Rodger J. Elble

Department of Neurology, Southern Illinois University School of Medicine, Springfield, USA

The Essential Tremor Rating Assessment Scale was developed by the Tremor Research Group (www.tremorresearchgroup.org) to quantify essential tremor severity and its impact on activities of daily living. This scale requires only a pen and paper, and can be completed in about 10 minutes. Upper extremity action tremor is the main focus of this scale, but action tremor is also assessed in the head, face, voice, and lower limbs. The scale has excellent face validity, inter- and intra-rater reliability, and sensitivity to change. The activities of daily living section correlates strongly with the performance section, and this scale also correlates strongly with transducer measures of tremor and with the Fahn-Tolosa-Marín tremor rating scale. In the Fahn-Tolosa-Marín tremor rating scale, upper extremity tremor greater than 4 cm corresponds to a maximum rating of 4, while grade 4 tremor in the Essential Tremor Rating Assessment Scale corresponds to an amplitude greater than 20 cm. Therefore, the Essential Tremor Rating Assessment Scale is better suited for assessment of severe essential tremor.

DOI: 10.29245/2572.942X/2016/4.1038 View / Download Pdf

So Mi Lee1,2, In-One Kim1

1Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
2Department of Radiology, Kyungpook National University Medical Center, Daegu, Korea

Germ cell tumor (GCT) arising in the basal ganglia or thalami is relatively uncommon. It occurs most commonly in boys during second decade of life. It is difficult to diagnose early stage GCTs in these regions because the images are not so typical and the symptom onset is insidious. However, early diagnosis of this tumor is important because of the high radiosensitivity and potential curability. Early stage GCTs originating from the basal ganglia or thalami appear as ill-defined small patchy lesions. They frequently present as hyperintense lesions compared with deep gray matter on T2-weighted image without cyst, mass effect, or prominent enhancement. Microhemorrhages can be accompanied infrequently. These tumors are mostly associated with ipsilateral hemiatrophy at the time of presentation. During tumor progression, tiny cysts develop at a relatively early stage, and intratumoral cyst, hemorrhage, and ipsilateral hemiatrophy gradually tend to be more pronounced. Ultimately, these become overt large mass with remarkable heterogenous enhancement, containing multiple cysts of various sizes and hemorrhage. This review aims to describe the serial MR imaging findings of the GCTs arising from basal ganglia or thalami, focusing on the early finding.

DOI: 10.29245/2572.942X/2016/4.1039 View / Download Pdf

Homajoun Maslehaty1*, Arya Nabavi2, Hubertus Maximilian Mehdorn2

1Department of Neurosurgery, University Hospitals Essen, Germany
2Department of Neurosurgery, University Hospitals Schleswig-Holstein, Kiel, Germany

DOI: 10.29245/2572.942X/2016/4.1009 View / Download Pdf

Qing Lu, Stephen M. Black

Department of Medicine, University of Arizona, AZ, USA

The neuronal cell death associated with perinatal asphyxia, or hypoxic-ischemic (HI) brain injury, plays an important role in neonatal mortality and neurodevelopment retardation. The types of cell death associated with HI in the brain have been classified as being either apoptotic or necrotic. Here we describe the recent discoveries of multiple non-apoptotic cell death pathways: necroptosis; ferroptosis; and autosis (autophagy). These new cell death pathways expand our understanding of the mechanisms underlying the cell death associated with perinatal asphyxia. By targeting specific regulators of these pathways, new therapies may be developed that could protect the neonatal brain from the HI mediated injury.

DOI: 10.29245/2572.942X/2016/4.1035 View / Download Pdf

Gonzalo Flores1 and Julio Morales-Medina2

1Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, Puebla, México
2Centro de Investigación en Reproducción Animal, CINVESTAV- Universidad Autónoma de Tlaxcala, AP 62, CP 90000, Tlaxcala, México

Schizophrenia is a complex mental disorder that starts at early adulthood with a combination of positive and negative symptoms as well as cognitive impairments. It is well known that dendritic spine density and dendritic length of the pyramidal neurons of the prefrontal cortex (PFC) are reduced in the post-mortem tissue of schizophrenia pateints. In addition, the volume of the PFC is reduced in this mental disorder. A possible hypothesis for these morphological changes suggests that the disruption between PFC and hippocampus, at an early age is involved in the pathophysiology of schizophrenia. Furthermore, rats with bilateral lesion of the neonatal ventral hippocampus (nVHL) at an early age is an example of the initial disruption between hippocampus and PFC and also exhibits a reduction in the synaptic connections in the PFC. The present mini-review discusses the neurochemical and morphological changes in the PFC of rats that underwent nVHL, an animal model of schizophrenia.

DOI: 10.29245/2572.942X/2016/3.1024 View / Download Pdf

Patrick L. McGeer* Edith G. McGeer and Moonhee Lee

Kinsmen Laboratory of Neurological Research, University of British Columbia, VancouverBC V6T 1Z3, Canada

Sodium thiosulfate (STS) is an industrial chemical which also has a long medical history. It was originally used as an intravenous medication for metal poisoning. It has since been approved for the treatment of certain rare medical conditions. These include cyanide poisoning, calciphylaxis, and cisplatin toxicity. In vitro assays have demonstrated that it is an anti-inflammatory and neuroprotective agent. It therefore has potential for treating neurodegenerative diseases such as Alzheimer disease and Parkinson disease. NaSH has similar properties and is somewhat more powerful than STS in these in vitro assays. However STS has already been approved as an orally available treatment. STS may therefore be a readily available candidate for treating neurodegenerative disorders such as Alzheimer disease and Parkinson disease.

DOI: 10.29245/2572.942X/2016/3.1032 View / Download Pdf

José Castillo1*, María Isabel Loza2,3, David Mirelman4, Tomás Sobrino1, Francisco Campos1*

1Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela, Spain
2Department of Pharmacology, Universidade de Santiago de Compostela, Discovery group BioFarma, Health Research Institute of Santiago de Compostela, Spain
3BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
4Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel

Glutamate-excitotoxicity is a primary contributor of ischemic neuronal death. Several strategies have been developed against glutamate-excitotoxicity, however any of them have not showed positive results in the clinical practice so far.

Nowadays, the concept of blood/brain glutamate grabbing is well recognized as a novel and attractive protective strategy to reduce the excitotoxic effect of excess extracellular glutamate that accumulates in the brain following an ischemic stroke. The main advantage of this novel therapeutic strategy is that occurs in the blood circulation and therefore does not affect the normal brain neurophysiology, as it has been described for other drug treatments used against glutamate excitotoxicity. In this work, we summarize all experimental data about the potential application of this therapy against stroke pathology.

Ischemic stroke, caused by interruption of the blood supply to the brain, is one of the most important causes of morbidity and mortality worldwide. Currently, the control of systemic parameters, such as body temperature, blood pressure, and glycemia, has considerably improved the outcome of stroke patients. In the absence of protective therapy, an early artery reperfusion, i.e. mechanical or enzymatic thrombolysis, remains the primary goal of treatment for acute ischemic stroke. However, because of the progressive increase in stroke incidence, the high morbidity and the limited therapeutic tools against stroke, seeking new alternatives that can be applied more universally and with less technological requirements is in high demand1, 2.

DOI: 10.29245/2572.942X/2016/3.1030 View / Download Pdf

Silvia Campioni1,2, Roland Riek1*

1Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich, Vladimir-Prelog-Veg 2, 8093 Zurich, Switzerland
2Laboratory of Food and Soft Materials, Department of Health Sciences and Technologies, Swiss Federal Institute of Technology Zurich, Schmelzbergstrasse 9, 8092 Zurich, Switzerland

Despite extensive research, a detailed description of the physiological function of α-Synuclein (α-Syn), the human neuronal protein involved in the pathogenesis of Parkinson’s Disease, is still lacking, most likely due to its highly dynamic conformation and behaviour. Recently, it has become increasingly evident that the interaction of α-Syn with membranes plays an important role in its function and misfunction. Strikingly, despite not having a membrane scaffolding domain, α-Syn can extensively reshape membrane bilayers. Moreover, stable and soluble nanometer-sized particles, whose morphology is ranging from tubules to discoids, can be obtained in vitro with different protocols and from different lipids. The focus of this review article is on the description of the membrane remodelling activity of α-Syn and on its possible physiological role.

DOI: 10.29245/2572.942X/2016/3.1031 View / Download Pdf

Jyotshna Kanungo*

Division of Neurotoxicology, National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA

Alzheimer’s disease (AD) is characterized by neuronal death with an accumulation of intra-cellular neurofibrillary tangles (NFT) and extracellular amyloid plaques. Reduced DNA repair ability has been reported in AD brains. In neurons, the predominant mechanism to repair double-strand DNA breaks (DSB) is non-homologous end joining (NHEJ) that requires DNA-dependent protein kinase (DNA-PK) activity. DNA-PK is a holoenzyme comprising the p460 kD DNA-PK catalytic subunit (DNA-PKcs) and its activator Ku, a heterodimer of p86 (Ku80) and p70 (Ku70) subunits. Upon binding to double-stranded DNA ends, Ku recruits DNA-PKcs to process NHEJ. In AD brains, reduced NHEJ activity as well as DNA-PKcs and Ku protein levels have been shown. Normal aging brains also show a reduction in both DNA-PKcs and Ku levels questioning a direct link between NHEJ ability and AD, and suggesting additional players/events in AD pathogenesis. Deficiency of Ku80, a somatostatin receptor, can disrupt somatostatin signaling thus inducing amyloid beta (Aβ) generation, which in turn can potentiate DNA-PKcs degradation and consequently loss of NHEJ activity, an additional step negatively affecting DSB repair. Trigger of these two different pathways culminating in genome instability may differentiate the outcomes between AD and normal aging.

DOI: 10.29245/2572.942X/2016/3.1016 View / Download Pdf

Ana Velasco* and Maruan Hijazi

Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain

Down syndrome (DS): or trisomy 21: is the most common autosomal aneuploidy and the leading genetic cause of intellectual disability. It is widely established that mental retardation is primarily a consequence of brain functioning and developmental abnormalities in neurogenesis. Some changes in the physical structure of the dendrites are a major cause of impaired synaptic plasticity of DS. The overexpression of the dual specificyty tyrsone phosphorylation-regulated kinase 1A (DYRK1A): located on chromosome 21: is involved in cellular plasticity and responsible for central nervous system disturbance in DS.

Oleic acid is a neurotrophic factor that promotes neuronal differentiation and increases the levels of choline acetyltransferase (ChAT). Furthermore: it has recently been shown that it induces migration and formation of new synapses in euploid cells. However: remarkably oleic acid fails to reproduce the same effects in trisomic cells.

Here we review the hypothesis that oleic acid-dependent synaptic plasticity may be dependent on the lipid environment. Thus: differences in membrane composition may be essential to understand why oleic acid promotes higher cell plasticity in euploid than in trisomic cells.

DOI: 10.29245/2572.942X/2016/3.1029 View / Download Pdf

Katz D1,2, Katz I1,2, Shoenfeld Y1,3*

1The Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer, Israel
2Faculty of Medicine, The Hebrew University of Jerusalem, Israel
3Incumbent of the Laura Schwarz-kipp chair for research of autoimmune diseases, Sackler Faculty of Medicine, Tel-Aviv University, Israel

DOI: 10.29245/2572.942X/2016/4.1017 View / Download Pdf

Alex Steimle and Julia-Stefanie Frick*

Institute of Medical Microbiology and Hygiene, University of Tuebingen, Tübingen, Germany

In recent years, a growing interest in pathophysiological processes that are associated with the endosomal and lysosomal protease cathepsin S (CTSS) results in an increasing number of various published methods for CTSS activity detection. CTSS has been reported to be involved in the pathology of autoimmune diseases like multiple sclerosis as well as in tumor growth and Alzheimer’s disease. These implications make this enzyme a first class drug target. In order to fully understand the involvement of CTSS in the formation of pathological processes, gene and protein expression analysis is not sufficient. Rather, one should focus on the regulation of its enzymatic activity. Different approaches for CTSS activity detection are available and described. However, some of these approaches are not suitable for a standard laboratory without special equipment or technical expertise or provide other limitations. We have recently published an easy-to-perform protocol for reliable, quantifiable and reproducible CTSS activity detection. In this review we want to discuss our application and compare it with other published methods and protocols. This might help researchers who are interested in CTSS research to decide which application fits best to their technical or personal facilities.

DOI: 10.29245/2572.942X/2016/3.1023 View / Download Pdf

Jianli Niu and Pappachan E. Kolattukudy*

Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA

Inflammatory response represents one of the first immune processes following injury. However, evidence indicates that inflammatory response can also induce cellular protection associated with preconditioning, a phenomenon in which brief episodes of a sublethal insult induce robust protection against subsequent lethal injuries. The elucidation of mechanisms that allow inflammatory response to confer cellular protection is critical to developing new therapeutic strategies against acute ischemic insults. In the present review, we will give a short overview on a novel zinc-finger protein, MCPIP (also known as Zc3h12a or Regnase-1), which may function as a master integrator of endogenous cellular protection exerted by preconditioning.

DOI: 10.29245/2572.942X/2016/3.1026 View / Download Pdf

Lorna Guéniot1a, Claire Latroche1ab, Cédric Thépenier1,2a, Laurent Chatre3,4, Aurélien Mazeraud1, Daniel Fiole1,2, Pierre L. Goossens1, Fabrice Chrétien1,5,6c and Gregory Jouvion1,5c

1Institut Pasteur, Histopathologie Humaine et Modèles Animaux, Paris, France
2Institut de Recherche Biomédicale des Armées, Interactions Hôte-Agents Pathogènes, Brétigny-sur-Orge, France
3Groupe Stabilité de l’ADN Nucléaire et Mitochondrial, CNRS UMR 3525, Paris, France
4Institut Pasteur, Cellules Souches et Développement, Paris, France
5Université Paris Descartes, PRES Sorbonne-Paris-Cité, Paris, France
6CH Sainte-Anne, Neuropathology Department, Paris, France
aGuéniot L, Latroche C, and Thépenier C contributed equally to this work
bPresent address: San Raffaele Telethon Institute for Gene Therapy; Division of Regenerative Medicine, Stem Cells and Gene Therapy; San Raffaele Scientific Institute; Milano, Italia.
cChrétien F and Jouvion G share senior co-authorship

Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease, caused by absence of functional dystrophin and inevitably leading to death. A variable proportion of women carriers (2.5 to 19%) can also manifest symptoms ranging from myalgia to cardiomyopathy, and pathophysiological mechanisms are still not completely understood. Our study focused on 12 month-old female mdx mice, displaying marked chronic muscle lesions, similar to the lesions observed in human DMD. Our aim was to focus on the alterations of the vascular network organisation, and functional repercussions using a combination of histology/morphometry techniques and totally non-invasive functional approach (multiparametric and functional nuclear magnetic resonance), clearly relevant for clinical diagnosis and research, combining arterial spin labeling imaging of perfusion, and 31P-spectroscopy of phosphocreatine kinetics. Collectively, our results demonstrate that the vasculature, both in its steady state organisation and dynamic behaviour after an ischemia-reperfusion stress, is altered in the 12 month-old female mdx mouse: increased density of vascular sections in histology, modification of the post-ischemic hyperemia profile, increase in mitochondrial oxidative rephosphorylation capacity, in striking opposition to what was observed in age-matched male mdx mice. We believe the apparent discordance between vascular and muscular features in the female mdx mouse make it an interesting tool to decipher further dystrophinopathy pathophysiological mechanisms.

DOI: 10.29245/2572.942X/2016/2.1014 View / Download Pdf

Domingos J1,3, Godinho C1,2,3, Ferreira JJ1,3,4*

1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
2Center for Interdisciplinary Research Egas Moniz (CiiEM), Instituto Superior de Ciências da Saúde Egas Moniz, Monte de Caparica, Portugal
3CNS-Campus Neurológico Sénior, Portugal
4Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal

Technologies may have implications for improving clinical diagnosis and prognosis, and for the development of therapeutic interventions, specific biomarkers, and preventive strategies. Given the amount of existing and ever-growing quantitative assessments using technology in PD, clinicians, patients and researchers are faced with the challenge of deciding which assessment tool to use in the laboratory, clinic and home environment. In order to facilitate this decision-making a systematic review was done to identify and classify the available monitoring technologies for individuals with PD over the last 2 decades. This is a commentary on the systematic review which adds on discussion on some controversial issues in the area. It tackles some of current open-to-discussion topics in the technology field, such as: which definitions to use, the heterogeneity of the clinimetric properties among technologies, standardization of a validation process, how to group different measuring technologies, and the need to conduct further studies on existing technologies before developing new ones. The strength of this comprehensive, timely and useful review is the detailed and robust approach taken by authors to classify technologies as listed, suggested, or recommended for the assessment of individuals with PD.

DOI: 10.29245/2572.942X/2016/2.1013 View / Download Pdf

J. Leigh Leasure1,2*, Rebecca West1

1Department of Psychology, University of Houston, 126 Heyne Building, Houston, TX 77204-5022, United States
2Department of Biology & Biochemistry, 3455 Cullen Boulevard, Room 342, Houston, TX 77204-5001, United States

Exercise has long been considered a useful means by which to maintain brain health and treat brain diseases. Yet many of the neural benefits of exercise, such as enhanced hippocampal neurogenesis, take weeks to manifest. Moreover, the brains most in need of the restorative effects of exercise are often paired with bodies that can tolerate very little physical activity, such as those deconditioned by stroke. It would therefore be of great utility to pinpoint ways in which the brain benefits of exercise could be augmented without adding additional exercise time. Exercise represents a significant challenge to the brain because of the heat produced by exercising muscles, but physical activity in the cold attenuates this physiological burden. Using a rat model of voluntary exercise, we recently tested the hypothesis that exercise in cold ambient temperature (4.5°C) would stimulate hippocampal neurogenesis more effectively than exercise at room temperature. We found that, compared to animals that ran at room temperature, animals that exercised in the cold ran a shorter distance and for less total time. Nonetheless, they had more significantly more newly generated neurons in the hippocampal dentate gyrus, indicating that running in the cold may be an effective means by which to maximize brain exercise benefits, yet minimize exercise time.

Exercise is increasingly becoming accepted as “medicine” for diseases of both brain and body1. For the brain, exercise offers chemical, cellular and structural benefits, including enhanced generation of new neurons, glia and blood vessels2-5, increased expression of neurotrophins (such as brain-derived neurotrophic factor (BDNF)6,7), dendritic remodeling8,9 and stabilization of stress responses10 and inflammatory signaling11. These mechanisms of action directly counteract those present in disease states. For example, the depressed brain is characterized by decreased synaptic plasticity, hippocampal neurogenesis and BDNF12, all of which can be reversed by exercise.

DOI: 10.29245/2572.942X/2016/2.1027 View / Download Pdf

Riikka-Liisa Uronen, Henri J. Huttunen*

Neuroscience Center, University of Helsinki, FI-00014 Helsinki, Finland

In Alzheimer’s disease (AD), loss of neurons and synapses parallels the formation of neurofibrillary tangles, protein aggregates mainly composed of hyperphosphorylated and aggregated Tau protein. Tau is mostly a cytosolic protein but can also be secreted by neurons. Cell-to-cell transfer of misfolded Tau protein plays a key role in the spread of neurofibrillary pathology between brain regions in AD and other tauopathies. Advances in genome-wide technologies have identified a large number of genetic risk factors for late-onset AD (LOAD). Currently, it remains unknown if genetic factors influence disease risk or progression rate by altering cell-to-cell propagation of Tau. Several LOAD risk genes are functionally associated with endocytic trafficking providing a potential link to Tau secretion and uptake. Recently, a LOAD risk gene FRMD4A was shown to regulate Tau secretion via a pathway linked to presynaptic vesicle machinery and polarity signaling. Tau release is linked to neuronal activity, and genetic factors that affect presynaptic vesicle release in the aging brain may also influence disease progression in AD and other tauopathies. In this mini review, we summarize the recent literature with a focus on the role of FRMD4A-cytohesin-Arf6 pathway and presynaptic vesicle machinery in the secretion of Tau.

DOI: 10.29245/2572.942X/2016/2.1022 View / Download Pdf

Gabriele C Saretzki

Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, UK

Telomerase is best known for its canonical function in telomere maintenance. However, a growing number of non-telomeric functions have been described. Several groups have found the telomerase protein TERT to persist in adult brain neurons. A protective role for the telomerase protein TERT had been demonstrated in cultivated mouse neurons during brain development, against excitotoxic stresses from N-methyl-D-aspartate (NMDA) and glutamate and agents known to be involved in neurodegenerative diseases such as beta amyloid peptides and hyperphosphorylated tau. In contrast, lack of telomerase and TERT protein increase oxidative stress and decrease neuronal survival. Research on telomerase and TERT protein in human neurodegenerative diseases is a relatively new field. However, there is emerging evidence of a beneficial role of telomerase in human brains and animal models of neurodegenerative diseases that suggests to explore the possibility of using telomerase activators as neuroprotective agents to combat brain ageing and to ameliorate neurodegenerative diseases such as Parkinson’s and Alzheimer’s diseases. The current mini-review summarises the knowledge about this developing novel area of brain research.

DOI: 10.29245/2572.942X/2016/2.1025 View / Download Pdf

Alejandra N. Martinez1, Mario T. Philipp1,2

1Division of Bacteriology & Parasitology, Tulane National Primate Research Center, Covington, LA, USA
2Department of Microbiology and Immunology, Tulane University Medical School, New Orleans, LA, USA

This review addresses the role that substance P (SP) and its preferred receptor neurokinin-1 (NK1R) play in neuroinflammation associated with select bacterial, viral, parasitic, and neurodegenerative diseases of the central nervous system. The SP/NK1R complex is a key player in the interaction between the immune and nervous systems. A common effect of this interaction is inflammation. For this reason and because of the predominance in the human brain of the NK1R, its antagonists are attractive potential therapeutic agents. Preventing the deleterious effects of SP through the use of NK1R antagonists has been shown to be a promising therapeutic strategy, as these antagonists are selective, potent, and safe. Here we evaluate their utility in the treatment of different neuroinfectious and neuroinflammatory diseases, as a novel approach to clinical management of CNS inflammation.

DOI: 10.29245/2572.942X/2016/2.1020 View / Download Pdf

Bernd Krone

1Center for Hygiene and Human Genetics, University Göttingen, Göttingen, Germany
2Medical Laboratory, Kurt-Reuber-Haus, Herkulesstraße 34a, 34119 Kassel, Germany

The sporadic forms of motor neuron diseases seem to depend on a multitude of factors that are, however, largely speculative. A main endogenous factor is seen in the enhanced expression of human endogenous retroviruses (HERVs). It is supposed that this is affecting the biogenesis of neuromelanin, in particular in the locus coeruleus. An altered neuromelanin with increased storage of metals and of precursors for toxins from food, respectively, might be of central importance. A hypothesis is described that is able to explain a number of observations and to understand roles and interactions of different factors. Toxins affecting the excitatory system are formed apparently within altered neuromelanin. Moreover, neuromelanin loaded with metal is also a microenvironment where pathological variants of proteins such as of alpha-synuclein are generated. Familial and toxin related forms of the diseases merge in formation and deposition of aberrant proteins and in a failure to destroy superoxide, thus increasing the harm from oxidative and nitrosative stress. This view might give some rational to revive investigations in methods for stabilization of storage and cautious squeezing out of metals and toxicants from neuromelanin, relying eventually on melatonin, iron chelators and chloroquine, respectively.

DOI: 10.29245/2572.942X/2016/2.1015 View / Download Pdf

Nathalie Khoury, Kevin B. Koronowski and Miguel A. Perez-Pinzon*

Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA

In the absence of effective neuroprotective agents in the clinic, ischemic and pharmacological preconditioning are gaining increased interest in the field of cerebral ischemia. Our lab recently reported that resveratrol preconditioning affords tolerance against a focal cerebral ischemic insult in mice that can last for at least 14 days in vivo making it the longest window of ischemic tolerance discovered to date by a single administration of a pharmacological agent. The mechanism behind this novel extended window of ischemic tolerance remains elusive. In the below commentary we discuss potential mechanisms that could explain this novel extended window of ischemic tolerance in the context of previously identified windows and the known mechanisms behind them. We also draw parallels from the fields of hibernation and hypoxia-tolerance, which are chronic adaptations to severe conditions of hypoxia and ischemia known to be mediated by a form of metabolic depression. We also briefly discuss the importance of epigenetic modifications in maintaining this depressed state of metabolism.

DOI: 10.29245/2572.942X/2016/2.1021 View / Download Pdf

Godinho C1,2,3, Domingos J1,3, Domingos J1,3,4*

1Instituto de Medicina Molecular, Faculty of Medicine, University of Lisbon, Portugal
2Center for Interdisciplinary Research Egas Moniz (CiiEM), Instituto Superior de Ciências da Saúde Egas Moniz, Monte de Caparica, Portugale
3CNS-Campus Neurológico Sénior, Torres Vedras, Portugal
4Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal

Healthcare professionals and pharmaceutical companies invest a great amount of time and effort in continuously creating electronic health solutions. These technology system developments may represent a step forward in care as ultimately it is not possible to manage what cannot be evaluated.

Yet, the use of future generations of technology depends on their specific design, fabrication, distribution, and, most importantly, patients adopting these new technologies as life companions. Data management and the use of artificial intelligence appear as new technological challenges. The overload, sharing and handling of information give rise to new legal, social, and ethical discussions in a field where there is a lack of universal criteria for data ownership, privacy and sharing.

Future technological progress requires much cooperation between multidisciplinary teams including sufficient sharing and benchmarking within open access frameworks.

DOI: 10.29245/2572.942X/2016/1.1019 View / Download Pdf

Yoshio Matsuda

Department of Obstetrics and Gynecology, International University of Health and Welfare Hospital, Japan

DOI: 10.29245/2572.942X/2016/1.1011 View / Download Pdf

Jeffrey L. Neul1,2, Alan K. Percy2

1Department of Neuroscience, University of California San Diego, USA
2Civitan International Research Center, University of Alabama at Birmingham, USA

DOI: 10.29245/2572.942X/2016/1.1008 View / Download Pdf

Kurt A. Jellinger

Institute of Clinical Neurobiology, Vienna, Austria

The aging brain is characterized by the simultaneous presence of multiple pathologies, and the prevalence of multi-morbidity increases with age. Large clinico-pathological correlative studies have shown that in brains of both cognitively intact and impaired aged subjects, the presence of a single disease is a rare finding, while most brains show neurodegenerative, cerebrovascular and other pathologies, which frequently have been missed clinically and may even be difficult to identify at postmortem examination. Since both clinical and autopsy studies differ in selection and classification criteria and in the applied evaluation methods, irrespective of the clinical symptoms, the reported frequency of cerebral pathologies varies considerably. The frequent co-occurrence of different pathologies indicates their mutual interaction in order to promote cognitive decline and other clinical symptoms. These facts have also implications for improvement of clinical diagnosis and prognosis, for the development of specific biomarkers, preventive strategies, and therapeutic targets for cerebral multi-morbidity.

DOI: 10.29245/2572.942X/2016/1.1001 View / Download Pdf