell line), which mimics HI in vitro, and found that treatment with MSCs-EVs increased the cell viability. The treatment was also found to reduce the expression of pro-inflammatory cytokines, induce the polarization of microglia towards the M2 phenotype, and suppress the phosphorylation of selective signal transducer and activator of transcription 3 (STAT3) in the microglia. These results were also obtained in vivo using neonatal mice with induced HI. We investigated the potential role of miR-21a-5p in mediating these effects, as it is the most highly expressed miRNA in MSCs-EVs and interacts with the STAT3 pathway. We found that treatment with MSCs-EVs increased the levels of miR-21a-5p in BV-2 cells, which had been lowered following oxygen-glucose deprivation. When the level of miR-21a-5p in the MSCs-EVs was reduced, the effects on microglial polarization and STAT3 phosphorylation were reduced, for both the in vitro and in vivo HI models. These results indicate that MSCs-EVs attenuate HI brain injury in neonatal mice by shuttling miR-21a-5p, which induces microglial M2 polarization by targeting STAT3.
    Related Articles | Metrics
    Temporal alterations in pericytes at the acute phase of ischemia/reperfusion in the mouse brain
    Shuang Zhang, Xue-Jing Liao, Jia Wang, Yi Shen, Han-Fen Shi, Yan Zou, Chong-Yang Ma, Xue-Qian Wang, Qing-Guo Wang, Xu Wang, Ming-Yang Xu, Fa-Feng Cheng, Wan-Zhu Bai
    2022, 17 (10):  2247-2252.  doi: 10.4103/1673-5374.336876
    Abstract ( 209 )   PDF (3791KB) ( 212 )   Save
    Pericytes, as the mural cells surrounding the microvasculature, play a critical role in the regulation of microcirculation; however, how these cells respond to ischemic stroke remains unclear. To determine the temporal alterations in pericytes after ischemia/reperfusion, we used the 1-hour middle cerebral artery occlusion model, which was examined at 2, 12, and 24 hours after reperfusion. Our results showed that in the reperfused regions, the cerebral blood flow decreased and the infarct volume increased with time. Furthermore, the pericytes in the infarct regions contracted and acted on the vascular endothelial cells within 24 hours after reperfusion. These effects may result in incomplete microcirculation reperfusion and a gradual worsening trend with time in the acute phase. These findings provide strong evidence for explaining the “no-reflow” phenomenon that occurs after recanalization in clinical practice.
    Related Articles | Metrics
    Cranial irradiation impairs intrinsic excitability and synaptic plasticity of hippocampal CA1 pyramidal neurons with implications for cognitive function
    Min-Yi Wu, Wen-Jun Zou, Pei Yu, Yuhua Yang, Shao-Jian Li, Qiang Liu, Jiatian Xie, Si-Qi Chen, Wei-Jye Lin, Yamei Tang
    2022, 17 (10):  2253-2259.  doi: 10.4103/1673-5374.336875
    Abstract ( 229 )   PDF (2247KB) ( 110 )   Save
    Radiation therapy is a standard treatment for head and neck tumors. However, patients often exhibit cognitive impairments following radiation therapy. Previous studies have revealed that hippocampal dysfunction, specifically abnormal hippocampal neurogenesis or neuroinflammation, plays a key role in radiation-induced cognitive impairment. However, the long-term effects of radiation with respect to the electrophysiological adaptation of hippocampal neurons remain poorly characterized. We found that mice exhibited cognitive impairment 3 months after undergoing 10 minutes of cranial irradiation at a dose rate of 3 Gy/min. Furthermore, we observed a remarkable reduction in spike firing and excitatory synaptic input, as well as greatly enhanced inhibitory inputs, in hippocampal CA1 pyramidal neurons. Corresponding to the electrophysiological adaptation, we found reduced expression of synaptic plasticity marker VGLUT1 and increased expression of VGAT. Furthermore, in irradiated mice, long-term potentiation in the hippocampus was weakened and GluR1 expression was inhibited. These findings suggest that radiation can impair intrinsic excitability and synaptic plasticity in hippocampal CA1 pyramidal neurons.
    Related Articles | Metrics
    A neurovascular unit-on-a-chip: culture and differentiation of human neural stem cells in a three-dimensional microfluidic environment
    Wen-Juan Wei, Ya-Chen Wang, Xin Guan, Wei-Gong Chen, Jing Liu
    2022, 17 (10):  2260-2266.  doi: 10.4103/1673-5374.337050
    Abstract ( 265 )   PDF (5743KB) ( 291 )   Save
    Biological studies typically rely on a simple monolayer cell culture, which does not reflect the complex functional characteristics of human tissues and organs, or their real response to external stimuli. Microfluidic technology has advantages of high-throughput screening, accurate control of the fluid velocity, low cell consumption, long-term culture, and high integration. By combining the multipotential differentiation of neural stem cells with high throughput and the integrated characteristics of microfluidic technology, an in vitro model of a functionalized neurovascular unit was established using human neural stem cell-derived neurons, astrocytes, oligodendrocytes, and a functional microvascular barrier. The model comprises a multi-layer vertical neural module and vascular module, both of which were connected with a syringe pump. This provides controllable conditions for cell inoculation and nutrient supply, and simultaneously simulates the process of ischemic/hypoxic injury and the process of inflammatory factors in the circulatory system passing through the blood-brain barrier and then acting on the nerve tissue in the brain. The in vitro functionalized neurovascular unit model will be conducive to central nervous system disease research, drug screening, and new drug development.
    Related Articles | Metrics
    A comparative analysis of differentially expressed genes in rostral and caudal regions after spinal cord injury in rats
    Xue-Min Cao, Sheng-Long Li, Yu-Qi Cao, Ye-Hua Lv, Ya-Xian Wang, Bin Yu, Chun Yao
    2022, 17 (10):  2267-2271.  doi: 10.4103/1673-5374.336874
    Abstract ( 167 )   PDF (3692KB) ( 99 )   Save
    The initial mechanical damage of a spinal cord injury (SCI) triggers a progressive secondary injury cascade, which is a complicated process integrating multiple systems and cells. It is crucial to explore the molecular and biological process alterations that occur after SCI for therapy development. The differences between the rostral and caudal regions around an SCI lesion have received little attention. Here, we analyzed the differentially expressed genes between rostral and caudal sites after injury to determine the biological processes in these two segments after SCI. We identified a set of differentially expressed genes, including Col3a1, Col1a1, Dcn, Fn1, Kcnk3, and Nrg1, between rostral and caudal regions at different time points following SCI. Functional enrichment analysis indicated that these genes were involved in response to mechanical stimulus, blood vessel development, and brain development. We then chose Col3a1, Col1a1, Dcn, Fn1, Kcnk3, and Nrg1 for quantitative real-time PCR and Fn1 for immunostaining validation. Our results indicate alterations in different biological events enriched in the rostral and caudal lesion areas, providing new insights into the pathology of SCI.
    Related Articles | Metrics
    The functional properties of synapses made by regenerated axons across spinal cord lesion sites in lamprey
    David Parker
    2022, 17 (10):  2272-2277.  doi: 10.4103/1673-5374.335828
    Abstract ( 340 )   PDF (3344KB) ( 113 )   Save
    While the anatomical properties of regenerated axons across spinal cord lesion sites have been studied extensively, little is known of how the functional properties of regenerated synapses compared to those in unlesioned animals. This study aims to compare the properties of synapses made by regenerated axons with unlesioned axons using the lamprey, a model system for spinal injury research, in which functional locomotor recovery after spinal cord lesions is associated with axonal regeneration across the lesion site. Regenerated synapses below the lesion site did not differ from synapses from unlesioned axons with respect to the amplitude and duration of single excitatory postsynaptic potentials. They also showed the same activity-dependent depression over spike trains. However, regenerated synapses did differ from unlesioned synapses as the estimated number of synaptic vesicles was greater and there was evidence for increased postsynaptic quantal amplitude. For axons above the lesion site, the amplitude and duration of single synaptic inputs also did not differ significantly from unlesioned animals. However, in this case, there was evidence of a reduction in release probability and inputs facilitated rather than depressed over spike trains. Synaptic inputs from single regenerated axons below the lesion site thus do not increase in amplitude to compensate for the reduced number of descending axons after functional recovery. However, the postsynaptic input was maintained at the unlesioned level using different synaptic properties. Conversely, the facilitation from the same initial amplitude above the lesion site made the synaptic input over spike trains functionally stronger. This may help to increase propriospinal activity across the lesion site to compensate for the lesion-induced reduction in supraspinal inputs. The animal experiments were approved by the Animal Ethics Committee of Cambridge University.
    Related Articles | Metrics
    Anodal transcranial direct current stimulation alleviates cognitive impairment in an APP/PS1 model of Alzheimer’s disease in the preclinical stage
    Yin-Pei Luo, Zhi Liu, Cong Wang, Xiu-Fang Yang, Xiao-Ying Wu, Xue-Long Tian, Hui-Zhong Wen
    2022, 17 (10):  2278-2285.  doi: 10.4103/1673-5374.337053
    Abstract ( 225 )   PDF (14127KB) ( 111 )   Save
    Anodal transcranial direct current stimulation (AtDCS) has been shown to alleviate cognitive impairment in an APP/PS1 model of Alzheimer’s disease in the preclinical stage. However, this enhancement was only observed immediately after AtDCS, and the long-term effect of AtDCS remains unknown. In this study, we treated 26-week-old mouse models of Alzheimer’s disease in the preclinical stage with 10 AtDCS sessions or sham stimulation. The Morris water maze, novel object recognition task, and novel object location test were implemented to evaluate spatial learning memory and recognition memory of mice. Western blotting was used to detect the relevant protein content. Morphological changes were observed using immunohistochemistry and immunofluorescence staining. Six weeks after treatment, the mice subjected to AtDCS sessions had a shorter escape latency, a shorter path length, more platform area crossings, and spent more time in the target quadrant than sham-stimulated mice. The mice subjected to AtDCS sessions also performed better in the novel object recognition and novel object location tests than sham-stimulated mice. Furthermore, AtDCS reduced the levels of amyloid-β42 and glial fibrillary acidic protein, a marker of astrocyte activation, and increased the level of neuronal marker NeuN in hippocampal tissue. These findings suggest that AtDCS can improve the spatial learning and memory abilities and pathological state of an APP/PS1 mouse model of Alzheimer’s disease in the preclinical stage, with improvements that last for at least 6 weeks.
    Related Articles | Metrics
    Toxicities of amyloid-beta and tau protein are reciprocally enhanced in the Drosophila model
    Zhen-Dong Sun, Jia-Xin Hu, Jia-Rui Wu, Bing Zhou, Yun-Peng Huang
    2022, 17 (10):  2286-2292.  doi: 10.4103/1673-5374.336872
    Abstract ( 159 )   PDF (3707KB) ( 117 )   Save
    Extracellular aggregation of amyloid-beta (Aβ) and intracellular tau tangles are two major pathogenic hallmarks and critical factors of Alzheimer’s disease. A linear interaction between Aβ and tau protein has been characterized in several models. Aβ induces tau hyperphosphorylation through a complex mechanism; however, the master regulators involved in this linear process are still unclear. In our study with Drosophila melanogaster, we found that Aβ regulated tau hyperphosphorylation and toxicity by activating c-Jun N-terminal kinase. Importantly, Aβ toxicity was dependent on tau hyperphosphorylation, and flies with hypophosphorylated tau were insulated against Aβ-induced toxicity. Strikingly, tau accumulation reciprocally interfered with Aβ degradation and correlated with the reduction in mRNA expression of genes encoding Aβ-degrading enzymes, including dNep1, dNep3, dMmp2, dNep4, and dIDE. Our results indicate that Aβ and tau protein work synergistically to further accelerate Alzheimer’s disease progression and may be considered as a combined target for future development of Alzheimer’s disease therapeutics.
    Related Articles | Metrics
    SARM1 participates in axonal degeneration and mitochondrial dysfunction in prion disease
    Meng-Yu Lai, Jie Li, Xi-Xi Zhang, Wei Wu, Zhi-Ping Li, Zhi-Xin Sun, Meng-Yang Zhao, Dong-Ming Yang, Dong-Dong Wang, Wen Li, De-Ming Zhao, Xiang-Mei Zhou, Li-Feng Yang
    2022, 17 (10):  2293-2299.  doi: 10.4103/1673-5374.337051
    Abstract ( 205 )   PDF (6001KB) ( 161 )   Save
    Prion disease represents a group of fatal neurogenerative diseases in humans and animals that are associated with energy loss, axonal degeneration, and mitochondrial dysfunction. Axonal degeneration is an early hallmark of neurodegeneration and is triggered by SARM1. We found that depletion or dysfunctional mutation of SARM1 protected against NAD+ loss, axonal degeneration, and mitochondrial functional disorder induced by the neurotoxic peptide PrP106–126. NAD+ supplementation rescued prion-triggered axonal degeneration and mitochondrial dysfunction and SARM1 overexpression suppressed this protective effect. NAD+ supplementation in PrP106–126-incubated N2a cells, SARM1 depletion, and SARM1 dysfunctional mutation each blocked neuronal apoptosis and increased cell survival. Our results indicate that the axonal degeneration and mitochondrial dysfunction triggered by PrP106–126 are partially dependent on SARM1 NADase activity. This pathway has potential as a therapeutic target in the early stages of prion disease.
    Related Articles | Metrics
    Expression and regulatory network of long noncoding RNA in rats after spinal cord hemisection injury
    Wei Liu, Jin-Cheng Tao, Sheng-Ze Zhu, Chao-Lun Dai, Ya-Xian Wang, Bin Yu, Chun Yao, Yu-Yu Sun
    2022, 17 (10):  2300-2304.  doi: 10.4103/1673-5374.337052
    Abstract ( 133 )   PDF (8475KB) ( 57 )   Save
    Long noncoding RNAs (lncRNAs) participate in a variety of biological processes and diseases. However, the expression and function of lncRNAs after spinal cord injury has not been extensively analyzed. In this study of right side hemisection of the spinal cord at T10, we detected the expression of lncRNAs in the proximal tissue of T10 lamina at different time points and found 445 lncRNAs and 6522 mRNA were differentially expressed. We divided the differentially expressed lncRNAs into 26 expression trends and analyzed Profile 25 and Profile 2, the two expression trends with the most significant difference. Our results showed that the expression of 68 lncRNAs in Profile 25 rose first and remained high 3 days post-injury. There were 387 mRNAs co-expressed with the 68 lncRNAs in Profile 25. The co-expression network showed that the co-expressed genes were mainly enriched in cell division, inflammatory response, FcγR-mediated cell phagocytosis signaling pathway, cell cycle and apoptosis. The expression of 56 lncRNAs in Profile2 first declined and remained low after 3 days post-injury. There were 387 mRNAs co-expressed with the 56 lncRNAs in Profile 2. The co-expression network showed that the co-expressed genes were mainly enriched in the chemical synaptic transmission process and in the signaling pathway of neuroactive ligand-receptor interaction. The results provided the expression and regulatory network of the main lncRNAs after spinal cord injury and clarified their co-expressed gene enriched biological processes and signaling pathways. These findings provide a new direction for the clinical treatment of spinal cord injury.
    Related Articles | Metrics
    DNA vaccines targeting amyloid-β oligomer ameliorate cognitive deficits of aged APP/PS1/tau triple-transgenic mouse models of Alzheimer’s disease
    Sha Sha, Xiao-Na Xing, Tao Wang, Ying Li, Rong-Wei Zhang, Xue-Li Shen, Yun-Peng Cao, Le Qu
    2022, 17 (10):  2305-2310.  doi: 10.4103/1673-5374.337054
    Abstract ( 174 )   PDF (3204KB) ( 112 )   Save
    The amyloid-β (Aβ) oligomer, rather than the Aβ monomer, is considered to be the primary initiator of Alzheimer’s disease. It was hypothesized that p(Aβ3–10)10-MT, the recombinant Aβ3–10 gene vaccine of the Aβ oligomer has the potential to treat Alzheimer’s disease. In this study, we intramuscularly injected the p(Aβ3–10)10-MT vaccine into the left hindlimb of APP/PS1/tau triple-transgenic mice, which are a model for Alzheimer’s disease. Our results showed that the p(Aβ3–10)10-MT vaccine effectively reduced Aβ oligomer levels and plaque deposition in the cerebral cortex and hippocampus, decreased the levels tau protein variants, reduced synaptic loss, protected synaptic function, reduced neuron loss, and ameliorated memory impairment without causing any cerebral hemorrhaging. Therefore, this novel DNA vaccine, which is safe and highly effective in mouse models of Alzheimer’s disease, holds a lot of promise for the treatment of Alzheimer’s disease in humans.
    Related Articles | Metrics
    Magnetic labeling of primary murine monocytes using very small superparamagnetic iron oxide nanoparticles
    Martin Pohland, Christoph Pohland, Jürgen Kiwit, Jana Glumm
    2022, 17 (10):  2311-2315.  doi: 10.4103/1673-5374.336873
    Abstract ( 156 )   PDF (2921KB) ( 105 )   Save
    Due to their very small size, nanoparticles can interact with all cells in the central nervous system. One of the most promising nanoparticle subgroups are very small superparamagnetic iron oxide nanoparticles (VSOP) that are citrate coated for electrostatic stabilization. To determine their influence on murine blood-derived monocytes, which easily enter the injured central nervous system, we applied VSOP and carboxydextran-coated superparamagnetic iron oxide nanoparticles (Resovist). We assessed their impact on the viability, cytokine, and chemokine secretion, as well as iron uptake of murine blood-derived monocytes. We found that (1) the monocytes accumulated VSOP and Resovist, (2) this uptake seemed to be nanoparticle- and time-dependent, (3) the decrease of monocytes viability was treatment-related, (4) VSOP and Resovist incubation did not alter cytokine homeostasis, and (5) overall a 6-hour treatment with 0.75 mM VSOP-R1 was probably sufficient to effectively label monocytes for future experiments. Since homeostasis is not altered, it is safe to label blood-derived monocles with VSOP. VSOP labeled monocytes can be used to study injured central nervous system sites further, for example with drug-carrying VSOP.
    Related Articles | Metrics
    Alexa Fluor 488-conjugated cholera toxin subunit B optimally labels neurons 3–7 days after injection into the rat gastrocnemius muscle
    Jing-Jing Cui, Jia Wang, Dong-Sheng Xu, Shuang Wu, Ya-Ting Guo, Yu-Xin Su, Yi-Han Liu, Yu-Qing Wang, Xiang-Hong Jing, Wan-Zhu Bai
    2022, 17 (10):  2316-2320.  doi: 10.4103/1673-5374.337055
    Abstract ( 343 )   PDF (10396KB) ( 185 )   Save
    Neural tract tracing is used to study neural pathways and evaluate neuronal regeneration following nerve injuries. However, it is not always clear which tracer should be used to yield optimal results. In this study, we examined the use of Alexa Fluor 488-conjugated cholera toxin subunit B (AF488-CTB). This was injected into the gastrocnemius muscle of rats, and it was found that motor, sensory, and sympathetic neurons were labeled in the spinal ventral horn, dorsal root ganglia, and sympathetic chain, respectively. Similar results were obtained when we injected AF594-CTB into the tibialis anterior muscle. The morphology and number of neurons were evaluated at different time points following the AF488-CTB injection. It was found that labeled motor and sensory neurons could be observed 12 hours post-injection. The intensity was found to increase over time, and the morphology appeared clear and complete 3–7 days post-injection, with clearly distinguishable motor neuron axons and dendrites. However, 14 days after the injection, the quality of the images decreased and the neurons appeared blurred and incomplete. Nissl and immunohistochemical staining showed that the AF488-CTB-labeled neurons retained normal neurochemical and morphological features, and the surrounding microglia were also found to be unaltered. Overall, these results imply that the cholera toxin subunit B, whether unconjugated or conjugated with Alexa Fluor, is effective for retrograde tracing in muscular tissues and that it would also be suitable for evaluating the regeneration or degeneration of injured nerves.
    Related Articles | Metrics