99951
|
Gracia P, Camino JD, Volpicelli-Daley L, Cremades N. Multiplicity of α-Synuclein Aggregated Species and Their Possible Roles in Disease. Int J Mol Sci 2020; 21:E8043. [PMID: 33126694 PMCID: PMC7663424 DOI: 10.3390/ijms21218043] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
α-Synuclein amyloid aggregation is a defining molecular feature of Parkinson's disease, Lewy body dementia, and multiple system atrophy, but can also be found in other neurodegenerative disorders such as Alzheimer's disease. The process of α-synuclein aggregation can be initiated through alternative nucleation mechanisms and dominated by different secondary processes giving rise to multiple amyloid polymorphs and intermediate species. Some aggregated species have more inherent abilities to induce cellular stress and toxicity, while others seem to be more potent in propagating neurodegeneration. The preference for particular types of polymorphs depends on the solution conditions and the cellular microenvironment that the protein encounters, which is likely related to the distinct cellular locations of α-synuclein inclusions in different synucleinopathies, and the existence of disease-specific amyloid polymorphs. In this review, we discuss our current understanding on the nature and structure of the various types of α-synuclein aggregated species and their possible roles in pathology. Precisely defining these distinct α-synuclein species will contribute to understanding the molecular origins of these disorders, developing accurate diagnoses, and designing effective therapeutic interventions for these highly debilitating neurodegenerative diseases.
Collapse
Affiliation(s)
- Pablo Gracia
- Joint Unit BIFI-IQFR (CSIC), Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018 Zaragoza, Spain; (P.G.); (J.D.C.)
| | - José D. Camino
- Joint Unit BIFI-IQFR (CSIC), Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018 Zaragoza, Spain; (P.G.); (J.D.C.)
| | - Laura Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Nunilo Cremades
- Joint Unit BIFI-IQFR (CSIC), Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018 Zaragoza, Spain; (P.G.); (J.D.C.)
| |
Collapse
|
99952
|
Unal DB, Caliari SR, Lampe KJ. 3D Hyaluronic Acid Hydrogels for Modeling Oligodendrocyte Progenitor Cell Behavior as a Function of Matrix Stiffness. Biomacromolecules 2020; 21:4962-4971. [PMID: 33112592 DOI: 10.1021/acs.biomac.0c01164] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The lack of regenerative solutions for demyelination within the central nervous system motivates the development of strategies to expand and drive the bioactivity of the cells, including oligodendrocyte progenitor cells (OPCs), that ultimately give rise to myelination. In this work, we introduce a 3D hyaluronic acid (HA) hydrogel system to study the effects of microenvironmental mechanical properties on the behavior of OPCs. We tuned the stiffness of the hydrogels to match the brain tissue (storage modulus 200-2000 Pa) and studied the effects of stiffness on metabolic activity, proliferation, and cell morphology of OPCs over a 7 day period. Although hydrogel mesh size decreased with increasing stiffness, all hydrogel groups facilitated OPC proliferation and mitochondrial metabolic activity to similar degrees. However, OPCs in the two lower stiffness hydrogel groups (170 ± 42 and 794 ± 203 Pa) supported greater adenosine triphosphate levels per cell than the highest stiffness hydrogels (2179 ± 127 Pa). Lower stiffness hydrogels also supported higher levels of cell viability and larger cell spheroid formation compared to the highest stiffness hydrogels. Together, these data suggest that 3D HA hydrogels are a useful platform for studying OPC behavior and that OPC growth/metabolic health may be favored in lower stiffness microenvironments mimicking brain tissue mechanics.
Collapse
Affiliation(s)
- Deniz B Unal
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States.,Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903, United States
| |
Collapse
|
99953
|
Paul BD, Snyder SH, Kashfi K. Effects of hydrogen sulfide on mitochondrial function and cellular bioenergetics. Redox Biol 2020; 38:101772. [PMID: 33137711 PMCID: PMC7606857 DOI: 10.1016/j.redox.2020.101772] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/17/2020] [Accepted: 10/26/2020] [Indexed: 01/06/2023] Open
Abstract
Hydrogen sulfide (H2S) was once considered to have only toxic properties, until it was discovered to be an endogenous signaling molecule. The effects of H2S are dose dependent, with lower concentrations being beneficial and higher concentrations, cytotoxic. This scenario is especially true for the effects of H2S on mitochondrial function, where higher concentrations of the gasotransmitter inhibit the electron transport chain, and lower concentrations stimulate bioenergetics in multiple ways. Here we review the role of H2S in mitochondrial function and its effects on cellular physiology. Hydrogen sulfide (H2S) plays central roles in mitochondrial homeostasis. Both excess H2S and a paucity of H2S have deleterious effects. One of the modes by which H2S signals in mitochondria is by sulfhydrating target proteins. Administering H2S (where scarcity of H2S occurs) or inhibiting H2S production (in case of excess H2S) may be beneficial.
Collapse
Affiliation(s)
- Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, USA.
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, USA; Department of Psychiatry and Behavioral Sciences, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, NY, 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, NY, 10016, USA.
| |
Collapse
|
99954
|
Lovett ML, Nieland TJ, Dingle YTL, Kaplan DL. Innovations in 3-Dimensional Tissue Models of Human Brain Physiology and Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909146. [PMID: 34211358 PMCID: PMC8240470 DOI: 10.1002/adfm.201909146] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Indexed: 05/04/2023]
Abstract
3-dimensional (3D) laboratory tissue cultures have emerged as an alternative to traditional 2-dimensional (2D) culture systems that do not recapitulate native cell behavior. The discrepancy between in vivo and in vitro tissue-cell-molecular responses impedes understanding of human physiology in general and creates roadblocks for the discovery of therapeutic solutions. Two parallel approaches have emerged for the design of 3D culture systems. The first is biomedical engineering methodology, including bioengineered materials, bioprinting, microfluidics and bioreactors, used alone or in combination, to mimic the microenvironments of native tissues. The second approach is organoid technology, in which stem cells are exposed to chemical and/or biological cues to activate differentiation programs that are reminiscent of human (prenatal) development. This review article describes recent technological advances in engineering 3D cultures that more closely resemble the human brain. The contributions of in vitro 3D tissue culture systems to new insights in neurophysiology, neurological diseases and regenerative medicine are highlighted. Perspectives on designing improved tissue models of the human brain are offered, focusing on an integrative approach merging biomedical engineering tools with organoid biology.
Collapse
Affiliation(s)
- Michael L. Lovett
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Thomas J.F. Nieland
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Yu-Ting L. Dingle
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| |
Collapse
|
99955
|
Davis CK, Vemuganti R. DNA damage and repair following traumatic brain injury. Neurobiol Dis 2020; 147:105143. [PMID: 33127471 DOI: 10.1016/j.nbd.2020.105143] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI) is known to promote significant DNA damage irrespective of age, sex, and species. Chemical as well as structural DNA modification start within minutes and persist for days after TBI. Although several DNA repair pathways are induced following TBI, the simultaneous downregulation of some of the genes and proteins of these pathways leads to an aberrant overall DNA repair process. In many instances, DNA damages escape even the most robust repair mechanisms, especially when the repair process becomes overwhelmed or becomes inefficient by severe or repeated injuries. The persisting DNA damage and/or lack of DNA repair contributes to long-term functional deficits. In this review, we discuss the mechanisms of TBI-induced DNA damage and repair. We further discussed the putative experimental therapies that target the members of the DNA repair process for improved outcome following TBI.
Collapse
Affiliation(s)
- Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA.
| |
Collapse
|
99956
|
Turchi R, Faraonio R, Lettieri-Barbato D, Aquilano K. An Overview of the Ferroptosis Hallmarks in Friedreich's Ataxia. Biomolecules 2020; 10:E1489. [PMID: 33126466 PMCID: PMC7693407 DOI: 10.3390/biom10111489] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/19/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Friedreich's ataxia (FRDA) is a neurodegenerative disease characterized by early mortality due to hypertrophic cardiomyopathy. FRDA is caused by reduced levels of frataxin (FXN), a mitochondrial protein involved in the synthesis of iron-sulphur clusters, leading to iron accumulation at the mitochondrial level, uncontrolled production of reactive oxygen species and lipid peroxidation. These features are also common to ferroptosis, an iron-mediated type of cell death triggered by accumulation of lipoperoxides with distinct morphological and molecular characteristics with respect to other known cell deaths. SCOPE OF REVIEW Even though ferroptosis has been associated with various neurodegenerative diseases including FRDA, the mechanisms leading to disease onset/progression have not been demonstrated yet. We describe the molecular alterations occurring in FRDA that overlap with those characterizing ferroptosis. MAJOR CONCLUSIONS The study of ferroptotic pathways is necessary for the understanding of FRDA pathogenesis, and anti-ferroptotic drugs could be envisaged as therapeutic strategies to cure FRDA.
Collapse
Affiliation(s)
- Riccardo Turchi
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy;
| | - Daniele Lettieri-Barbato
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy;
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy;
| |
Collapse
|
99957
|
Bartosz G, Grzesik-Pietrasiewicz M, Sadowska-Bartosz I. Fluorescent Products of Anthocyanidin and Anthocyanin Oxidation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12019-12027. [PMID: 33048534 DOI: 10.1021/acs.jafc.0c04755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We found that the oxidation of cyanidin with hydrogen peroxide, tert-butyl hydroperoxide, 2,2'-azobis(2-methylpropionamidine), 3-morpholinosydnonimine hydrochloride, sodium hypochlorite, Fe3+, and Fe2+ induced the appearance of a new fluorescence band, centered at 525-540 nm when excited at 470-490 nm. The intensity of this fluorescence was related to the oxidant concentration. The same fluorescence was induced by the oxidation of other anthocyanidins, an anthocyanin (cyanidin-3-O-glucoside) as well as anthocyanin-rich red cabbage extract, and blackcurrant and elderberry juices. Peroxidized blood plasma also induced the appearance of cyanidin fluorescence. We also showed that the formation of the fluorescent product of pelargonidin can be observed in a culture of the MRC-5 human fetal lung fibroblast cell line and the DU-145 human prostate carcinoma cell line. Our results suggest that this new anthocyanidin/anthocyanin fluorescence may be an indicator of oxidation, especially of food products, where these compounds are present or added as colorants, and can also be useful to detect oxidation in biomedical experiments.
Collapse
Affiliation(s)
- Grzegorz Bartosz
- Department of Bioenergetics, Food Analysis and Microbiology, Institute of Food Technology and Nutrition, College of Natural Sciences, Rzeszow University, 4 Zelwerowicza Street, 35-601 Rzeszow, Poland
| | - Michalina Grzesik-Pietrasiewicz
- Department of Analytical Biochemistry, Institute of Food Technology and Nutrition, College of Natural Sciences, Rzeszow University, 4 Zelwerowicza Street, 35-601 Rzeszow, Poland
| | - Izabela Sadowska-Bartosz
- Department of Analytical Biochemistry, Institute of Food Technology and Nutrition, College of Natural Sciences, Rzeszow University, 4 Zelwerowicza Street, 35-601 Rzeszow, Poland
| |
Collapse
|
99958
|
Moreira JD, Siqueira LV, Müller AP, Porciúncula LO, Vinadé L, Souza DO. Dietary omega-3 fatty acids prevent neonatal seizure-induced early alterations in the hippocampal glutamatergic system and memory deficits in adulthood. Nutr Neurosci 2020; 25:1066-1077. [PMID: 33107813 DOI: 10.1080/1028415x.2020.1837569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE We investigated the influence of dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) on glutamatergic system modulation after a single episode of neonatal seizures and their possible effects on seizure-induced long-lasting behavioral deficits. METHODS Male Wistar rats receiving an omega-3 diet (n-3) or an n-3 deficient diet (D) from the prenatal period were subjected to a kainate-induced seizure model at P7. Glutamate transporter activity and immunocontents (GLT-1 and GLAST) were assessed in the hippocampus at 12, 24, and 48 h after the seizure episode. Fluorescence intensity for glial cells (GFAP) and neurons (NeuN) was assessed 24 h after seizure in the hippocampus. Behavioral analysis (elevated-plus maze and inhibitory avoidance memory task) was performed at 60 days of age. RESULTS The D group showed a decrease in glutamate uptake 24 h after seizure. In this group only, the GLT1 content increased at 12 h, followed by a decrease at 24 h. GLAST increased up to 24 h after seizure. GFAP fluorescence was higher, and NeuN fluorescence decreased, in the D group independent of seizures. In adulthood, the D group presented memory deficits independent of seizures, but short-term memory (1.5 h after a training session) was abolished in the D group treated with kainate. SIGNIFICANCE N-3 PUFA positively influenced the glutamatergic system during seizure and prevented seizure-related memory deficits in adulthood.
Collapse
Affiliation(s)
- Júlia D Moreira
- Postgraduate Program in Nutrition, Translational Nutrition Neuroscience Working Group, Health Science Centre, Universidade Federal de Santa Catarina, Santa Catarina, Brazil
| | - Letícia Vicari Siqueira
- Postgraduate Program in Biological Science - Biochemistry, Basic Health Science Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alexandre P Müller
- Postgraduate Program in Pharmacology, Health Science Centre, Universidade Federal de Santa Catarina, Brazil
| | - Lisiane O Porciúncula
- Postgraduate Program in Biological Science - Biochemistry, Basic Health Science Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lúcia Vinadé
- Master Graduation Program in Biological Sciences (Programa de Pós-Graduação em Ciências Biológicas), Universidade Federal do Pampa - UNIPAMPA, Campus São Gabriel, São Gabriel, Brazil
| | - Diogo O Souza
- Postgraduate Program in Biological Science - Biochemistry, Basic Health Science Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
99959
|
Heredia RR, Blackburn AM, Vega LA. Moderation-Mediation Effects in Bilingualism and Cognitive Reserve. Front Psychol 2020; 11:572555. [PMID: 33101142 PMCID: PMC7554512 DOI: 10.3389/fpsyg.2020.572555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023] Open
Abstract
We first provide a critical review of the existing findings on bilingualism as a contributor to cognitive reserve from moderator-mediator warranting cause-effect research conclusions. We next address the question of direct or indirect effects between bilingualism and neurocognitive protective factors influencing the associated age-related mental deficits. The existing findings support bilingualism as a predictor and as a moderator. Third, we propose cognitive reserve models of bilingualism describing analytical approaches that allow testing of these models and hypotheses related to path strength and causal relationships between predictors, moderators, and mediators. Lastly and most importantly, we suggest using large datasets available via open repositories. This can aid in the testing of theoretical models, clarifying the roles of moderators and mediators, and assessing the research viability of multi-causal paths that can influence cognitive reserve. Creating collaborative datasets to test these models would greatly advance our field and identify critical variables in the study of the bilingual aging brain.
Collapse
Affiliation(s)
- Roberto R Heredia
- Department of Psychology and Communication, Texas A&M International University, Laredo, TX, United States
| | - Angélique M Blackburn
- Department of Psychology and Communication, Texas A&M International University, Laredo, TX, United States
| | - Luis A Vega
- Department of Psychology, California State University-Bakersfield, Bakersfield, CA, United States
| |
Collapse
|
99960
|
Morris M, Mańkowska A, Heilman KM. Upper Vertical Spatial Neglect With A Right Temporal Lobe Stroke. Cogn Behav Neurol 2020; 33:63-66. [PMID: 32132404 DOI: 10.1097/wnn.0000000000000221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Individuals with a hemispheric infarction often reveal inattention to, or neglect of, contralesional lateral space (ie, hemispatial neglect). Individuals with a bilateral ventral temporal-occipital lesion have been shown to demonstrate upper vertical neglect, and those with a bilateral parietal-occipital lesion have been shown to demonstrate lower vertical neglect. However, to our knowledge, there have been no reports of individuals with vertical neglect from a unilateral hemispheric lesion. We report on a 72-year-old, right-handed male who developed transient left hemiparesis. On examination, he had left facial weakness and he bisected horizontal lines to the left of the midline (ie, ipsilesional neglect). In addition, on a line bisection test involving nine vertical line bisections, he demonstrated downward deviation in the majority of the trials; healthy individuals deviate upward. On brain imaging, our patient revealed a cerebral infarction, primarily affecting the right temporal lobe; the temporal lobes contain the ventral attentional network that allocates attention upward. There is also some evidence that, whereas the right hemisphere mediates attention upward, the left mediates attention downward. Therefore, injury to the right temporal lobe may account for our patient's upward neglect with downward deviation. However, further studies are needed to better understand the pathophysiology of vertical neglect.
Collapse
Affiliation(s)
- Michael Morris
- Division of Cognitive and Behavioral Neurology, Department of Neurology, University of Florida College of Medicine, Gainesville, Florida.,Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| | - Aleksandra Mańkowska
- Division of Clinical Psychology and Neuropsychology, Institute of Psychology, Department of the Social Sciences, University of Gdańsk, Gdańsk, Poland
| | - Kenneth M Heilman
- Division of Cognitive and Behavioral Neurology, Department of Neurology, University of Florida College of Medicine, Gainesville, Florida.,Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida
| |
Collapse
|
99961
|
Levanovich PE, Diaczok A, Rossi NF. Clinical and Molecular Perspectives of Monogenic Hypertension. Curr Hypertens Rev 2020; 16:91-107. [PMID: 30963979 PMCID: PMC7499356 DOI: 10.2174/1573402115666190409115330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022]
Abstract
Advances in molecular research techniques have enabled a new frontier in discerning the mechanisms responsible for monogenic diseases. In this review, we discuss the current research on the molecular pathways governing blood pressure disorders with a Mendelian inheritance pattern, each presenting with a unique pathophysiology. Glucocorticoid Remediable Aldosteronism (GRA) and Apparent Mineralocorticoid Excess (AME) are caused by mutations in regulatory enzymes that induce increased production of mineralocorticoids or inhibit degradation of glucocorticoids, respectively. Geller syndrome is due to a point mutation in the hormone responsive element of the promotor for the mineralocorticoid receptor, rendering the receptor susceptible to activation by progesterone, leading to hypertension during pregnancy. Pseudohypoaldosteronism type II (PHA-II), also known as Gordon's syndrome or familial hyperkalemic hypertension, is a more variable disorder typically characterized by hypertension, high plasma potassium and metabolic acidosis. Mutations in a variety of intracellular enzymes that lead to enhanced sodium reabsorption have been identified. In contrast, hypertension in Liddle's syndrome, which results from mutations in the Epithelial sodium Channel (ENaC), is associated with low plasma potassium and metabolic alkalosis. In Liddle's syndrome, truncation of one the ENaC protein subunits removes a binding site necessary protein for ubiquitination and degradation, thereby promoting accumulation along the apical membrane and enhanced sodium reabsorption. The myriad effects due to mutation in phosphodiesterase 3A (PDE3A) lead to severe hypertension underlying sodium-independent autosomal dominant hypertension with brachydactyly. How mutations in PDE3A result in the phenotypic features of this disorder are discussed. Understanding the pathologies of these monogenic hypertensive disorders may provide insight into the causes of the more prevalent essential hypertension and new avenues to unravel the complexities of blood pressure regulation.
Collapse
Affiliation(s)
- Peter E Levanovich
- Department of Physiology, Wayne State University School of Medicine and the John D. Dingell VA Medical Center, Detroit, Michigan, MI 48201, United States
| | - Alexander Diaczok
- Department of Internal Medicine, Wayne State University School of Medicine and the John D. Dingell VA Medical Center, Detroit, Michigan, MI 48201, United States
| | - Noreen F Rossi
- Department of Physiology, Wayne State University School of Medicine and the John D. Dingell VA Medical Center, Detroit, Michigan, MI 48201, United States
| |
Collapse
|
99962
|
West CL, Mao YK, Delungahawatta T, Amin JY, Farhin S, McQuade RM, Diwakarla S, Pustovit R, Stanisz AM, Bienenstock J, Barbut D, Zasloff M, Furness JB, Kunze WA. Squalamine Restores the Function of the Enteric Nervous System in Mouse Models of Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2020; 10:1477-1491. [PMID: 32925094 DOI: 10.3233/jpd-202076] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative disorder thought to be caused by accumulation of α-synuclein (α-syn) within the brain, autonomic nerves, and the enteric nervous system (ENS). Involvement of the ENS in PD often precedes the onset of the classic motor signs of PD by many years at a time when severe constipation represents a major morbidity. Studies conducted in vitro and in vivo, have shown that squalamine, a zwitterionic amphipathic aminosterol, originally isolated from the liver of the dogfish shark, effectively displaces membrane-bound α-syn. OBJECTIVE Here we explore the electrophysiological effect of squalamine on the gastrointestinal (GI) tract of mouse models of PD engineered to express the highly aggregating A53T human α-syn mutant. METHODS GI motility and in vivo response to oral squalamine in PD model mice and controls were assessed using an in vitro tissue motility protocol and via fecal pellet output. Vagal afferent response to squalamine was measured using extracellular mesenteric nerve recordings from the jejunum. Whole cell patch clamp was performed to measure response to squalamine in the myenteric plexus. RESULTS Squalamine effectively restores disordered colonic motility in vivo and within minutes of local application to the bowel. We show that topical squalamine exposure to intrinsic primary afferent neurons (IPANs) of the ENS rapidly restores excitability. CONCLUSION These observations may help to explain how squalamine may promote gut propulsive activity through local effects on IPANs in the ENS, and further support its possible utility in the treatment of constipation in patients with PD.
Collapse
Affiliation(s)
- Christine L West
- Brain-Body Institute, St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Yu-Kang Mao
- Brain-Body Institute, St. Joseph's Healthcare, Hamilton, ON, Canada
| | | | - Jessica Y Amin
- Brain-Body Institute, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Sohana Farhin
- Brain-Body Institute, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Rachel M McQuade
- Department of Anatomy and Neuroscience, University of Melbourne, and the Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Shanti Diwakarla
- Department of Anatomy and Neuroscience, University of Melbourne, and the Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Ruslan Pustovit
- Department of Anatomy and Neuroscience, University of Melbourne, and the Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Andrew M Stanisz
- Brain-Body Institute, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - John Bienenstock
- Brain-Body Institute, St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Michael Zasloff
- Enterin, Inc., Philadelphia, PA, USA.,MedStar-Georgetown Transplant Institute, Georgetown University School of Medicine, Washington, DC, USA
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne, and the Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Wolfgang A Kunze
- Brain-Body Institute, St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Biology, McMaster University, Hamilton, ON, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
99963
|
Styczen H, Fischer S, Yeo LL, Yong-Qiang Tan B, Maurer CJ, Berlis A, Abdullayev N, Kabbasch C, Kastrup A, Papanagiotou P, Clajus C, Lobsien D, Piechowiak E, Kaesmacher J, Maus V. Approaching the Boundaries of Endovascular Treatment in Acute Ischemic Stroke : Multicenter Experience with Mechanical Thrombectomy in Vertebrobasilar Artery Branch Occlusions. Clin Neuroradiol 2020; 31:791-798. [PMID: 33108470 DOI: 10.1007/s00062-020-00970-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Little is known about catheter-based endovascular treatment of vertebrobasilar artery branch occlusion (VEBABO) in acute ischemic stroke (AIS). Nonetheless, the experience of mechanical thrombectomy (MT) in distal small sized arteries of the anterior circulation seems promising in AIS. In this multicenter study, we report the feasibility, efficacy and safety of MT in VEBABO. METHODS Retrospective analysis of consecutive AIS patients treated with MT due to VEBABO including posterior and anterior inferior cerebellar artery (PICA, AICA) and superior cerebellar artery (SCA) occlusions at seven tertiary care centers between January 2013 and May 2020. Baseline demographics and angiographic outcomes including recanalization success of the affected cerebellar arteries and procedural complications were recorded. Clinical outcomes were evaluated by the modified Rankin scale (mRS) at discharge and 90 days. RESULTS Out of 668 endovascularly treated posterior circulation strokes we identified 16 (0.02%) cases with MT for VEBABO. Most frequently, MT of the SCA was done (13/16, 81%). Most VEBABOs occurred after MT of initial basilar/posterior cerebral artery occlusion (9/16, 56%). In 10/16 (63%) procedures, the affected VEBABO was successfully recanalized. Out of four patients three (75%) with isolated VEBABO had benefited from endovascular therapy. Subarachnoid hemorrhage was observed in 3/16 (19%) procedures. The rate of favorable outcome (mRS ≤2) was 40% at discharge and 47% at 90-day follow-up. Mortality was 13% (2/15). CONCLUSION The use of MT for VEBABO is rare but appears to be feasible and effective; however, the comparatively high rate of procedure-related hemorrhage highlights that the indications for MT in these occlusion sites should be carefully weighed up.
Collapse
Affiliation(s)
- Hanna Styczen
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstraße 55, 45122, Essen, Germany.
| | - Sebastian Fischer
- Department of Radiology, Neuroradiology and Nuclear Medicine, Knappschaftskrankenhaus Langendreer, Ruhr-University Bochum, Bochum, Germany
| | - Leonard Ll Yeo
- Division of Neurology, Department of Medicine, National University Health System, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Benjamin Yong-Qiang Tan
- Division of Neurology, Department of Medicine, National University Health System, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christoph J Maurer
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Ansgar Berlis
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Nuran Abdullayev
- Department of Diagnostic and Interventional Radiology, University Hospital Cologne, Cologne, Germany
| | - Christoph Kabbasch
- Department of Diagnostic and Interventional Radiology, University Hospital Cologne, Cologne, Germany
| | | | - Panagiotis Papanagiotou
- Department of Diagnostic and Interventional Neuroradiology, Klinikum Bremen-Mitte, Bremen, Germany.,Department of Radiology, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christin Clajus
- Department of Diagnostic and Interventional Radiology and Neuroradiology, Helios General Hospital Erfurt, Erfurt, Germany
| | - Donald Lobsien
- Department of Diagnostic and Interventional Radiology and Neuroradiology, Helios General Hospital Erfurt, Erfurt, Germany
| | - Eike Piechowiak
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Johannes Kaesmacher
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland.,University Institute of Diagnostic and Interventional and Pediatric Radiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Volker Maus
- Department of Radiology, Neuroradiology and Nuclear Medicine, Knappschaftskrankenhaus Langendreer, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
99964
|
Beyens A, Boel A, Symoens S, Callewaert B. Cutis laxa: A comprehensive overview of clinical characteristics and pathophysiology. Clin Genet 2020; 99:53-66. [PMID: 33058140 DOI: 10.1111/cge.13865] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/17/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022]
Abstract
Cutis laxa (CL) syndromes comprise a rare group of multisystem disorders that share loose redundant skin folds as hallmark clinical feature. CL results from impaired elastic fiber assembly and homeostasis, and the known underlying gene defects affect different extracellular matrix proteins, intracellular trafficking, or cellular metabolism. Due to the underlying clinical and molecular heterogeneity, the diagnostic work-up of CL patients is often challenging. In this review, we provide a practical approach to the broad differential diagnosis of CL syndromes, provide an overview of the molecular pathogenesis of the different subtypes, and suggest general management guidelines.
Collapse
Affiliation(s)
- Aude Beyens
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium.,Department of Dermatology, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Annekatrien Boel
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Sofie Symoens
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
99965
|
Pietroboni AM, Colombi A, Carandini T, Scarpini E, Galimberti D, Bozzali M. The Role of Amyloid-β in White Matter Damage: Possible Common Pathogenetic Mechanisms in Neurodegenerative and Demyelinating Diseases. J Alzheimers Dis 2020; 78:13-22. [PMID: 32925075 DOI: 10.3233/jad-200868] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Just as multiple sclerosis (MS) has long been primarily considered a white matter (WM) disease, Alzheimer's disease (AD) has for decades been regarded only as a grey matter disorder. However, convergent evidences have suggested that WM abnormalities are also important components of AD, at the same extent as axonal and neuronal loss is critically involved in MS pathophysiology since early clinical stages. These observations have motivated a more thorough investigation about the possible mechanisms that could link neuroinflammation and neurodegeneration, focusing on amyloid-β (Aβ). Neuroimaging studies have found that patients with AD have widespread WM abnormalities already at the earliest disease stages and prior to the presence of Aβ plaques. Moreover, a correlation between cerebrospinal fluid (CSF) Aβ levels and WM lesion load was found. On the other hand, recent studies suggest a predictive role for CSF Aβ levels in MS, possibly due in the first instance to the reduced capacity for remyelination, consequently to a higher risk of WM damage progression, and ultimately to neuronal loss. We undertook a review of the recent findings concerning the involvement of CSF Aβ levels in the MS disease course and of the latest evidence of AD related WM abnormalities, with the aim to discuss the potential causes that may connect WM damage and amyloid pathology.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | | | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Marco Bozzali
- Department of Neuroscience 'Rita Levi Montalcini', University of Torino, Turin, Italy.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
99966
|
Möhn N, Luo Y, Skripuletz T, Schwenkenbecher P, Ladwig A, Warnke C, Meuth SG, Wiendl H, Gross CC, Schröder C, Haghikia A, Stangel M. Cerebrospinal fluid analysis in 108 patients with progressive multifocal leukoencephalopathy. Fluids Barriers CNS 2020; 17:65. [PMID: 33109245 PMCID: PMC7590675 DOI: 10.1186/s12987-020-00227-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/17/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Progressive multifocal leukoencephalopathy (PML) is caused by an opportunistic infection with JC polyoma virus (JCPyV) and mainly affects immunocompromised patients. It leads to pronounced demyelination of the central nervous system (CNS) resulting in severe disability or even death. Detection of JCPyV DNA in the cerebrospinal fluid (CSF) is usually accepted as proof for the diagnosis of PML. Routine CSF parameters, like CSF cell count, protein concentration, Qalbumin, or intrathecal immunoglobulin synthesis are mostly considered normal. However, this has not been investigated systematically. METHODS We analyzed routine CSF parameters in a cohort of 108 PML patients that were treated at four different neurological centers in Germany. The patients exhibited different underlying conditions with natalizumab-treated multiple sclerosis (n = 54) and human immunodeficiency virus (HIV)-infection (n = 25) being the most frequent. The data were collected at the respective centers in accordance with local requirements and then jointly analyzed. The total PML cohort was compared with a control group of patients with normal pressure hydrocephalus (NPH) and idiopathic intracranial hypertension (IIH). Multiple sclerosis and HIV patients were additionally compared with their own non-PML control groups. RESULTS The PML group showed an elevated cell count (p < 0.001) compared to the control group, however, this effect was mainly driven by HIV-PML patients. This subgroup also demonstrated a significantly higher proportion of patients with a disturbed blood-CSF-barrier function. CONCLUSIONS This comprehensive, retrospective study on CSF diagnostic analysis in PML patients provides insight into the CSF of those patients. It demonstrates that CSF composition in PML patients may be specific for the underlying condition that predisposes for the development of PML and thus data have to be interpreted in this context.
Collapse
Affiliation(s)
- Nora Möhn
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Yi Luo
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Skripuletz
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Philipp Schwenkenbecher
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Anne Ladwig
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Clemens Warnke
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Christoph Schröder
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital Bochum, Bochum, Germany
| | - Aiden Haghikia
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital Bochum, Bochum, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
99967
|
Alyenbaawi H, Allison WT, Mok SA. Prion-Like Propagation Mechanisms in Tauopathies and Traumatic Brain Injury: Challenges and Prospects. Biomolecules 2020; 10:E1487. [PMID: 33121065 PMCID: PMC7692808 DOI: 10.3390/biom10111487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022] Open
Abstract
The accumulation of tau protein in the form of filamentous aggregates is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). These dementias share traumatic brain injury (TBI) as a prominent risk factor. Tau aggregates can transfer between cells and tissues in a "prion-like" manner, where they initiate the templated misfolding of normal tau molecules. This enables the spread of tau pathology to distinct parts of the brain. The evidence that tauopathies spread via prion-like mechanisms is considerable, but work detailing the mechanisms of spread has mostly used in vitro platforms that cannot fully reveal the tissue-level vectors or etiology of progression. We review these issues and then briefly use TBI and CTE as a case study to illustrate aspects of tauopathy that warrant further attention in vivo. These include seizures and sleep/wake disturbances, emphasizing the urgent need for improved animal models. Dissecting these mechanisms of tauopathy progression continues to provide fresh inspiration for the design of diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hadeel Alyenbaawi
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Medical Laboratories, Majmaah University, Majmaah 11952, Saudi Arabia
| | - W. Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Sue-Ann Mok
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
99968
|
Bergoug M, Doudeau M, Godin F, Mosrin C, Vallée B, Bénédetti H. Neurofibromin Structure, Functions and Regulation. Cells 2020; 9:cells9112365. [PMID: 33121128 PMCID: PMC7692384 DOI: 10.3390/cells9112365] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Neurofibromin is a large and multifunctional protein encoded by the tumor suppressor gene NF1, mutations of which cause the tumor predisposition syndrome neurofibromatosis type 1 (NF1). Over the last three decades, studies of neurofibromin structure, interacting partners, and functions have shown that it is involved in several cell signaling pathways, including the Ras/MAPK, Akt/mTOR, ROCK/LIMK/cofilin, and cAMP/PKA pathways, and regulates many fundamental cellular processes, such as proliferation and migration, cytoskeletal dynamics, neurite outgrowth, dendritic-spine density, and dopamine levels. The crystallographic structure has been resolved for two of its functional domains, GRD (GAP-related (GTPase-activating protein) domain) and SecPH, and its post-translational modifications studied, showing it to be localized to several cell compartments. These findings have been of particular interest in the identification of many therapeutic targets and in the proposal of various therapeutic strategies to treat the symptoms of NF1. In this review, we provide an overview of the literature on neurofibromin structure, function, interactions, and regulation and highlight the relationships between them.
Collapse
|
99969
|
Menozzi E, Macnaughtan J, Schapira AHV. LRRK2 Parkinsonism: Does the Response to Gut Bacteria Mitigate the Neurological Picture? Mov Disord 2020; 36:71-75. [PMID: 33107648 DOI: 10.1002/mds.28347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/02/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Jane Macnaughtan
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
99970
|
Abstract
Migraine headaches can be a disabling condition for patients. Fortunately, most patients can be successfully managed in the outpatient setting, however, there are a number of patients who may not respond to the abortive treatments that they have been prescribed. These patients often present to the emergency department (ED) for further assistance with the management of their condition. Migraines are the fourth most common cause of ED visits and are associated with an estimated annual cost of $17 billion in the United States. Familiarity with abortive treatments is critical for providers in the ED as are treatments, such as valproic acid, that may be considered in patients who do not respond to other treatment options. Many providers are more familiar with the role of valproic acid in the treatment of mood and seizure disorders, but its tolerability and the successes reported in the primary literature make it a reasonable consideration for patients with migraine who fail to respond to other therapies. This article briefly summarizes the therapies considered first line for abortive treatment in the setting of migraines and provides an overview of the primary literature describing the use of valproic acid in these patients.
Collapse
|
99971
|
Hallab A, Lange C, Apostolova I, Özden C, Gonzalez-Escamilla G, Klutmann S, Brenner W, Grothe MJ, Buchert R. Impairment of Everyday Spatial Navigation Abilities in Mild Cognitive Impairment Is Weakly Associated with Reduced Grey Matter Volume in the Medial Part of the Entorhinal Cortex. J Alzheimers Dis 2020; 78:1149-1159. [PMID: 33104026 DOI: 10.3233/jad-200520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Research in rodents identified specific neuron populations encoding information for spatial navigation with particularly high density in the medial part of the entorhinal cortex (ERC), which may be homologous with Brodmann area 34 (BA34) in the human brain. OBJECTIVE The aim of this study was to test whether impaired spatial navigation frequently occurring in mild cognitive impairment (MCI) is specifically associated with neurodegeneration in BA34. METHODS The study included baseline data of MCI patients enrolled in the Alzheimer's Disease Neuroimaging Initiative with high-resolution structural MRI, brain FDG PET, and complete visuospatial ability scores of the Everyday Cognition test (VS-ECog) within 30 days of PET. A standard mask of BA34 predefined in MNI space was mapped to individual native space to determine grey matter volume and metabolic activity in BA34 on MRI and on (partial volume corrected) FDG PET, respectively. The association of the VS-ECog sum score with grey matter volume and metabolic activity in BA34, APOE4 carrier status, age, education, and global cognition (ADAS-cog-13 score) was tested by linear regression. BA28, which constitutes the lateral part of the ERC, was used as control region. RESULTS The eligibility criteria led to inclusion of 379 MCI subjects. The VS-ECog sum score was negatively correlated with grey matter volume in BA34 (β= -0.229, p = 0.022) and age (β= -0.124, p = 0.036), and was positively correlated with ADAS-cog-13 (β= 0.175, p = 0.003). None of the other predictor variables contributed significantly. CONCLUSION Impairment of spatial navigation in MCI is weakly associated with BA34 atrophy.
Collapse
Affiliation(s)
- Asma Hallab
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Catharina Lange
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ivayla Apostolova
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cansu Özden
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriel Gonzalez-Escamilla
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Rostock, Germany.,Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Susanne Klutmann
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Rostock, Germany.,Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
99972
|
Castiglione JI, Marrodan M, Alessandro L, Taratuto AL, Brand P, Nogués M, Barroso F. Vasculitic Peripheral Neuropathy, Differences Between Systemic and Non-Systemic Etiologies: A Case Series and Biopsy Report. J Neuromuscul Dis 2020; 8:155-161. [PMID: 33104037 DOI: 10.3233/jnd-200576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Vasculitic peripheral neuropathy (VPN) is caused by vessel inflammation leading to peripheral nerve injury of acute-to-subacute onset. When VPN occurs in the context of systemic disease it is classified as Systemic Vasculitic Neuropathy (SVN) and as Non-Systemic Vasculitic Neuropathy (NSVN) when restricted to the nerves. OBJECTIVE This study aimed to compare the clinical characteristics, biopsy findings and disease outcome in patients with VPN. METHODS Clinical records of adult patients with VPN diagnosed at our institution between June-2002 and June-2019 were retrospectively reviewed. Demographic characteristics, clinical manifestations, nerve conduction studies, nerve biopsies, treatment and clinical evolution were analyzed in all patients with at least 6 months follow-up. RESULTS Twenty-five patients with VPN were included (SVN, n = 10; NSVN, n = 15). No significant differences in demographic or clinical features were found between groups. The median delay between symptom onset and nerve biopsy was significantly longer in NSVN patients (10 vs 5.5 months, p = 0.009). Erythrocyte sedimentation rate (ESR) values over 20 mm/h were significantly more common in SVN patients (100% vs. 60%, p = 0.024). Nerve biopsies showed active lesions more frequently in treatment-naive patients compared to those who had received at least 2 weeks of corticosteroids (92% vs 38%; p = 0.03), with a higher proportion of definite VPN cases (92 vs 46%; p = 0.04). CONCLUSIONS Although the clinical manifestations are similar, ESR is an important tool to help distinguish between both conditions. Early nerve biopsy in untreated patients increases diagnostic accuracy, avoiding misdiagnosis.
Collapse
Affiliation(s)
| | - M Marrodan
- Neurology department, FLENI, Buenos Aires, Argentina
| | - L Alessandro
- Neurology department, FLENI, Buenos Aires, Argentina
| | - A L Taratuto
- Neuropathology department, FLENI, Buenos Aires, Argentina
| | - P Brand
- Neurology department, FLENI, Buenos Aires, Argentina
| | - M Nogués
- Neurology department, FLENI, Buenos Aires, Argentina
| | - F Barroso
- Neurology department, FLENI, Buenos Aires, Argentina
| |
Collapse
|
99973
|
Belter L, Jarecki J, Reyna SP, Cruz R, Jones CC, Schroth M, O'Toole CM, O'Brien S, Hall SA, Johnson NB, Paradis AD. The Cure SMA Membership Surveys: Highlights of Key Demographic and Clinical Characteristics of Individuals with Spinal Muscular Atrophy. J Neuromuscul Dis 2020; 8:109-123. [PMID: 33104036 PMCID: PMC7902958 DOI: 10.3233/jnd-200563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background: Cure SMA maintains the largest patient-reported database for people affected with spinal muscular atrophy (SMA). In 2017, Cure SMA initiated annual surveys with their membership to collect demographic and disease characteristics, healthcare, and burden of disease information from patients and caregivers. Objective: To summarize results from two large-scale Cure SMA surveys in 2017 and 2018. Methods: Cure SMA database members were invited to complete surveys; these were completed by caregivers for living or deceased individuals with SMA and/or affected adults. Results: In 2017, 726 surveys were completed for 695 individuals with SMA; in 2018, 796 surveys were completed for 760 individuals with SMA. Data from both survey years are available for 313 affected individuals. Age at symptom onset, distribution of SMN2 gene copy number, and representation of each SMA type in the surveys were consistent with that expected in the SMA population. In the 2018 survey, the average age at diagnosis was 5.2 months for SMA type I and the reported mean age at death for this subgroup was 27.8 months. Between survey years, there was consistency in responses for factors that should not change within individuals over time (e.g., reported age at diagnosis). Conclusions: Results from the Cure SMA surveys advance the understanding of SMA and facilitate advocacy efforts and healthcare services planning. Longitudinal surveys are important for evaluating the impact of effective treatments on changing phenotypes, and burden of disease and care in individuals with SMA.
Collapse
|
99974
|
Ma X, Ma L, Wang Z, Liu Y, Long L, Ma X, Chen H, Chen Z, Lin X, Si L, Chen X. Clinical Features and Gut Microbial Alterations in Anti-leucine-rich Glioma-Inactivated 1 Encephalitis-A Pilot Study. Front Neurol 2020; 11:585977. [PMID: 33193049 PMCID: PMC7655127 DOI: 10.3389/fneur.2020.585977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Anti-leucine-rich glioma-inactivated 1 (anti-LGI1) encephalitis is a rare autoimmune encephalitis (AE). We investigated the clinical features and gut microbial alterations of anti-LGI1 encephalitis. Fifteen patients newly diagnosed with anti-LGI1 encephalitis were recruited in the study prior to the administration of immunotherapy. The control group contains 25 well-matched healthy controls (HCs). All participants were Han Chinese from South China. Their clinical data and fecal samples were collected. The diversity and composition of gut microbiota were analyzed by 16S ribosomal RNA (16S rRNA) gene sequencing. The results showed that anti-LGI1 encephalitis was characterized by cognitive impairment, faciobrachial dystonic seizures, hyponatremia, and psychiatric symptoms. Abnormal EEG and brain MRI were presented in 9 and 10 patients, respectively. Compared to HCs, the anti-LGI1 encephalitis patients exhibited a decreased microbial diversity and an altered overall composition of gut microbiome. At the phylum level, anti-LGI1 encephalitis patients exhibited a higher abundance of Proteobacteria and a lower abundance of Firmicutes. The alterations in the phylum level were associated with autoimmune and inflammatory disorders. At the genus level, there was an increase in Sphingomonas, Anaerofustis, Succinvibrio, Clostridium, and SMB53 (genera related to movement disorders, psychiatric diseases, and with proinflammatory effects). However, the Faecalibacterium, Roseburia, Lachnospira, Ruminococcus, and Blautia [genera with ability to produce short-chain fatty acids (SCFAs)] were obviously reduced in the patient group. Our results suggest that anti-LGI1 encephalitis is characterized by special clinical features and is accompanied by alterations in specific gut microbiota. For the limited sample size and non-applicability to other populations, further studies are warranted to explore the relationships between gut microbiota and anti-LGI1 encephalitis.
Collapse
Affiliation(s)
- Xueying Ma
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lili Ma
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhanhang Wang
- Department of Neurology, Guangdong 999 Brain Hospital, Guangzhou, China
| | - Yingying Liu
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ling Long
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaomeng Ma
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Chen
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaoyu Chen
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiuli Lin
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Si
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Chen
- Department of Neurology and Multiple Sclerosis Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
99975
|
Melikyan AG, Vorobiev AN, Shishkina LV, Kozlova AB, Vlasov PA, Ayvazyan SO, Shults EI, Korsakova MB, Koptelova AM, Buklina SB, Demin MO, Agrba SB, Shevchenko AM. [Surgical treatment of epilepsy in children with focal cortical dysplasia]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2020; 84:5-20. [PMID: 33095529 DOI: 10.17116/neiro2020840515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Surgery is the first-line treatment option in children with FCD and refractory epilepsy, but the rate of success and patient numbers who became free of seizures vary widely from series to series. STUDY AIMS To elicit variables affecting the outcome and predicting achievement of the long-term seizure-free status. MATERIAL AND METHODS One hundred sixty-nine children with cortical dysplasia and DR-epilepsy underwent surgery Preoperative evaluation included prolonged video-EEG and MRI (in all patients) and neuropsychological testing when possible. Fourteen patients underwent invasive EEG, fMRI and MEG were used also in some cases. Including 27 repeat procedures the list of overall 196 surgeries performed consists of: cortectomy (lesionectomy with or without adjacent epileptogenic cortices) – in 116 cases; lobectomy – in 46; and various disconnective procedures – in 34 patients. Almost routinely employed intraoperative ECOG (134 surgeries) was combined with stimulation and/or SSEP in 47 cases to map eloquent cortex (with CST-tracking in some). A new permanent and not anticipated neurological deficit developed post-surgery in 5 cases (2,5%). Patients were follow-upped using video-EEG and MRI and FU which lasts more than 2 years (median – 3 years) is known in 56 cases. Thirty-two children were free of seizures at the last check (57,2% rate of Engel IA). A list of variables regarding patients’ demography, seizure type, lesion pathology and localization, and those related to surgery and its extent were evaluated to figure out anyone associated with favorable outcome. RESULTS Both Type II FCDs and their anatomically complete excision are positive predictors for favorable outcome and achievement of SF-status (p<0,05). Residual epileptic activity on immediate post-resection ECOG do not affect the outcome. CONCLUSION Patients with Type II FCD, particularly with Type IIb malformations are the best candidates for curative surgery, including cases with lesions in brain eloquent areas. Kids with Type I FCD have much less chances to become free of seizures when attempting focal cortectomy. However, some of them with early onset catastrophic epilepsies may benefit from larger surgeries using lobectomy or various disconnections.
Collapse
Affiliation(s)
| | | | | | - A B Kozlova
- Burdenko Neurosurgical Center, Moscow, Russia
| | - P A Vlasov
- Burdenko Neurosurgical Center, Moscow, Russia
| | | | - E I Shults
- Burdenko Neurosurgical Center, Moscow, Russia
| | | | - A M Koptelova
- Center for Neurocognitive research (MEG-center), MSUPE, Moscow, Russia
| | - S B Buklina
- Burdenko Neurosurgical Center, Moscow, Russia
| | - M O Demin
- Burdenko Neurosurgical Center, Moscow, Russia
| | - S B Agrba
- Burdenko Neurosurgical Center, Moscow, Russia
| | | |
Collapse
|
99976
|
Molazadeh N, Mohebi F, Altafi D, Sahraian MA. Prevalence and incidence of multiple sclerosis in Ardabil, Northwest of Iran. Mult Scler Relat Disord 2020; 47:102605. [PMID: 33217696 DOI: 10.1016/j.msard.2020.102605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/17/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) epidemiology is studies in many populations; however, studying populations with unique characteristics could provide opportunities to deepen the understanding of the underlying reason of the disease. In this regard, we aimed to study the epidemiology of MS in Ardabil, a province in northwest Iran, where the majority are of Iranian Azerbaijanis. METHOD A retrospective population-based study was conducted from 2008 to 2018 in Ardabil, based on the data of Iran's Ministry Of Health. Collected information includes sex, age, age at disease onset, education and type of MS. We used t-test to compare means and chi-square test to analyze the association among variables. RESULTS The total number of patients was 760 with 533 (70.13%) females and 227 (29.87%) males. The crude prevalence was 59.37 per 100,000 in 2018 (95% CI: 55.31, 63.73). The crude incidence rate was 7.65 per 100,000 in 2018 (95% CI: 6.28, 9.32). The most frequent educational level was high school diploma (38.36%). The relapsing-remitting (RR) form was the most frequent type of MS (48.16%). The F/M ratio was 2.92:1 and the mean onset age was 33.14 (95% CI: 31.56, 34.72) in 2018, significantly higher in males (P value= 0.01). CONCLUSION Ardabil is a medium risk zone of MS. The different ethnicity of its population, climate and environmental features, may explain the factors modulating the risk of MS in similar areas and present windows of opportunity to understand the causes of MS.
Collapse
Affiliation(s)
- Negar Molazadeh
- MS Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnam Mohebi
- Non-communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Davar Altafi
- Department of Neurology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Ali Sahraian
- MS Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
99977
|
Fan Z, Li Z, Shen F, Zhang X, Lei L, Su S, Lu Y, Di L, Wang M, Xu M, Da Y. Favorable Effects of Tacrolimus Monotherapy on Myasthenia Gravis Patients. Front Neurol 2020; 11:594152. [PMID: 33193063 PMCID: PMC7652845 DOI: 10.3389/fneur.2020.594152] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Background and Purpose: Tacrolimus (TAC) has been proven to be a rapid-acting, steroid-sparing agent for myasthenia gravis (MG) therapy. However, evidence related to the effectiveness of TAC alone is rare. Therefore, this study was performed to investigate the effect of TAC monotherapy in MG patients. Methods: Forty-four MG patients who received TAC monotherapy were retrospectively analyzed. A mixed effect model was used to analyze improvements in MG-specific activities of daily living scale (MG-ADL), quantitative MG score (QMG) and MG-ADL subscores. Kaplan-Meier analysis was used to estimate the cumulative probability of minimal manifestations (MM) or better. Adverse events (AEs) were recorded for safety analyses. Results: Of the patients receiving TAC monotherapy, MG-ADL scores were remarkably improved at 3, 6 and 12 months compared with scores at baseline (mean difference and 95% CIs: −3.29 [−4.94, −1.64], −3.97 [−5.67, −2.27], and −4.67 [−6.48, −2.85], respectively). QMG scores significantly decreased at 6 and 12 months, with mean differences and 95% CIs of −4.67(−6.88, −2.45) and −5.77 (−7.55, −4.00), respectively. Estimated median period to achieve “MM or better” was 5.0 (95% CIs, 2.8, 7.2) months. Ocular MG (OMG) and generalized MG (GMG) showed similar therapeutic effects in cumulative probabilities of “MM or better” (P-value = 0.764). A better response was observed in MG-ADL subscores for ptosis and bulbar symptoms. AEs occurred in 37.5% of patients and were generally mild and reversible. Conclusions: TAC monotherapy is a promising option to rapidly alleviate all symptoms of MG, especially for ptosis and bulbar symptoms.
Collapse
Affiliation(s)
- Zhirong Fan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zunbo Li
- Department of Neurology, Xi'an Gaoxin Hospital, Xi'an, China
| | - Faxiu Shen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xueping Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lin Lei
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shengyao Su
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yan Lu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Li Di
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Xu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
99978
|
Spotorno N, Lindberg O, Nilsson C, Landqvist Waldö M, van Westen D, Nilsson K, Vestberg S, Englund E, Zetterberg H, Blennow K, Lätt J, Markus N, Lars-Olof W, Alexander S. Plasma neurofilament light protein correlates with diffusion tensor imaging metrics in frontotemporal dementia. PLoS One 2020; 15:e0236384. [PMID: 33108404 PMCID: PMC7591030 DOI: 10.1371/journal.pone.0236384] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/12/2020] [Indexed: 12/02/2022] Open
Abstract
Neurofilaments are structural components of neurons and are particularly abundant in highly myelinated axons. The levels of neurofilament light chain (NfL) in both cerebrospinal fluid (CSF) and plasma have been related to degeneration in several neurodegenerative conditions including frontotemporal dementia (FTD) and NfL is currently considered as the most promising diagnostic and prognostic fluid biomarker in FTD. Although the location and function of filaments in the healthy nervous system suggests a link between increased NfL and white matter degeneration, such a claim has not been fully elucidated in vivo, especially in the context of FTD. The present study provides evidence of an association between the plasma levels of NfL and white matter involvement in behavioral variant FTD (bvFTD) by relating plasma concentration of NfL to diffusion tensor imaging (DTI) metrics in a group of 20 bvFTD patients. The results of both voxel-wise and tract specific analysis showed that increased plasma NfL concentration is associated with a reduction in fractional anisotropy (FA) in a widespread set of white matter tracts including the superior longitudinal fasciculus, the fronto-occipital fasciculus the anterior thalamic radiation and the dorsal cingulum bundle. Plasma NfL concentration also correlated with cortical thinning in a portion of the right medial prefrontal cortex and of the right lateral orbitofrontal cortex. These results support the hypothesis that blood NfL levels reflect the global level of neurodegeneration in bvFTD and help to advance our understanding of the association between this blood biomarker for FTD and the disease process.
Collapse
Affiliation(s)
- Nicola Spotorno
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Olof Lindberg
- Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden
| | - Christer Nilsson
- Division of Neurology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Landqvist Waldö
- Department of clinical Sciences, Clinical Sciences Helsingborg, Lund, Lund University, Lund, Sweden
| | - Danielle van Westen
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Karin Nilsson
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | | | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jimmy Lätt
- Center for Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
| | - Nilsson Markus
- Department of Diagnostic Radiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Wahlund Lars-Olof
- Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden
| | - Santillo Alexander
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Malmö, Sweden
| |
Collapse
|
99979
|
Katagiri N, Kawakami S, Okuyama S, Koseki T, Kudo D, Namba S, Tanabe S, Yamaguchi T. Single-Session Cerebellar Transcranial Direct Current Stimulation Affects Postural Control Learning and Cerebellar Brain Inhibition in Healthy Individuals. THE CEREBELLUM 2020; 20:203-211. [PMID: 33108574 DOI: 10.1007/s12311-020-01208-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
Cerebellar transcranial direct current stimulation (ctDCS) modulates cerebellar activity and postural control. However, the effects of ctDCS on postural control learning and the mechanisms associated with these effects remain unclear. To examine the effects of single-session ctDCS on postural control learning and cerebellar brain inhibition (CBI) of the primary motor cortex in healthy individuals. In this triple-blind, sham-controlled study, 36 participants were allocated randomly to one of three groups: (1) anodal ctDCS group, (2) cathodal ctDCS group, and (3) sham ctDCS group. ctDCS (2 mA) was applied to the cerebellar brain for 20 min prior to six blocks of standing postural control training (each block consisted of five trials of a 30-s tracking task). CBI and corticospinal excitability of the tibialis anterior muscle were assessed at baseline, immediately after, 1 day after, and 7 days after training. Skill acquisition following training was significantly reduced in both the anodal and cathodal ctDCS groups compared with the sham ctDCS group. Changes in performance measured 1 day after and 7 days after training did not differ among the groups. In the anodal ctDCS group, CBI significantly increased after training, whereas corticospinal excitability decreased. Anodal ctDCS-induced CBI changes were correlated with the learning formation of postural control (r = 0.55, P = 0.04). Single-session anodal and cathodal ctDCS could suppress the skill acquisition of postural control in healthy individuals. The CBI changes induced by anodal ctDCS may affect the learning process of postural control.
Collapse
Affiliation(s)
- Natsuki Katagiri
- Department of Physical Therapy, Graduate School of Health Sciences, Yamagata Prefectural University of Health Sciences, 260 Kamiyanagi, Yamagata-shi, Yamagata, 990-2212, Japan
| | - Saki Kawakami
- Department of Physical Therapy, Graduate School of Health Sciences, Yamagata Prefectural University of Health Sciences, 260 Kamiyanagi, Yamagata-shi, Yamagata, 990-2212, Japan
| | - Sayuri Okuyama
- Department of Physical Therapy, Graduate School of Health Sciences, Yamagata Prefectural University of Health Sciences, 260 Kamiyanagi, Yamagata-shi, Yamagata, 990-2212, Japan
| | - Tadaki Koseki
- Department of Physical Therapy, Graduate School of Health Sciences, Yamagata Prefectural University of Health Sciences, 260 Kamiyanagi, Yamagata-shi, Yamagata, 990-2212, Japan
| | - Daisuke Kudo
- Department of Physical Therapy, Graduate School of Health Sciences, Yamagata Prefectural University of Health Sciences, 260 Kamiyanagi, Yamagata-shi, Yamagata, 990-2212, Japan
| | - Shigehiro Namba
- Department of Physical Therapy, Graduate School of Health Sciences, Yamagata Prefectural University of Health Sciences, 260 Kamiyanagi, Yamagata-shi, Yamagata, 990-2212, Japan
| | - Shigeo Tanabe
- Faculty of Rehabilitation, School of Health Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake-shi, Aichi, 470-1192, Japan
| | - Tomofumi Yamaguchi
- Department of Physical Therapy, Graduate School of Health Sciences, Yamagata Prefectural University of Health Sciences, 260 Kamiyanagi, Yamagata-shi, Yamagata, 990-2212, Japan. .,Department of Physical Therapy, Faculty of Health Science, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
99980
|
Strik M, Chard DT, Dekker I, Meijer KA, Eijlers AJ, Pardini M, Uitdehaag BM, Kolbe SC, Geurts JJ, Schoonheim MM. Increased functional sensorimotor network efficiency relates to disability in multiple sclerosis. Mult Scler 2020; 27:1364-1373. [PMID: 33104448 PMCID: PMC8358536 DOI: 10.1177/1352458520966292] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Network abnormalities could help explain physical disability in multiple sclerosis (MS), which remains poorly understood. OBJECTIVE This study investigates functional network efficiency changes in the sensorimotor system. METHODS We included 222 MS patients, divided into low disability (LD, Expanded Disability Status Scale (EDSS) ⩽3.5, n = 185) and high disability (HD, EDSS ⩾6, n = 37), and 82 healthy controls (HC). Functional connectivity was assessed between 23 sensorimotor regions. Measures of efficiency were computed and compared between groups using general linear models corrected for age and sex. Binary logistic regression models related disability status to local functional network efficiency (LE), brain volumes and demographics. Functional connectivity patterns of regions important for disability were explored. RESULTS HD patients demonstrated significantly higher LE of the left primary somatosensory cortex (S1) and right pallidum compared to LD and HC, and left premotor cortex compared to HC only. The logistic regression model for disability (R2 = 0.38) included age, deep grey matter volume and left S1 LE. S1 functional connectivity was increased with prefrontal and secondary sensory areas in HD patients, compared to LD and HC. CONCLUSION Clinical disability in MS associates with functional sensorimotor increases in efficiency and connectivity, centred around S1, independent of structural damage.
Collapse
Affiliation(s)
- Myrte Strik
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands/Department of Radiology and Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Declan T Chard
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, London, UK/National Institute for Health Research, University College London Hospitals Biomedical Research Centre, London, UK
| | - Iris Dekker
- Department of Neurology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands/Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kim A Meijer
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anand Jc Eijlers
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Matteo Pardini
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Institute of Neurology, London, UK/Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genoa, Genoa, Italy/Ospedale Policlinico San Martino-IRCCS, Genoa, Italy
| | - Bernard Mj Uitdehaag
- Department of Neurology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Scott C Kolbe
- Department of Radiology and Medicine, The University of Melbourne, Melbourne, VIC, Australia/Department of Neurosciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jeroen Jg Geurts
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
99981
|
Briggs FBS, Hill E, Abboud H. The prevalence of hypertension in multiple sclerosis based on 37 million electronic health records from the United States. Eur J Neurol 2020; 28:558-566. [PMID: 32981133 DOI: 10.1111/ene.14557] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Hypertension (HTN) is a common comorbidity in multiple sclerosis (MS), and it significantly contributes to adverse outcomes. Unfortunately, the distribution of HTN in persons with MS has not been well characterized, and prior estimates have primarily relied on modest sample sizes. The objective of this study was to robustly describe the distribution of HTN in the MS population in comparison to the non-MS population with considerations for age, sex, and race. To date, this is the largest investigation of its kind. METHODS We conducted a cross-sectional study of 37 million unique electronic health records available in the IBM Explorys Enterprise Performance Management: Explore database (Explorys) spanning the United States. This resource has previously been validated for use in MS. We evaluated the prevalence of HTN in MS (N = 122 660) and non-MS (N = 37 075 350) cohorts, stratifying by age, sex, and race. RESULTS The prevalence of HTN was significantly greater among those with MS than among those without MS across age, sex, and race subpopulations, even after adjusting for age and sex. HTN was 25% more common in MS. In both MS and non-MS cohorts, the prevalence of HTN progressively increased with age and was higher in Black Americans and in males. DISCUSSION This study demonstrated that HTN is significantly more common in the MS population compared to the non-MS population, irrespective of sex and race. Because HTN is the leading global risk factor for disability and death, these results emphasize the need for aggressive screening for, and management of, HTN in the MS population.
Collapse
Affiliation(s)
- F B S Briggs
- Neuroimmunological Disorders Gene-Environment Epidemiology Laboratory, Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - E Hill
- Neuroimmunological Disorders Gene-Environment Epidemiology Laboratory, Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - H Abboud
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Multiple Sclerosis and Neuroimmunology Program, University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
99982
|
Yu X, Xie Y. Effect of dexmedetomidine combined with etomidate on IL-17A and S-100β expression levels in rats with postoperative cognitive dysfunction. Exp Ther Med 2020; 20:275. [PMID: 33200000 DOI: 10.3892/etm.2020.9405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 08/03/2020] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to explore the effects of dexmedetomidine combined with etomidate on the expression levels of interleukin (IL)-17A and S-100β in rats with postoperative cognitive dysfunction (POCD). A total of 50 SD rats were randomly allocated into the control group, model group, etomidate group (Eto group), dexmedetomidine group (Dex group) and dexmedetomidine combined with etomidate group (Dex-Eto group). Inhalation anesthesia was used in all five groups. Apart from the control group, partial lobectomy was performed to construct a rat model of cognitive dysfunction. The rats of the model group received no intravenous anesthesia, except general anesthesia with intubation. Morris water maze test was performed before injection (T0), at the 1st day (T1), the 3rd day (T2) and the 5th day (T3) after operation to assess the memory ability of the rats. At the end of T3, the expression levels of IL-17A, S-100β, TNF-α, IL-6 and IL-1β in serum were detected by ELISA and the expression of NF-κB p65 by western blot analysis. Compared with the control group, the model group showed an increased escape latency and swimming distance, decreased number of times of crossing the platform and target quadrant residence time, and increased expression levels of IL-17A, S-100β, TNF-α, IL-6, IL-1β and NF-κB p65. Compared with the model group, the escape latency and swimming distance in the Dex, Eto and Dex-Eto groups were reduced, whereas the number of times of crossing the platform and the target quadrant residence time were increased. In addition, the expression levels of IL-17A, S-100β, TNF-α, IL-6, IL-1β and NF-κB p65 were decreased in the Dex, Eto and Dex-Eto groups, compared with the model group. Among the Dex, Eto and Dex-Eto groups, the escape latency and swimming distance in the Dex-Eto group were the shortest, the number of times of crossing the platform and the target quadrant residence time were the highest, and IL-17A, S-100β, TNF-α, IL-6, IL-1β and NF-κB p65 expression levels were the lowest. In conclusion, dexmedetomidine combined with etomidate can effectively improve POCD.
Collapse
Affiliation(s)
- Xiaoli Yu
- Department of Anesthesiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yunfang Xie
- Department of Anesthesiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
99983
|
Sahay B, Mergia A. The Potential Contribution of Caveolin 1 to HIV Latent Infection. Pathogens 2020; 9:pathogens9110896. [PMID: 33121153 PMCID: PMC7692328 DOI: 10.3390/pathogens9110896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
Combinatorial antiretroviral therapy (cART) suppresses HIV replication to undetectable levels and has been effective in prolonging the lives of HIV infected individuals. However, cART is not capable of eradicating HIV from infected individuals mainly due to HIV’s persistence in small reservoirs of latently infected resting cells. Latent infection occurs when the HIV-1 provirus becomes transcriptionally inactive and several mechanisms that contribute to the silencing of HIV transcription have been described. Despite these advances, latent infection remains a major hurdle to cure HIV infected individuals. Therefore, there is a need for more understanding of novel mechanisms that are associated with latent infection to purge HIV from infected individuals thoroughly. Caveolin 1(Cav-1) is a multifaceted functional protein expressed in many cell types. The expression of Cav-1 in lymphocytes has been controversial. Recent evidence, however, convincingly established the expression of Cav-1 in lymphocytes. In lieu of this finding, the current review examines the potential role of Cav-1 in HIV latent infection and provides a perspective that helps uncover new insights to understand HIV latent infection.
Collapse
Affiliation(s)
| | - Ayalew Mergia
- Correspondence: ; Tel.: +352-294-4139; Fax: +352-392-9704
| |
Collapse
|
99984
|
Disrupted white matter integrity and network connectivity are related to poor motor performance. Sci Rep 2020; 10:18369. [PMID: 33110225 PMCID: PMC7591496 DOI: 10.1038/s41598-020-75617-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 10/15/2020] [Indexed: 11/24/2022] Open
Abstract
Motor impairment is common in the elderly population. Disrupted white matter tracts and the resultant loss of connectivity between cortical regions play an essential role in motor control. Using diffusion tensor imaging (DTI), we investigated the effect of white matter microstructure on upper-extremity and lower-extremity motor function in a community-based sample. A total of 766 participants (57.3 ± 9.2 years) completed the assessment of motor performance, including 3-m walking speed, 5-repeat chair-stand time, 10-repeat hand pronation-supination time, and 10-repeat finger-tapping time. Fractional anisotropy (FA), mean diffusivity (MD), and structural network connectivity parameters were calculated based on DTI. Lower FA and higher MD were associated with poor performance in walking, chair-stand, hand pronation-supination, and finger-tapping tests, independent of the presence of lacunes, white matter hyperintensities volume, and brain atrophy. Reduced network density, network strength, and global efficiency related to slower hand pronation-supination and finger-tapping, but not related to walking speed and chair-stand time. Disrupted white matter integrity and reduced cerebral network connectivity were associated with poor motor performance. Diffusion-based methods provide a more in-depth insight into the neural basis of motor dysfunction.
Collapse
|
99985
|
Karimi SA, Hosseinmardi N, Sayyah M, Hajisoltani R, Janahmadi M. Enhancement of intrinsic neuronal excitability-mediated by a reduction in hyperpolarization-activated cation current (I h ) in hippocampal CA1 neurons in a rat model of traumatic brain injury. Hippocampus 2020; 31:156-169. [PMID: 33107111 DOI: 10.1002/hipo.23270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 01/13/2023]
Abstract
Traumatic brain injury (TBI) is associated with epileptiform activity in the hippocampus; however, the underlying mechanisms have not been fully determined. The goal was to understand what changes take place in intrinsic neuronal physiology in the hippocampus after blunt force trauma to the cortex. In this context, hyperpolarization-activated cation current (Ih ) currents may have a critical role in modulating the neuronal intrinsic membrane excitability; therefore, its contribution to the TBI-induced hyperexcitability was assessed. In a model of TBI caused by controlled cortical impact (CCI), the intrinsic electrophysiological properties of pyramidal neurons were examined 1 week after TBI induction in rats. Whole-cell patch-clamp recordings were performed under current- and voltage-clamp conditions following ionotropic receptors blockade. Induction of TBI caused changes in the intrinsic excitability of pyramidal neurons, as shown by a significant increase and decrease in firing frequency and in the rheobase current, respectively (p < .05). The evoked firing rate and the action potential time to peak were also significantly increased and decreased, respectively (p < .05). In the TBI group, the amplitude of instantaneous and steady-state Ih currents was both significantly smaller than those in the control group (p < .05). The Ih current density was also significantly decreased (p < .001). Findings indicated that TBI led to an increase in the intrinsic excitability in CA1 pyramidal neurons and changes in Ih current could be, in part, one of the underlying mechanisms involved in this hyperexcitability.
Collapse
Affiliation(s)
- Seyed Asaad Karimi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Razieh Hajisoltani
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
99986
|
Cao Z, Luo Z, Huang X, Pinto JM, Simonsick EM, Shiroma EJ, Chen H. Self-Reported Versus Objectively Assessed Olfaction and Parkinson’s Disease Risk. JOURNAL OF PARKINSONS DISEASE 2020; 10:1789-1795. [DOI: 10.3233/jpd-202164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Poor olfaction is a prodromal symptom of Parkinson’s disease (PD); however, self-reported sense of smell is often dismissed as unreliable. Objective: To assess self-reported and objectively assessed sense of smell, independently and jointly, in relation to future risk for PD. Methods: We conducted a prospective analysis using data from 2,424 participants, ages 71–82 at baseline, from the Health, Aging, and Body Composition study. Exposures were self-reported poor sense of smell or taste and the objectively measured 12-item Brief Smell Identification Test score. The outcome was incident PD, analyzed using Cox proportional hazard models adjusted for age, sex, race, and cognitive function. Results: After approximately 10 years of follow-up, both self-reported and objectively tested poor sense of smell were independently associated with a higher risk of developing PD: the hazard ratios (95% confidence interval) were 2.8 (1.3, 5.9) and 4.0 (2.1, 7.5), respectively. When analyzed jointly, compared with participants who reported and tested normal, the hazard ratio was 2.2 (1.0, 4.6) for those reported poor sense of smell but tested normal, 3.6 (1.9, 6.9) for reported normal but tested poor, and 7.8 (3.2, 19.4) for both reported and tested poor. We did not find significant interactions between self-reported and objectively tested sense of smell in predicting PD risk. Conclusion: This study provides preliminary evidence that self-reported poor sense of smell or taste should not be simply dismissed as useless in predicting risk of PD. Future studies should confirm our finding and evaluate whether structured questionnaires may further improve the predictability.
Collapse
Affiliation(s)
- Zichun Cao
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Zhehui Luo
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jayant M. Pinto
- Section of Otolaryngology-Head and Neck Surgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Eleanor M. Simonsick
- Laboratory of Epidemiology and Population Science, Intramural Research Program of the National Institutes of Health, National Institute on Aging, Bethesda, MD, USA
| | - Eric J. Shiroma
- Laboratory of Epidemiology and Population Science, Intramural Research Program of the National Institutes of Health, National Institute on Aging, Bethesda, MD, USA
| | - Honglei Chen
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
99987
|
Shadrin AA, Mucha S, Ellinghaus D, Makarious MB, Blauwendraat C, Sreelatha AAK, Heras-Garvin A, Ding J, Hammer M, Foubert-Samier A, Meissner WG, Rascol O, Pavy-Le Traon A, Frei O, O'Connell KS, Bahrami S, Schreiber S, Lieb W, Müller-Nurasyid M, Schminke U, Homuth G, Schmidt CO, Nöthen MM, Hoffmann P, Gieger C, Wenning G, Gibbs JR, Franke A, Hardy J, Stefanova N, Gasser T, Singleton A, Houlden H, Scholz SW, Andreassen OA, Sharma M. Shared Genetics of Multiple System Atrophy and Inflammatory Bowel Disease. Mov Disord 2020; 36:449-459. [PMID: 33107653 PMCID: PMC8985479 DOI: 10.1002/mds.28338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/01/2020] [Accepted: 09/21/2020] [Indexed: 11/22/2022] Open
Abstract
Background: Multiple system atrophy (MSA) is a rare neurodegenerative disease characterized by intracellular accumulations of α-synuclein and nerve cell loss in striatonigral and olivopontocerebellar structures. Epidemiological and clinical studies have reported potential involvement of autoimmune mechanisms in MSA pathogenesis. However, genetic etiology of this interaction remains unknown. We aimed to investigate genetic overlap between MSA and 7 autoimmune diseases and to identify shared genetic loci. Methods: Genome-wide association study summary statistics of MSA and 7 autoimmune diseases were combined in cross-trait conjunctional false discovery rate analysis to explore overlapping genetic background. Expression of selected candidate genes was compared in transgenic MSA mice and wild-type mice. Genetic variability of candidate genes was further investigated using independent whole-exome genotyping data from large cohorts of MSA and autoimmune disease patients and healthy controls. Results: We observed substantial polygenic overlap between MSA and inflammatory bowel disease and identified 3 shared genetic loci with leading variants upstream of the DENND1B and RSP04 genes, and in intron of the C7 gene. Further, the C7 gene showed significantly dysregulated expression in the degenerating midbrain of transgenic MSA mice compared with wild-type mice and had elevated burden of protein-coding variants in independent MSA and inflammatory bowel disease cohorts. Conclusion: Our study provides evidence of shared genetic etiology between MSA and inflammatory bowel disease with an important role of the C7 gene in both phenotypes, with the implication of immune and gut dysfunction in MSA pathophysiology.
Collapse
Affiliation(s)
- Alexey A Shadrin
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sören Mucha
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Mary B Makarious
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and, Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Ashwin A K Sreelatha
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | | | - Jinhui Ding
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Monia Hammer
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexandra Foubert-Samier
- Service de Neurologie, CRMR Atrophie Multisystématisée, CHU Bordeaux, Bordeaux, France.,Inserm, UMR1219, Bordeaux Population Health Research Center, Bordeaux University, ISPED, Bordeaux, France
| | - Wassilios G Meissner
- Service de Neurologie, CRMR Atrophie Multisystématisée, CHU Bordeaux, Bordeaux, France.,Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, CNRS, Bordeaux, France
| | - Olivier Rascol
- Centre de Reference Maladie Rare Atrophie MultiSystématisée, Centre d'Investigation, Clinique CIC 1436, Services de Pharmacologie Clinique et Neurosciences, NeuroToul COEN Center, Toulouse, France.,Centre Hospitalo-Universitaire de Toulouse, 3, INSERM, Toulouse, France
| | - Anne Pavy-Le Traon
- Neurology Department, French Reference Centre for MSA, University Hospital of Toulouse and INSERM U 1048, Institute of Cardiovascular and Metabolic Diseases, Toulouse, France
| | - Oleksandr Frei
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Kevin S O'Connell
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Shahram Bahrami
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany.,First Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank PopGen, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Genetic Epidemiology, IBE, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.,Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Ulf Schminke
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Carsten O Schmidt
- Institute for Community Medicine, Study of Health in Pomerania/KEF, University Medicine Greifswald, Greifswald, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Gregor Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - J Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - John Hardy
- Rita Lila Weston Institute, University College London, London, UK
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Henry Houlden
- Rita Lila Weston Institute, University College London, London, UK
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and, Stroke, National Institutes of Health, Bethesda, Maryland, USA.,Department of Neurology, Johns Hopkins University Medical Center, Baltimore, Maryland, USA
| | - Ole A Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Manu Sharma
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
99988
|
Yılmaz Kara B, Kalcan S, Özyurt S, Gümüş A, Özçelik N, Karadoğan D, Şahin Ü. Weight Loss as the First-Line Therapy in Patients with Severe Obesity and Obstructive Sleep Apnea Syndrome: the Role of Laparoscopic Sleeve Gastrectomy. Obes Surg 2020; 31:1082-1091. [PMID: 33108591 DOI: 10.1007/s11695-020-05080-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE The objective of this study is to investigate the effects of laparoscopic sleeve gastrectomy (LSG) on the polysomnographic parameters related to OSAS. MATERIALS AND METHODS We conducted this 3-year prospective cohort study in a tertiary care center between December 2016 and December 2019. In total, we enrolled 31 patients with severe obesity who underwent full-night polysomnography (PSG) before LSG. Later, the patients were re-evaluated by full-night PSG 12 months after the surgery. RESULTS The mean age of the patients was 44.1 ± 9.6 years. The mean body mass index (BMI) decreased significantly from a mean value of 49.8 ± 8.5 kg/m2 at baseline to 33.2 ± 8.2 kg/m2 and a percent BMI (%BMI) reduction of 33.8 ± 10.4% and a percent total weight loss (%TWL) of 35.4 ± 10.8% was achieved on the same day of the postsurgical PSG (p < 0.001). There was a remarkable improvement in the AHI (baseline: 36.1 ± 27.1, 12 months after the surgery: 10.3 ± 11.8; difference: 25.8 ± 22.8 events per hour) (p < 0.001). Importantly, there was a decrease in the percentage of non-rapid eye movement (NREM) 2 (p < 0.001), whereas NREM 3 and REM stages witnessed a significant increase (p = 0.001 and p < 0.001, respectively) after the surgery. CONCLUSION The results of this study showed that weight loss after LSG yields improvement not only in AHI but also in many polysomnographic parameters such as sleep quality and desaturation indices.
Collapse
Affiliation(s)
- Bilge Yılmaz Kara
- Department of Pulmonology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey.
| | - Süleyman Kalcan
- Department of General Surgery, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Songül Özyurt
- Department of Pulmonology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Aziz Gümüş
- Department of Pulmonology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Neslihan Özçelik
- Department of Pulmonology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Dilek Karadoğan
- Department of Pulmonology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Ünal Şahin
- Department of Pulmonology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| |
Collapse
|
99989
|
Opfer R, Krüger J, Spies L, Hamann M, Wicki CA, Kitzler HH, Gocke C, Silva D, Schippling S. Age-dependent cut-offs for pathological deep gray matter and thalamic volume loss using Jacobian integration. NEUROIMAGE-CLINICAL 2020; 28:102478. [PMID: 33269702 PMCID: PMC7645288 DOI: 10.1016/j.nicl.2020.102478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 11/28/2022]
Abstract
Longitudinal MRI data of 189 healthy controls and 127 MS patients were analyzed. Deep gray matter (deep GMVL) thalamic volume loss (ThalaVL) was assessed. Magnitude of measurement error was estimated by means of 162 MRI scan-rescans. Age-dependent cut-offs for pathological deep GMVL and ThalaVL are provided. Result help interpreting deep GMVL and ThalaVL measurements in individual MS patients.
Introduction Several recent studies indicate that deep gray matter or thalamic volume loss (VL) might be promising surrogate markers of disease activity in multiple sclerosis (MS) patients. To allow applying these markers to individual MS patients in clinical routine, age-dependent cut-offs distinguishing physiological from pathological VL and an estimation of the measurement error, which provides the confidence of the result, are to be defined. Methods Longitudinal MRI scans of the following cohorts were analyzed in this study: 189 healthy controls (HC) (mean age 54 years, 22% female), 98 MS patients from Zurich university hospital (mean age 34 years, 62% female), 33 MS patients from Dresden university hospital (mean age 38 years, 60% female), and publicly available reliability data sets consisting of 162 short-term MRI scan-rescan pairs with scan intervals of days or few weeks. Percentage annualized whole brain volume loss (BVL), gray matter (GM) volume loss (GMVL), deep gray matter volume loss (deep GMVL), and thalamic volume loss (ThalaVL) were computed deploying the Jacobian integration (JI) method. BVL was additionally computed using Siena, an established method used in many Phase III drug trials. A linear mixed effect model was used to estimate the measurement error as the standard deviation (SD) of model residuals of all 162 scan-rescan pairs For estimation of age-dependent cut-offs, a quadratic regression function between age and the corresponding annualized VL values of the HC was computed. The 5th percentile was defined as the threshold for pathological VL per year since 95% of HC subjects exhibit a less pronounced VL for a given age. For the MS patients BVL, GMVL, deep GMVL, and ThalaVL were mutually compared and a paired t-test was used to test whether there are systematic differences in VL between these brain regions. Results Siena and JI showed a high agreement for BVL measures, with a median absolute difference of 0.1% and a correlation coefficient of r = 0.78. Siena and GMVL showed a similar standard deviation (SD) of the scan-rescan error of 0.28% and 0.29%, respectively. For deep GMVL, ThalaVL the SD of the scan-rescan error was slightly higher (0.43% and 0.5%, respectively). Among the HC the thalamus showed the highest mean VL (−0.16%, −0.39%, and −0.59% at ages 35, 55, and 75, respectively). Corresponding cut-offs for a pathological VL/year were −0.68%, −0.91%, and −1.11%. The MS cohorts did not differ in BVL and GMVL. However, both MS cohorts showed a significantly (p = 0.05) stronger deep GMVL than BVL per year. Conclusion It might be methodologically feasible to assess deep GMVL using JI in individual MS patients. However, age and the measurement error need to be taken into account. Furthermore, deep GMVL may be used as a complementary marker to BVL since MS patients exhibit a significantly stronger deep GMVL than BVL.
Collapse
Affiliation(s)
| | | | | | | | - Carla A Wicki
- Multimodal Imaging in Neuroimmunological Diseases (MINDS), University of Zurich, Zurich, Switzerland; Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Hagen H Kitzler
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Carola Gocke
- Conradia Medical Prevention Hamburg, Hamburg, Germany
| | - Diego Silva
- Bristol Myers Squibb, Princeton, NJ, United States
| | - Sven Schippling
- Multimodal Imaging in Neuroimmunological Diseases (MINDS), University of Zurich, Zurich, Switzerland; Center for Neuroscience Zurich (ZNZ), ETH Zurich, Zurich, Switzerland
| |
Collapse
|
99990
|
Friedman A, Hueske E, Drammis SM, Toro Arana SE, Nelson ED, Carter CW, Delcasso S, Rodriguez RX, Lutwak H, DiMarco KS, Zhang Q, Rakocevic LI, Hu D, Xiong JK, Zhao J, Gibb LG, Yoshida T, Siciliano CA, Diefenbach TJ, Ramakrishnan C, Deisseroth K, Graybiel AM. Striosomes Mediate Value-Based Learning Vulnerable in Age and a Huntington's Disease Model. Cell 2020; 183:918-934.e49. [PMID: 33113354 DOI: 10.1016/j.cell.2020.09.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/10/2020] [Accepted: 09/24/2020] [Indexed: 12/21/2022]
Abstract
Learning valence-based responses to favorable and unfavorable options requires judgments of the relative value of the options, a process necessary for species survival. We found, using engineered mice, that circuit connectivity and function of the striosome compartment of the striatum are critical for this type of learning. Calcium imaging during valence-based learning exhibited a selective correlation between learning and striosomal but not matrix signals. This striosomal activity encoded discrimination learning and was correlated with task engagement, which, in turn, could be regulated by chemogenetic excitation and inhibition. Striosomal function during discrimination learning was disturbed with aging and severely so in a mouse model of Huntington's disease. Anatomical and functional connectivity of parvalbumin-positive, putative fast-spiking interneurons (FSIs) to striatal projection neurons was enhanced in striosomes compared with matrix in mice that learned. Computational modeling of these findings suggests that FSIs can modulate the striosomal signal-to-noise ratio, crucial for discrimination and learning.
Collapse
Affiliation(s)
- Alexander Friedman
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emily Hueske
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sabrina M Drammis
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sebastian E Toro Arana
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Erik D Nelson
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cody W Carter
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sebastien Delcasso
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Raimundo X Rodriguez
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hope Lutwak
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kaden S DiMarco
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Qingyang Zhang
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lara I Rakocevic
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dan Hu
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua K Xiong
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jiajia Zhao
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leif G Gibb
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tomoko Yoshida
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cody A Siciliano
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
99991
|
Rafii MS, Ances BM, Schupf N, Krinsky‐McHale SJ, Mapstone M, Silverman W, Lott I, Klunk W, Head E, Christian B, Lai F, Rosas HD, Zaman S, Petersen ME, Strydom A, Fortea J, Handen B, O'Bryant S. The AT(N) framework for Alzheimer's disease in adults with Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12062. [PMID: 33134477 PMCID: PMC7588820 DOI: 10.1002/dad2.12062] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022]
Abstract
The National Institute on Aging in conjunction with the Alzheimer's Association (NIA-AA) recently proposed a biological framework for defining the Alzheimer's disease (AD) continuum. This new framework is based upon the key AD biomarkers (amyloid, tau, neurodegeneration, AT[N]) instead of clinical symptoms and represents the latest understanding that the pathological processes underlying AD begin decades before the manifestation of symptoms. By using these same biomarkers, individuals with Down syndrome (DS), who are genetically predisposed to developing AD, can also be placed more precisely along the AD continuum. The A/T(N) framework is therefore thought to provide an objective manner by which to select and enrich samples for clinical trials. This new framework is highly flexible and allows the addition of newly confirmed AD biomarkers into the existing AT(N) groups. As biomarkers for other pathological processes are validated, they can also be added to the AT(N) classification scheme, which will allow for better characterization and staging of AD in DS. These biological classifications can then be merged with clinical staging for an examination of factors that impact the biological and clinical progression of the disease. Here, we leverage previously published guidelines for the AT(N) framework to generate such a plan for AD among adults with DS.
Collapse
Affiliation(s)
- Michael S. Rafii
- Alzheimer's Therapeutic Research Institute (ATRI)Keck School of MedicineUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Beau M. Ances
- Center for Advanced Medicine NeuroscienceWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain/G.H. Sergievsky CenterColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of EpidemiologyMailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyNeurological Institute of New York, Columbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of PsychiatryColumbia University Medical CenterNew YorkNew YorkUSA
| | - Sharon J. Krinsky‐McHale
- Department of PsychologyNYS Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Mark Mapstone
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
| | - Wayne Silverman
- Department of PediatricsSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Ira Lott
- Department of PediatricsSchool of MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - William Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Elizabeth Head
- Department of PathologyGillespie Neuroscience Research Facility, University of CaliforniaIrvineCaliforniaUSA
| | - Brad Christian
- Department of Medical Physics and PsychiatryUniversity of Wisconsin MadisonMadisonWisconsinUSA
| | - Florence Lai
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMassachusettsUSA
| | - H. Diana Rosas
- Departments of Neurology and RadiologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMassachusettsUSA
| | - Shahid Zaman
- Department of PsychiatrySchool of Clinical MedicineUniversity of CambridgeCambridgeUK
- Cambridgeshire and Peterborough NHS Foundation TrustFulbourn HospitalCambridgeUK
| | - Melissa E. Petersen
- Department of Family Medicine and Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental SciencesInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Juan Fortea
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Benjamin Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Sid O'Bryant
- Institute for Translational Research and Department of Pharmacology and NeuroscienceUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
99992
|
Shore AN, Colombo S, Tobin WF, Petri S, Cullen ER, Dominguez S, Bostick CD, Beaumont MA, Williams D, Khodagholy D, Yang M, Lutz CM, Peng Y, Gelinas JN, Goldstein DB, Boland MJ, Frankel WN, Weston MC. Reduced GABAergic Neuron Excitability, Altered Synaptic Connectivity, and Seizures in a KCNT1 Gain-of-Function Mouse Model of Childhood Epilepsy. Cell Rep 2020; 33:108303. [PMID: 33113364 PMCID: PMC7712469 DOI: 10.1016/j.celrep.2020.108303] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 08/06/2020] [Accepted: 10/01/2020] [Indexed: 01/07/2023] Open
Abstract
Gain-of-function (GOF) variants in K+ channels cause severe childhood epilepsies, but there are no mechanisms to explain how increased K+ currents lead to network hyperexcitability. Here, we introduce a human Na+-activated K+ (KNa) channel variant (KCNT1-Y796H) into mice and, using a multiplatform approach, find motor cortex hyperexcitability and early-onset seizures, phenotypes strikingly similar to those of human patients. Although the variant increases KNa currents in cortical excitatory and inhibitory neurons, there is an increase in the KNa current across subthreshold voltages only in inhibitory neurons, particularly in those with non-fast-spiking properties, resulting in inhibitory-neuron-specific impairments in excitability and action potential (AP) generation. We further observe evidence of synaptic rewiring, including increases in homotypic synaptic connectivity, accompanied by network hyperexcitability and hypersynchronicity. These findings support inhibitory-neuron-specific mechanisms in mediating the epileptogenic effects of KCNT1 channel GOF, offering cell-type-specific currents and effects as promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Amy N Shore
- Department of Neurological Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Sophie Colombo
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | - William F Tobin
- Department of Neurological Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Sabrina Petri
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | - Erin R Cullen
- Department of Neurological Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Soledad Dominguez
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | | | - Michael A Beaumont
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA; Axion BioSystems, Atlanta, GA 30309, USA
| | - Damian Williams
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | - Dion Khodagholy
- Department of Electrical Engineering, Columbia University, New York, NY 10032, USA
| | - Mu Yang
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA
| | | | - Yueqing Peng
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Jennifer N Gelinas
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA; Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Michael J Boland
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA; Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Wayne N Frankel
- Institute for Genomic Medicine, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Matthew C Weston
- Department of Neurological Sciences, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|
99993
|
Alafuzoff I, Libard S. Mixed Brain Pathology Is the Most Common Cause of Cognitive Impairment in the Elderly. J Alzheimers Dis 2020; 78:453-465. [PMID: 33016922 DOI: 10.3233/jad-200925] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Systemic diseases, diabetes mellitus (DM), and cardiovascular disease (CaVD) have been suggested being risk factors for cognitive impairment (CI) and/or influence Alzheimer's disease neuropathologic change (ADNC). OBJECTIVE The purpose was to assess the type and the extent of neuropathological alterations in the brain and to assess whether brain pathology was associated with CaVD or DM related alterations in peripheral organs, i.e., vessels, heart, and kidney. METHODS 119 subjects, 15% with DM and 24% with CI, age range 80 to 89 years, were chosen and neuropathological alterations were assessed applying immunohistochemistry. RESULTS Hyperphosphorylated τ (HPτ) was seen in 99%, amyloid-β (Aβ) in 71%, transactive DNA binding protein 43 (TDP43) in 62%, and α-synuclein (αS) in 21% of the subjects. Primary age related tauopathy was diagnosed in 29% (more common in females), limbic predominant age-related TDP encephalopathy in 4% (14% of subjects with CI), and dementia with Lewy bodies in 3% (14% of subjects with CI) of the subjects. High/intermediate level of ADNC was seen in 47% and the extent of HPτ increased with age. The extent of ADNC was not associated with the extent of pathology observed in peripheral organs, i.e., DM or CaVD. Contrary, brain alterations such as pTDP43 and cerebrovascular lesions (CeVL) were influenced by DM, and CeVL correlated significantly with the extent of vessel pathology. CONCLUSION In most (66%) subjects with CI, the cause of impairment was "mixed pathology", i.e., ADNC combined with TDP43, αS, or vascular brain lesions. Furthermore, our results suggest that systemic diseases, DM and CaVD, are risk factors for CI but not related to ADNC.
Collapse
Affiliation(s)
- Irina Alafuzoff
- Department of Pathology, Uppsala University Hospital, Sweden
| | - Sylwia Libard
- Department of Pathology, Uppsala University Hospital, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| |
Collapse
|
99994
|
Kolbaev SN, Mohapatra N, Chen R, Lombardi A, Staiger JF, Luhmann HJ, Jedlicka P, Kilb W. NKCC-1 mediated Cl - uptake in immature CA3 pyramidal neurons is sufficient to compensate phasic GABAergic inputs. Sci Rep 2020; 10:18399. [PMID: 33110147 PMCID: PMC7591924 DOI: 10.1038/s41598-020-75382-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Activation of GABAA receptors causes in immature neurons a functionally relevant decrease in the intracellular Cl- concentration ([Cl-]i), a process termed ionic plasticity. Amount and duration of ionic plasticity depends on kinetic properties of [Cl-]i homeostasis. In order to characterize the capacity of Cl- accumulation and to quantify the effect of persistent GABAergic activity on [Cl-]i, we performed gramicidin-perforated patch-clamp recordings from CA3 pyramidal neurons of immature (postnatal day 4-7) rat hippocampal slices. These experiments revealed that inhibition of NKCC1 decreased [Cl-]i toward passive distribution with a time constant of 381 s. In contrast, active Cl- accumulation occurred with a time constant of 155 s, corresponding to a rate of 15.4 µM/s. Inhibition of phasic GABAergic activity had no significant effect on steady state [Cl-]i. Inhibition of tonic GABAergic currents induced a significant [Cl-]i increase by 1.6 mM, while activation of tonic extrasynaptic GABAA receptors with THIP significantly reduced [Cl-]i.. Simulations of neuronal [Cl-]i homeostasis supported the observation, that basal levels of synaptic GABAergic activation do not affect [Cl-]i. In summary, these results indicate that active Cl--uptake in immature hippocampal neurons is sufficient to maintain stable [Cl-]i at basal levels of phasic and to some extent also to compensate tonic GABAergic activity.
Collapse
Affiliation(s)
- Sergey N Kolbaev
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany.,Research Center of Neurology, Volokolamskoyeshosse, 80, Moscow, Russia, 125367
| | - Namrata Mohapatra
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Rongqing Chen
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany.,Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Aniello Lombardi
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany
| | - Jochen F Staiger
- Institute of Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.,ICAR3R-Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, Rudolf-Buchheim-Str. 6, 35392, Giessen, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany.
| |
Collapse
|
99995
|
Lui EH. Editorial for "Thalamic Nuclei Volumes and Their Relationships to Neuroperformance in Multiple Sclerosis: A Cross-Sectional Structural MRI Study". J Magn Reson Imaging 2020; 53:740-741. [PMID: 33103820 DOI: 10.1002/jmri.27413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 11/06/2022] Open
Affiliation(s)
- Elaine H Lui
- Department of Medicine and Radiology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Radiology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
99996
|
Zakaria Z, Abdullah MM, Halim SA, Ghani ARI, Idris Z, Abdullah JM. The Neurological Exam of a Comatose Patient: An Essential Practical Guide. Malays J Med Sci 2020; 27:108-123. [PMID: 33154707 PMCID: PMC7605838 DOI: 10.21315/mjms2020.27.5.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 09/10/2020] [Indexed: 11/28/2022] Open
Abstract
A thorough examination of a comatose patient is essential given the spectrum of clinical diagnoses. The most immediate threat to patients is airway, breathing and circulation. All attending physician should employ a structured and focused approach in dealing with a comatose patient. It is important to recognise the urgent steps needed at the time to prevent further deterioration, followed by the final diagnosis of patient’s neurologic status. Here we provide the essential practical guide to the neurological exam of a comatose patient that would assist to determine the aetiology, location and nature of the neurological lesion.
Collapse
Affiliation(s)
- Zaitun Zakaria
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Mohamad Muhaimin Abdullah
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Neurosurgery, Hospital Sultanah Bahiyah, Alor Setar, Kedah, Malaysia
| | - Sanihah Abdul Halim
- Department of Neurosciences, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Neurology Unit, Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Abdul Rahman Izaini Ghani
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Zamzuri Idris
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Jafri Malin Abdullah
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
99997
|
Intracranial Hemorrhage Detection in Head CT Using Double-Branch Convolutional Neural Network, Support Vector Machine, and Random Forest. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10217577] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Brain hemorrhage is a severe threat to human life, and its timely and correct diagnosis and treatment are of great importance. Multiple types of brain hemorrhage are distinguished depending on the location and character of bleeding. The main division covers five subtypes: subdural, epidural, intraventricular, intraparenchymal, and subarachnoid hemorrhage. This paper presents an approach to detect these intracranial hemorrhage types in computed tomography images of the head. The model trained for each hemorrhage subtype is based on a double-branch convolutional neural network of ResNet-50 architecture. It extracts features from two chromatic representations of the input data: a concatenation of the image normalized in different intensity windows and a stack of three consecutive slices creating a 3D spatial context. The joint feature vector is passed to the classifier to produce the final decision. We tested two tools: the support vector machine and the random forest. The experiments involved 372,556 images from 11,454 CT series of 9997 patients, with each image annotated with labels related to the hemorrhage subtypes. We validated deep networks from both branches of our framework and the model with either of two classifiers under consideration. The obtained results justify the use of a combination of double-source features with the random forest classifier. The system outperforms state-of-the-art methods in terms of F1 score. The highest detection accuracy was obtained in intraventricular (96.7%) and intraparenchymal hemorrhages (93.3%).
Collapse
|
99998
|
Yu Y, Wu Y, Cao X, Li J, Liao X, Wei J, Huang W. The Clinical Features and Prognosis of Anti-NMDAR Encephalitis Depends on Blood Brain Barrier Integrity. Mult Scler Relat Disord 2020; 47:102604. [PMID: 33130468 DOI: 10.1016/j.msard.2020.102604] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/04/2020] [Accepted: 10/25/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis is an autoimmune nervous system disease that has become increasingly recognized. This retrospective study is aimed to analyze the relations between clinical manifestations and blood brain barrier (BBB) integrity in anti-NMDAR encephalitis patients. METHODS Anti-NMDAR encephalitis patients were admitted to the First Affiliated Hospital of Guangxi Medical University from April 2014 to April 2020. Patients were grouped by the normal BBB and damaged BBB groups according to the cerebrospinal fluid (CSF) albumin/serum albumin (QAlb). Neutrophil-to-lymphocyte ratio (NLR) in peripheral blood was used for estimating the inflammatory status. The modified Rankin Scale (mRS) was used to assess prognosis. RESULTS Seventy-three anti-NMDAR encephalitis patients were diagnosed based on the autoimmune encephalitis diagnosis criteria of 2016. Fifty-three (72.6%) patients were in the normal BBB group and twenty (27.4%) were in the BBB damaged group. There were no significant differences in gender, age, psychiatric disturbances, epilepsy, speech disorder, motor dysfunction, memory dysfunction, and autonomic dysfunction between the two groups (p>0.05). Nevertheless, the proportions of decreased consciousness, ICU admission, NLR, CSF protein and intrathecal IgG synthesis (IgGIF, IgGLoc) in the damaged BBB group were higher than that in the normal BBB group (p<0.05). Patients (79.2%) with normal BBB had good prognosis compared to patients with damaged BBB (50%) after 2 months follow-up. The median mRS before and after immunotherapy in the damaged BBB group were significantly higher than that in the normal BBB group (p<0.01, p<0.05, respectively). Additionally, QAlb increased was positively correlated with the quantitative intrathecal IgG synthesis (IgGLoc: r=0.66; IgGIF: r=0.433, all p<0.001). CONCLUSION The dysfunction of BBB can be helpful in evaluating its prognosis since QAlb showed associations with ICU admission, NLR, a higher CSF protein, intrathecal IgG synthesis (IgGLoc, IgGIF) and mRS score after 2 months follow-up.
Collapse
Affiliation(s)
- Yachun Yu
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, P. R. China, 530021
| | - Yu Wu
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, P. R. China, 530021
| | - Xiaoli Cao
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, P. R. China, 530021
| | - Jing Li
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, P. R. China, 530021
| | - Xiangling Liao
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, P. R. China, 530021
| | - Junxiang Wei
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, P. R. China, 530021
| | - Wen Huang
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, Nanning, P. R. China, 530021.
| |
Collapse
|
99999
|
Erb ML, Moore DJ. LRRK2 and the Endolysosomal System in Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2020; 10:1271-1291. [PMID: 33044192 PMCID: PMC7677880 DOI: 10.3233/jpd-202138] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal dominant familial Parkinson’s disease (PD), with pathogenic mutations enhancing LRRK2 kinase activity. There is a growing body of evidence indicating that LRRK2 contributes to neuronal damage and pathology both in familial and sporadic PD, making it of particular interest for understanding the molecular pathways that underlie PD. Although LRRK2 has been extensively studied to date, our understanding of the seemingly diverse functions of LRRK2 throughout the cell remains incomplete. In this review, we discuss the functions of LRRK2 within the endolysosomal pathway. Endocytosis, vesicle trafficking pathways, and lysosomal degradation are commonly disrupted in many neurodegenerative diseases, including PD. Additionally, many PD-linked gene products function in these intersecting pathways, suggesting an important role for the endolysosomal system in maintaining protein homeostasis and neuronal health in PD. LRRK2 activity can regulate synaptic vesicle endocytosis, lysosomal function, Golgi network maintenance and sorting, vesicular trafficking and autophagy, with alterations in LRRK2 kinase activity serving to disrupt or regulate these pathways depending on the distinct cell type or model system. LRRK2 is critically regulated by at least two proteins in the endolysosomal pathway, Rab29 and VPS35, which may serve as master regulators of LRRK2 kinase activity. Investigating the function and regulation of LRRK2 in the endolysosomal pathway in diverse PD models, especially in vivo models, will provide critical insight into the cellular and molecular pathophysiological mechanisms driving PD and whether LRRK2 represents a viable drug target for disease-modification in familial and sporadic PD.
Collapse
Affiliation(s)
- Madalynn L Erb
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Darren J Moore
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
100000
|
Zhang RQ, Chen SD, Shen XN, Yang YX, Lu JY, Cui M, Zuo CT, Dong Q, Tan L, Yu JT. Elevated Tau PET Signal Depends on Abnormal Amyloid Levels and Correlates with Cognitive Impairment in Elderly Persons without Dementia. J Alzheimers Dis 2020; 78:395-404. [PMID: 32986669 DOI: 10.3233/jad-200526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The recent developed PET ligands for amyloid-β (Aβ) and tau allow these two neuropathological hallmarks of Alzheimer's disease (AD) to be mapped and quantified in vivo and to be examined in relation to cognition. OBJECTIVE To assess the associations among Aβ, tau, and cognition in non-demented subjects. METHODS Three hundred eighty-nine elderly participants without dementia from the Alzheimer's Disease Neuroimaging Initiative underwent tau and amyloid PET scans. Cross-sectional comparisons and longitudinal analyses were used to evaluate the relationship between Aβ and tau accumulation. The correlations between biomarkers of both pathologies and performance in memory and executive function were measured. RESULTS Increased amyloid-PET retention was associated with greater tau-PET retention in widespread cortices. We observed a significant tau increase in the temporal composite regions of interest over 24 months in Aβ+ but not Aβ- subjects. Finally, tau-PET retention but not amyloid-PET retention significantly explained the variance in memory and executive function. Higher level of tau was associated with greater longitudinal memory decline. CONCLUSION These findings suggested PET-detectable Aβ plaque pathology may be a necessary antecedent for tau-PET signal elevation. Greater tau-PET retention may demonstrate poorer cognition and predict prospective memory decline in non-demented subjects.
Collapse
Affiliation(s)
- Rui-Qi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Xiang Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Ying Lu
- Department of PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chuan-Tao Zuo
- Department of PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | | |
Collapse
|