201
|
Riancho J, Gonzalo I, Ruiz-Soto M, Berciano J. Why do motor neurons degenerate? Actualisation in the pathogenesis of amyotrophic lateral sclerosis. NEUROLOGÍA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.nrleng.2015.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
202
|
Riancho J, Gonzalo I, Ruiz-Soto M, Berciano J. ¿Por qué degeneran las motoneuronas? Actualización en la patogenia de la esclerosis lateral amiotrófica. Neurologia 2019; 34:27-37. [DOI: 10.1016/j.nrl.2015.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/06/2015] [Indexed: 12/11/2022] Open
|
203
|
Pickett SB, Thomas ED, Sebe JY, Linbo T, Esterberg R, Hailey DW, Raible DW. Cumulative mitochondrial activity correlates with ototoxin susceptibility in zebrafish mechanosensory hair cells. eLife 2018; 7:38062. [PMID: 30596476 PMCID: PMC6345563 DOI: 10.7554/elife.38062] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 12/31/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a prominent role in mechanosensory hair cell damage and death. Although hair cells are thought to be energetically demanding cells, how mitochondria respond to these demands and how this might relate to cell death is largely unexplored. Using genetically encoded indicators, we found that mitochondrial calcium flux and oxidation are regulated by mechanotransduction and demonstrate that hair cell activity has both acute and long-term consequences on mitochondrial function. We tested whether variation in mitochondrial activity reflected differences in the vulnerability of hair cells to the toxic drug neomycin. We observed that susceptibility did not correspond to the acute level of mitochondrial activity but rather to the cumulative history of that activity.
Collapse
Affiliation(s)
- Sarah B Pickett
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Eric D Thomas
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Joy Y Sebe
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Tor Linbo
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Robert Esterberg
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - Dale W Hailey
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| |
Collapse
|
204
|
When safeguarding goes wrong: Impact of oxidative stress on protein homeostasis in health and neurodegenerative disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 114:221-264. [PMID: 30635082 DOI: 10.1016/bs.apcsb.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cellular redox status is an established player in many different cellular functions. The buildup of oxidants within the cell is tightly regulated to maintain a balance between the positive and negative outcomes of cellular oxidants. Proteins are highly sensitive to oxidation, since modification can cause widespread unfolding and the formation of toxic aggregates. In response, cells have developed highly regulated systems that contribute to the maintenance of both the global redox status and protein homeostasis at large. Changes to these systems have been found to correlate with aging and age-related disorders, such as neurodegenerative pathologies. This raises intriguing questions as to the source of the imbalance in the redox and protein homeostasis systems, their interconnectivity, and their role in disease progression. Here we focus on the crosstalk between the redox and protein homeostasis systems in neurodegenerative diseases, specifically in Alzheimer's, Parkinson's, and ALS. We elaborate on some of the main players of the stress response systems, including the master regulators of oxidative stress and the heat shock response, Nrf2 and Hsf1, which are essential features of protein folding, and mediators of protein turnover. We illustrate the elegant mechanisms used by these components to provide an immediate response, including protein plasticity controlled by redox-sensing cysteines and the recruitment of naive proteins to the redox homeostasis array that act as chaperons in an ATP-independent manner.
Collapse
|
205
|
Fu H, Possenti A, Freer R, Nakano Y, Hernandez Villegas NC, Tang M, Cauhy PVM, Lassus BA, Chen S, Fowler SL, Figueroa HY, Huey ED, Johnson GVW, Vendruscolo M, Duff KE. A tau homeostasis signature is linked with the cellular and regional vulnerability of excitatory neurons to tau pathology. Nat Neurosci 2018; 22:47-56. [PMID: 30559469 PMCID: PMC6330709 DOI: 10.1038/s41593-018-0298-7] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/23/2018] [Indexed: 01/04/2023]
Abstract
Excitatory neurons are preferentially impaired in early Alzheimer's disease but the pathways contributing to their relative vulnerability remain largely unknown. Here we report that pathological tau accumulation takes place predominantly in excitatory neurons compared to inhibitory neurons, not only in the entorhinal cortex, a brain region affected in early Alzheimer's disease, but also in areas affected later by the disease. By analyzing RNA transcripts from single-nucleus RNA datasets, we identified a specific tau homeostasis signature of genes differentially expressed in excitatory compared to inhibitory neurons. One of the genes, BCL2-associated athanogene 3 (BAG3), a facilitator of autophagy, was identified as a hub, or master regulator, gene. We verified that reducing BAG3 levels in primary neurons exacerbated pathological tau accumulation, whereas BAG3 overexpression attenuated it. These results define a tau homeostasis signature that underlies the cellular and regional vulnerability of excitatory neurons to tau pathology.
Collapse
Affiliation(s)
- Hongjun Fu
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA.
| | - Andrea Possenti
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Rosie Freer
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Yoshikazu Nakano
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | | | - Maoping Tang
- Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Paula V M Cauhy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA.,Federal University of Uberlândia, Uberlândia, Brazil
| | - Benjamin A Lassus
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Shuo Chen
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Stephanie L Fowler
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Helen Y Figueroa
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA
| | - Edward D Huey
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA.,Departments of Psychiatry and Neurology, Columbia University, New York, NY, USA
| | - Gail V W Johnson
- Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Karen E Duff
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
206
|
Yang S, Li S, Li XJ. Shortening the Half-Life of Cas9 Maintains Its Gene Editing Ability and Reduces Neuronal Toxicity. Cell Rep 2018; 25:2653-2659.e3. [PMID: 30517854 PMCID: PMC6314484 DOI: 10.1016/j.celrep.2018.11.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 09/20/2018] [Accepted: 11/01/2018] [Indexed: 12/22/2022] Open
Abstract
Virus-mediated expression of CRISPR/Cas9 is commonly used for genome editing in animal brains to model or treat neurological diseases, but the potential neurotoxicity of overexpressing bacterial Cas9 in the mammalian brain remains unknown. Through RNA sequencing (RNA-seq) analysis, we find that virus-mediated expression of Cas9 influences the expression of genes involved in neuronal functions. Reducing the half-life of Cas9 by tagging with geminin, whose expression is regulated by the cell cycle, maintains the genome editing capacity of Cas9 but significantly alleviates neurotoxicity. Thus, modification of Cas9 by shortening its half-life can help develop CRISPR/Cas9-based therapeutic approaches for treating neurological disorders.
Collapse
Affiliation(s)
- Su Yang
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA.
| | - Shihua Li
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA
| | - Xiao-Jiang Li
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Room 355, Atlanta, GA 30322, USA.
| |
Collapse
|
207
|
An H, Williams NG, Shelkovnikova TA. NEAT1 and paraspeckles in neurodegenerative diseases: A missing lnc found? Noncoding RNA Res 2018; 3:243-252. [PMID: 30533572 PMCID: PMC6257911 DOI: 10.1016/j.ncrna.2018.11.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are among the most common causes of disability worldwide. Although neurodegenerative diseases are heterogeneous in both their clinical features and the underlying physiology, they are all characterised by progressive loss of specific neuronal populations. Recent experimental evidence suggests that long non-coding RNAs (lncRNAs) play important roles in the CNS in health and disease. Nuclear Paraspeckle Assembly Transcript 1 (NEAT1) is an abundant, ubiquitously expressed lncRNA, which forms a scaffold for a specific RNA granule in the nucleus, or nuclear body, the paraspeckle. Paraspeckles act as molecular hubs for cellular processes commonly affected by neurodegeneration. Transcriptomic analyses of the diseased human tissue have revealed altered NEAT1 levels in the CNS in major neurodegenerative disorders as well as in some disease models. Although it is clear that changes in NEAT1 expression (and in some cases, paraspeckle assembly) accompany neuronal damage, our understanding of NEAT1 contribution to the disease pathogenesis is still rudimentary. In this review, we have summarised the available knowledge on NEAT1 involvement in the molecular processes linked to neurodegeneration and on NEAT1 dysregulation in this type of disease, with a special focus on amyotrophic lateral sclerosis. The goal of this review is to attract the attention of researchers in the field of neurodegeneration to NEAT1 and paraspeckles.
Collapse
Affiliation(s)
- Haiyan An
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Park Place, Cardiff, CF10 3AT, United Kingdom
| | - Non G Williams
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Park Place, Cardiff, CF10 3AT, United Kingdom
| | - Tatyana A Shelkovnikova
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Park Place, Cardiff, CF10 3AT, United Kingdom
| |
Collapse
|
208
|
Díaz J, Martínez-Martín P, Rodríguez-Blázquez C, Vázquez B, Forjaz MJ, Ortiz C, Carmona R, Linares C. Short-term association between road traffic noise and healthcare demand generated by Parkinson's disease in Madrid, Spain. GACETA SANITARIA 2018; 32:553-558. [DOI: 10.1016/j.gaceta.2017.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 01/11/2023]
|
209
|
Noyce A, Bandopadhyay R. Parkinson's Disease: Basic Pathomechanisms and a Clinical Overview. ADVANCES IN NEUROBIOLOGY 2018; 15:55-92. [PMID: 28674978 DOI: 10.1007/978-3-319-57193-5_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PD is a common and a debilitating degenerative movement disorder. The number of patients is increasing worldwide and as yet there is no cure for the disease. The majority of existing treatments target motor symptom control. Over the last two decades the impact of the genetic contribution to PD has been appreciated. Significant discoveries have been made, which have advanced our understanding of the pathophysiological and molecular basis of PD. In this chapter we outline current knowledge of the clinical aspects of PD and the basic mechanistic understanding.
Collapse
Affiliation(s)
- Alastair Noyce
- Department of Molecular Neuroscience, Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1, Wakefield Street, London, WC1N 1PJ, UK
| | - Rina Bandopadhyay
- Department of Molecular Neuroscience, Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, 1, Wakefield Street, London, WC1N 1PJ, UK.
| |
Collapse
|
210
|
Di Gregorio SE, Duennwald ML. ALS Yeast Models-Past Success Stories and New Opportunities. Front Mol Neurosci 2018; 11:394. [PMID: 30425620 PMCID: PMC6218427 DOI: 10.3389/fnmol.2018.00394] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
In the past two decades, yeast models have delivered profound insights into basic mechanisms of protein misfolding and the dysfunction of key cellular pathways associated with amyotrophic lateral sclerosis (ALS). Expressing ALS-associated proteins, such as superoxide dismutase (SOD1), TAR DNA binding protein 43 (TDP-43) and Fused in sarcoma (FUS), in yeast recapitulates major hallmarks of ALS pathology, including protein aggregation, mislocalization and cellular toxicity. Results from yeast have consistently been recapitulated in other model systems and even specimens from human patients, thus providing evidence for the power and validity of ALS yeast models. Focusing on impaired ribonucleic acid (RNA) metabolism and protein misfolding and their cytotoxic consequences in ALS, we summarize exemplary discoveries that originated from work in yeast. We also propose previously unexplored experimental strategies to modernize ALS yeast models, which will help to decipher the basic pathomechanisms underlying ALS and thus, possibly contribute to finding a cure.
Collapse
Affiliation(s)
- Sonja E Di Gregorio
- Schulich School of Medicine and Dentistry, Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Martin L Duennwald
- Schulich School of Medicine and Dentistry, Pathology and Laboratory Medicine, Western University, London, ON, Canada
| |
Collapse
|
211
|
Caldwell KA, Thies JL, Caldwell GA. No Country for Old Worms: A Systematic Review of the Application of C. elegans to Investigate a Bacterial Source of Environmental Neurotoxicity in Parkinson's Disease. Metabolites 2018; 8:metabo8040070. [PMID: 30380609 PMCID: PMC6315381 DOI: 10.3390/metabo8040070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 10/21/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
While progress has been made in discerning genetic associations with Parkinson's disease (PD), identifying elusive environmental contributors necessitates the application of unconventional hypotheses and experimental strategies. Here, we provide an overview of studies that we conducted on a neurotoxic metabolite produced by a species of common soil bacteria, Streptomyces venezuelae (S. ven), indicating that the toxicity displayed by this bacterium causes stress in diverse cellular mechanisms, such as the ubiquitin proteasome system and mitochondrial homeostasis. This dysfunction eventually leads to age and dose-dependent neurodegeneration in the nematode Caenorhabditis elegans. Notably, dopaminergic neurons have heightened susceptibility, but all of the neuronal classes eventually degenerate following exposure. Toxicity further extends to human SH-SY5Y cells, which also degenerate following exposure. Additionally, the neurons of nematodes expressing heterologous aggregation-prone proteins display enhanced metabolite vulnerability. These mechanistic analyses collectively reveal a unique metabolomic fingerprint for this bacterially-derived neurotoxin. In considering that epidemiological distinctions in locales influence the incidence of PD, we surveyed soils from diverse regions of Alabama, and found that exposure to ~30% of isolated Streptomyces species caused worm dopaminergic neurons to die. In addition to aging, one of the few established contributors to PD appears to be a rural lifestyle, where exposure to soil on a regular basis might increase the risk of interaction with bacteria producing such toxins. Taken together, these data suggest that a novel toxicant within the Streptomyces genus might represent an environmental contributor to the progressive neurodegeneration that is associated with PD.
Collapse
Affiliation(s)
- Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487, USA.
- Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA.
| | - Jennifer L Thies
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487, USA.
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Box 870344, Tuscaloosa, AL 35487, USA.
- Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA.
| |
Collapse
|
212
|
Yokoi T, Watanabe H, Yamaguchi H, Bagarinao E, Masuda M, Imai K, Ogura A, Ohdake R, Kawabata K, Hara K, Riku Y, Ishigaki S, Katsuno M, Miyao S, Kato K, Naganawa S, Harada R, Okamura N, Yanai K, Yoshida M, Sobue G. Involvement of the Precuneus/Posterior Cingulate Cortex Is Significant for the Development of Alzheimer's Disease: A PET (THK5351, PiB) and Resting fMRI Study. Front Aging Neurosci 2018; 10:304. [PMID: 30344488 PMCID: PMC6182068 DOI: 10.3389/fnagi.2018.00304] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/13/2018] [Indexed: 01/02/2023] Open
Abstract
Background: Imaging studies in Alzheimer’s disease (AD) have yet to answer the underlying questions concerning the relationship among tau retention, neuroinflammation, network disruption and cognitive decline. We compared the spatial retention patterns of 18F-THK5351 and resting state network (RSN) disruption in patients with early AD and healthy controls. Methods: We enrolled 23 11C-Pittsburgh compound B (PiB)-positive patients with early AD and 24 11C-PiB-negative participants as healthy controls. All participants underwent resting state functional MRI and 18F-THK5351 PET scans. We used scaled subprofile modeling/principal component analysis (SSM/PCA) to reduce the complexity of multivariate data and to identify patterns that exhibited the largest statistical effects (variances) in THK5351 concentration in AD and healthy controls. Findings: SSM/PCA identified a significant spatial THK5351 pattern composed by mainly three clusters including precuneus/posterior cingulate cortex (PCC), right and left dorsolateral prefrontal cortex (DLPFC) which accounted for 23.6% of the total subject voxel variance of the data and had 82.6% sensitivity and 79.1% specificity in discriminating AD from healthy controls. There was a significant relationship between the intensity of the 18F-THK5351 covariation pattern and cognitive scores in AD. The spatial patterns of 18F-THK5351 uptake showed significant similarity with intrinsic functional connectivity, especially in the PCC network. Seed-based connectivity analysis from the PCC showed significant decrease in connectivity over widespread brain regions in AD patients. An evaluation of an autopsied AD patient with Braak V showed that 18F-THK5351 retention corresponded to tau deposition, monoamine oxidase-B (MAO-B) and astrogliosis in the precuneus/PCC. Interpretation: We identified an AD-specific spatial pattern of 18F-THK5351 retention in the precuneus/PCC, an important connectivity hub region in the brain. Disruption of the functional connections of this important network hub may play an important role in developing dementia in AD.
Collapse
Affiliation(s)
- Takamasa Yokoi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | | | | | - Michihito Masuda
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazunori Imai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Aya Ogura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Reiko Ohdake
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Kazuya Kawabata
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhiro Hara
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Riku
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Miyao
- Department of Neurology, Meitetsu Hospital, Nagoya, Japan
| | - Katsuhiko Kato
- Department of Radiological and Medical Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryuichi Harada
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| |
Collapse
|
213
|
Non-cell-autonomous actions of α-synuclein: Implications in glial synucleinopathies. Prog Neurobiol 2018; 169:158-171. [DOI: 10.1016/j.pneurobio.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 07/24/2017] [Accepted: 06/30/2018] [Indexed: 01/11/2023]
|
214
|
Soto C, Pritzkow S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat Neurosci 2018; 21:1332-1340. [PMID: 30250260 DOI: 10.1038/s41593-018-0235-9] [Citation(s) in RCA: 648] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022]
Abstract
A hallmark event in neurodegenerative diseases (NDs) is the misfolding, aggregation, and accumulation of proteins, leading to cellular dysfunction, loss of synaptic connections, and brain damage. Despite the involvement of distinct proteins in different NDs, the process of protein misfolding and aggregation is remarkably similar. A recent breakthrough in the field was the discovery that misfolded protein aggregates can self-propagate through seeding and spread the pathological abnormalities between cells and tissues in a manner akin to the behavior of infectious prions in prion diseases. This discovery has vast implications for understanding the mechanisms involved in the initiation and progression of NDs, as well as for the design of novel strategies for treatment and diagnosis. In this Review, we provide a critical discussion of the role of protein misfolding and aggregation in NDs. Commonalities and differences between distinct protein aggregates will be highlighted, in addition to evidence supporting the hypothesis that misfolded aggregates can be transmissible by the prion principle. We will also describe the molecular basis and implications for prion-like conformational strains, cross-interaction between different misfolded proteins in the brain, and how these concepts can be applied to the development of novel strategies for therapy and diagnosis.
Collapse
Affiliation(s)
- Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas, USA.
| | - Sandra Pritzkow
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, University of Texas McGovern Medical School, Houston, Texas, USA
| |
Collapse
|
215
|
Fu H, Hardy J, Duff KE. Selective vulnerability in neurodegenerative diseases. Nat Neurosci 2018; 21:1350-1358. [PMID: 30250262 DOI: 10.1038/s41593-018-0221-2] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases have two general characteristics that are so fundamental we usually take them for granted. The first is that the pathology associated with the disease only affects particular neurons ('selective neuronal vulnerability'); the second is that the pathology worsens with time and impacts more regions in a stereotypical and predictable fashion. The mechanisms underpinning selective neuronal and regional vulnerability have been difficult to dissect, but the recent application of whole-genome technologies, the development of mouse models that reproduce spatial and temporal features of the pathology, and the identification of intrinsic morphological, electrophysiological, and biochemical properties of vulnerable neurons are beginning to shed some light on these fundamental features of neurodegenerative diseases. Here we detail our emerging understanding of the underlying biology of selective neuronal vulnerability and outline some of the areas in which our understanding is incomplete.
Collapse
Affiliation(s)
- Hongjun Fu
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain; and Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.,Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - John Hardy
- Department of Molecular Neuroscience and Reta Lilla Weston Laboratories, Institute of Neurology, London, UK
| | - Karen E Duff
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain; and Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Department of Psychiatry, Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
216
|
Chung CG, Lee H, Lee SB. Mechanisms of protein toxicity in neurodegenerative diseases. Cell Mol Life Sci 2018; 75:3159-3180. [PMID: 29947927 PMCID: PMC6063327 DOI: 10.1007/s00018-018-2854-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022]
Abstract
Protein toxicity can be defined as all the pathological changes that ensue from accumulation, mis-localization, and/or multimerization of disease-specific proteins. Most neurodegenerative diseases manifest protein toxicity as one of their key pathogenic mechanisms, the details of which remain unclear. By systematically deconstructing the nature of toxic proteins, we aim to elucidate and illuminate some of the key mechanisms of protein toxicity from which therapeutic insights may be drawn. In this review, we focus specifically on protein toxicity from the point of view of various cellular compartments such as the nucleus and the mitochondria. We also discuss the cell-to-cell propagation of toxic disease proteins that complicates the mechanistic understanding of the disease progression as well as the spatiotemporal point at which to therapeutically intervene. Finally, we discuss selective neuronal vulnerability, which still remains largely enigmatic.
Collapse
Affiliation(s)
- Chang Geon Chung
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, Republic of Korea
| | - Hyosang Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, Republic of Korea.
| | - Sung Bae Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, Republic of Korea.
| |
Collapse
|
217
|
Enabling Precision Medicine through Integrative Network Models. J Mol Biol 2018; 430:2913-2923. [DOI: 10.1016/j.jmb.2018.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/15/2018] [Accepted: 07/03/2018] [Indexed: 11/17/2022]
|
218
|
Aymerich MS, Aso E, Abellanas MA, Tolon RM, Ramos JA, Ferrer I, Romero J, Fernández-Ruiz J. Cannabinoid pharmacology/therapeutics in chronic degenerative disorders affecting the central nervous system. Biochem Pharmacol 2018; 157:67-84. [PMID: 30121249 DOI: 10.1016/j.bcp.2018.08.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
The endocannabinoid system (ECS) exerts a modulatory effect of important functions such as neurotransmission, glial activation, oxidative stress, or protein homeostasis. Dysregulation of these cellular processes is a common neuropathological hallmark in aging and in neurodegenerative diseases of the central nervous system (CNS). The broad spectrum of actions of cannabinoids allows targeting different aspects of these multifactorial diseases. In this review, we examine the therapeutic potential of the ECS for the treatment of chronic neurodegenerative diseases of the CNS focusing on Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. First, we describe the localization of the molecular components of the ECS and how they are altered under neurodegenerative conditions, either contributing to or protecting cells from degeneration. Second, we address recent advances in the modulation of the ECS using experimental models through different strategies including the direct targeting of cannabinoid receptors with agonists or antagonists, increasing the endocannabinoid tone by the inhibition of endocannabinoid hydrolysis, and activation of cannabinoid receptor-independent effects. Preclinical evidence indicates that cannabinoid pharmacology is complex but supports the therapeutic potential of targeting the ECS. Third, we review the clinical evidence and discuss the future perspectives on how to bridge human and animal studies to develop cannabinoid-based therapies for each neurodegenerative disorder. Finally, we summarize the most relevant opportunities of cannabinoid pharmacology related to each disease and the multiple unexplored pathways in cannabinoid pharmacology that could be useful for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria S Aymerich
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; Universidad de Navarra, CIMA, Programa de Neurociencias, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Spain.
| | - Ester Aso
- Departamento de Patología y Terapéutica Experimental, Universidad de Barcelona, L'Hospitalet de Llobregat, Spain; CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain
| | - Miguel A Abellanas
- Universidad de Navarra, Facultad de Ciencias, Departamento de Bioquímica y Genética, Pamplona, Spain; Universidad de Navarra, CIMA, Programa de Neurociencias, Pamplona, Spain
| | - Rosa M Tolon
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
| | - Jose A Ramos
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Isidre Ferrer
- Departamento de Patología y Terapéutica Experimental, Universidad de Barcelona, L'Hospitalet de Llobregat, Spain; CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain
| | - Julian Romero
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
| | - Javier Fernández-Ruiz
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| |
Collapse
|
219
|
Rosas-Arellano A, Estrada-Mondragón A, Piña R, Mantellero CA, Castro MA. The Tiny Drosophila Melanogaster for the Biggest Answers in Huntington's Disease. Int J Mol Sci 2018; 19:E2398. [PMID: 30110961 PMCID: PMC6121572 DOI: 10.3390/ijms19082398] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 12/18/2022] Open
Abstract
The average life expectancy for humans has increased over the last years. However, the quality of the later stages of life is low and is considered a public health issue of global importance. Late adulthood and the transition into the later stage of life occasionally leads to neurodegenerative diseases that selectively affect different types of neurons and brain regions, producing motor dysfunctions, cognitive impairment, and psychiatric disorders that are progressive, irreversible, without remission periods, and incurable. Huntington's disease (HD) is a common neurodegenerative disorder. In the 25 years since the mutation of the huntingtin (HTT) gene was identified as the molecule responsible for this neural disorder, a variety of animal models, including the fruit fly, have been used to study the disease. Here, we review recent research that used Drosophila as an experimental tool for improving knowledge about the molecular and cellular mechanisms underpinning HD.
Collapse
Affiliation(s)
- Abraham Rosas-Arellano
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Argel Estrada-Mondragón
- Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden.
| | - Ricardo Piña
- Laboratorio de Neurociencias, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9160000, Chile.
- Departamento de Ciencias Químicas y Biológicas, Universidad Bernardo O'Higgins, Santiago 8370993, Chile.
| | - Carola A Mantellero
- Facultad de Ciencias de la Salud, Universidad de Las Américas, Santiago 7500972, Chile.
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile.
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile.
| |
Collapse
|
220
|
Raj A, Powell F. Models of Network Spread and Network Degeneration in Brain Disorders. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 3:788-797. [PMID: 30170711 DOI: 10.1016/j.bpsc.2018.07.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 01/01/2023]
Abstract
Network analysis can provide insight into key organizational principles of brain structure and help identify structural changes associated with brain disease. Though static differences between diseased and healthy networks are well characterized, the study of network dynamics, or how brain networks change over time, is increasingly central to understanding ongoing brain changes throughout disease. Accordingly, we present a short review of network models of spread, network dynamics, and network degeneration. Borrowing from recent suggestions, we divide this review into two processes by which brain networks can change: dynamics on networks, which are functional and pathological consequences taking place atop a static structural brain network; and dynamics of networks, which constitutes a changing structural brain network. We focus on diffusion magnetic resonance imaging-based structural or anatomic connectivity graphs. We address psychiatric disorders like schizophrenia; developmental disorders like epilepsy; stroke; and Alzheimer's disease and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.
| | - Fon Powell
- Department of Radiology, Weill Cornell Medicine, New York, New York
| |
Collapse
|
221
|
Hess JL, Akutagava-Martins GC, Patak JD, Glatt SJ, Faraone SV. Why is there selective subcortical vulnerability in ADHD? Clues from postmortem brain gene expression data. Mol Psychiatry 2018; 23:1787-1793. [PMID: 29180674 PMCID: PMC6985986 DOI: 10.1038/mp.2017.242] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/17/2017] [Accepted: 08/24/2017] [Indexed: 01/25/2023]
Abstract
Sub-cortical volumetric differences were associated with attention-deficit/hyperactivity disorder (ADHD) in a recent multi-site, mega-analysis of 1713 ADHD persons and 1529 controls. As there was a wide range of effect sizes among the sub-cortical volumes, it is possible that selective neuronal vulnerability has a role in these volumetric losses. To address this possibility, we used data from Allen Brain Atlas to investigate variability in gene expression profiles between subcortical regions of typically developing brains. We tested the hypothesis that the expression of genes in a set of curated ADHD candidate genes and five a priori selected, biological pathways would be associated with the Enhancing NeuroImaging Genetics through Meta-Analysis (ENIGMA) findings. Across the subcortical regions studied by ENIGMA, gene expression profiles for three pathways were significantly correlated with ADHD-associated volumetric reductions: apoptosis, oxidative stress and autophagy. These correlations were strong and significant for children with ADHD, but not for adults. Although preliminary, these data suggest that variability of structural brain anomalies in ADHD can be explained, in part, by the differential vulnerability of these regions to mechanisms mediating apoptosis, oxidative stress and autophagy.
Collapse
Affiliation(s)
- J L Hess
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - G C Akutagava-Martins
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - J D Patak
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - S J Glatt
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - S V Faraone
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA.
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
- K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway.
| |
Collapse
|
222
|
Gomes C, Cunha C, Nascimento F, Ribeiro JA, Vaz AR, Brites D. Cortical Neurotoxic Astrocytes with Early ALS Pathology and miR-146a Deficit Replicate Gliosis Markers of Symptomatic SOD1G93A Mouse Model. Mol Neurobiol 2018; 56:2137-2158. [DOI: 10.1007/s12035-018-1220-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/29/2018] [Indexed: 12/13/2022]
|
223
|
Chang HY, Sang TK, Chiang AS. Untangling the Tauopathy for Alzheimer's disease and parkinsonism. J Biomed Sci 2018; 25:54. [PMID: 29991349 PMCID: PMC6038292 DOI: 10.1186/s12929-018-0457-x] [Citation(s) in RCA: 32] [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: 03/06/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022] Open
Abstract
Tau is a microtubule-associated protein that mainly localizes to the axon to stabilize axonal microtubule structure and neuronal connectivity. Tau pathology is one of the most common proteinopathies that associates with age-dependent neurodegenerative diseases including Alzheimer's disease (AD), and various Parkinsonism. Tau protein undergoes a plethora of intra-molecular modifications and some altered forms promote the production of toxic oligomeric tau and paired helical filaments, and through which further assemble into neurofibrillary tangles, also known as tauopathy. In this review, we will discuss the recent advances of the tauopathy research, primarily focusing on its association with the early axonal manifestation of axonal transport defect, axonal mitochondrial stress, autophagic vesicle accumulation and the proceeding of axon destruction, and the pathogenic Tau spreading across the synapse. Two alternative strategies either by targeting tau protein itself or by improving the age-related physiological decline are currently racing to find the hopeful treatment for tauopathy. Undoubtedly, more studies are needed to combat this devastating condition that has already affected millions of people in our aging population.
Collapse
Affiliation(s)
- Hui-Yun Chang
- Department of Medical Science, Institute of Systems Neuroscience, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Brain Research Center, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Tzu-Kang Sang
- Department of Life Science, Institute of Biotechnology, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Brain Research Center, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Ann-Shyn Chiang
- Department of Medical Science, Institute of Systems Neuroscience, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Department of Life Science, Institute of Biotechnology, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
- Brain Research Center, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| |
Collapse
|
224
|
Srinivasan G, Morgan D, Varun D, Brookhouser N, Brafman DA. An integrated biomanufacturing platform for the large-scale expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells. Acta Biomater 2018; 74:168-179. [PMID: 29775730 DOI: 10.1016/j.actbio.2018.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cell derived neural progenitor cells (hNPCs) have the unique properties of long-term in vitro expansion as well as differentiation into the various neurons and supporting cell types of the central nervous system (CNS). Because of these characteristics, hNPCs have tremendous potential in the modeling and treatment of various CNS diseases and disorders. However, expansion and neuronal differentiation of hNPCs in quantities necessary for these applications is not possible with current two dimensional (2-D) approaches. Here, we used a fully defined peptide substrate as the basis for a microcarrier (MC)-based suspension culture system. Several independently derived hNPC lines were cultured on MCs for multiple passages as well as efficiently differentiated to neurons. Finally, this MC-based system was used in conjunction with a low shear rotating wall vessel (RWV) bioreactor for the integrated, large-scale expansion and neuronal differentiation of hNPCs. Overall, this fully defined and scalable biomanufacturing system will facilitate the generation of hNPCs and their neuronal derivatives in quantities necessary for basic and translational applications. STATEMENT OF SIGNIFICANCE In this work, we developed a microcarrier (MC)-based culture system that allows for the expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells (hNPCs) under defined conditions. In turn, this MC approach was implemented in a rotating wall vessel (RWV) bioreactor for the large-scale expansion and neuronal differentiation of hNPCs. This work is of significance as it overcomes current limitations of conventional two dimensional (2-D) culture systems to enable the generation of hNPCs and their neuronal derivatives in quantities required for downstream applications in disease modeling, drug screening, and regenerative medicine.
Collapse
Affiliation(s)
- Gayathri Srinivasan
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Daylin Morgan
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Divya Varun
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Nicholas Brookhouser
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, United States.
| |
Collapse
|
225
|
Driven to decay: Excitability and synaptic abnormalities in amyotrophic lateral sclerosis. Brain Res Bull 2018; 140:318-333. [PMID: 29870780 DOI: 10.1016/j.brainresbull.2018.05.023] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/26/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron (MN) disease and is clinically characterised by the death of corticospinal motor neurons (CSMNs), spinal and brainstem MNs and the degeneration of the corticospinal tract. Degeneration of CSMNs and MNs leads inexorably to muscle wastage and weakness, progressing to eventual death within 3-5 years of diagnosis. The CSMNs, located within layer V of the primary motor cortex, project axons constituting the corticospinal tract, forming synaptic connections with brainstem and spinal cord interneurons and MNs. Clinical ALS may be divided into familial (∼10% of cases) or sporadic (∼90% of cases), based on apparent random incidence. The emergence of transgenic murine models, expressing different ALS-associated mutations has accelerated our understanding of ALS pathogenesis, although precise mechanisms remain elusive. Multiple avenues of investigation suggest that cortical electrical abnormalities have pre-eminence in the pathophysiology of ALS. In addition, glutamate-mediated functional and structural alterations in both CSMNs and MNs are present in both sporadic and familial forms of ALS. This review aims to promulgate debate in the field with regard to the common aetiology of sporadic and familial ALS. A specific focus on a nexus point in ALS pathogenesis, namely, the synaptic and intrinsic hyperexcitability of CSMNs and MNs and alterations to their structure are comprehensively detailed. The association of extramotor dysfunction with neuronal structural/functional alterations will be discussed. Finally, the implications of the latest research on the dying-forward and dying-back controversy are considered.
Collapse
|
226
|
Baggio HC, Abos A, Segura B, Campabadal A, Garcia‐Diaz A, Uribe C, Compta Y, Marti MJ, Valldeoriola F, Junque C. Statistical inference in brain graphs using threshold-free network-based statistics. Hum Brain Mapp 2018; 39:2289-2302. [PMID: 29450940 PMCID: PMC6619254 DOI: 10.1002/hbm.24007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/30/2018] [Accepted: 02/06/2018] [Indexed: 01/06/2023] Open
Abstract
The description of brain networks as graphs where nodes represent different brain regions and edges represent a measure of connectivity between a pair of nodes is an increasingly used approach in neuroimaging research. The development of powerful methods for edge-wise group-level statistical inference in brain graphs while controlling for multiple-testing associated false-positive rates, however, remains a difficult task. In this study, we use simulated data to assess the properties of threshold-free network-based statistics (TFNBS). The TFNBS combines threshold-free cluster enhancement, a method commonly used in voxel-wise statistical inference, and network-based statistic (NBS), which is frequently used for statistical analysis of brain graphs. Unlike the NBS, TFNBS generates edge-wise significance values and does not require the a priori definition of a hard cluster-defining threshold. Other test parameters, nonetheless, need to be set. We show that it is possible to find parameters that make TFNBS sensitive to strong and topologically clustered effects, while appropriately controlling false-positive rates. Our results show that the TFNBS is an adequate technique for the statistical assessment of brain graphs.
Collapse
Affiliation(s)
- Hugo C. Baggio
- Medical Psychology Unit, Department of Medicine, Institute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
| | - Alexandra Abos
- Medical Psychology Unit, Department of Medicine, Institute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
| | - Barbara Segura
- Medical Psychology Unit, Department of Medicine, Institute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
| | - Anna Campabadal
- Medical Psychology Unit, Department of Medicine, Institute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
| | - Anna Garcia‐Diaz
- Medical Psychology Unit, Department of Medicine, Institute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
| | - Carme Uribe
- Medical Psychology Unit, Department of Medicine, Institute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
| | - Yaroslau Compta
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de BarcelonaBarcelonaCataloniaSpain
- Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona. Institute of Neuroscience, University of BarcelonaBarcelonaCataloniaSpain
| | - Maria Jose Marti
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de BarcelonaBarcelonaCataloniaSpain
- Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona. Institute of Neuroscience, University of BarcelonaBarcelonaCataloniaSpain
| | - Francesc Valldeoriola
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de BarcelonaBarcelonaCataloniaSpain
- Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona. Institute of Neuroscience, University of BarcelonaBarcelonaCataloniaSpain
| | - Carme Junque
- Medical Psychology Unit, Department of Medicine, Institute of NeuroscienceUniversity of BarcelonaBarcelonaCataloniaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic de BarcelonaBarcelonaCataloniaSpain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS)BarcelonaCataloniaSpain
| |
Collapse
|
227
|
Acosta D, Powell F, Zhao Y, Raj A. Regional vulnerability in Alzheimer's disease: The role of cell-autonomous and transneuronal processes. Alzheimers Dement 2018; 14:797-810. [PMID: 29306583 PMCID: PMC5994366 DOI: 10.1016/j.jalz.2017.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 11/08/2017] [Accepted: 11/30/2017] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The stereotypical progression of Alzheimer's disease (AD) pathology is not fully understood. The selective impact of AD on distinct regions has led the field to question if innate vulnerability exists. This study aims to determine if the causative factors of regional vulnerability are dependent on cell-autonomous or transneuronal (non-cell autonomous) processes. METHODS Using mathematical and statistical models, we analyzed the contribution of cell-autonomous and non-cell autonomous factors to predictive linear models of AD pathology. RESULTS Results indicate gene expression as a weak contributor to predictive linear models of AD. Instead, the network diffusion model acts as a strong predictor of observed AD atrophy and hypometabolism. DISCUSSION We propose a convenient methodology for identifying genes and their role in determining AD topography, in comparison with network spread. Results reinforce the role of transneuronal network spread on disease progression and suggest that innate gene expression plays a secondary role in seeding and subsequent disease progression.
Collapse
Affiliation(s)
- Diana Acosta
- Department of Neuroscience, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Fontasha Powell
- Department of Neuroscience, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Yize Zhao
- Department of Healthcare Policy and Research, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Ashish Raj
- Department of Neuroscience, Weill Cornell Medical College of Cornell University, New York, NY, USA; Department of Radiology and Biomedical Imaging, UCSF School of Medicine, San Francisco, CA, USA.
| |
Collapse
|
228
|
Herrera MI, Udovin LD, Toro-Urrego N, Kusnier CF, Luaces JP, Otero-Losada M, Capani F. Neuroprotection Targeting Protein Misfolding on Chronic Cerebral Hypoperfusion in the Context of Metabolic Syndrome. Front Neurosci 2018; 12:339. [PMID: 29904335 PMCID: PMC5990610 DOI: 10.3389/fnins.2018.00339] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 04/30/2018] [Indexed: 01/04/2023] Open
Abstract
Metabolic syndrome (MetS) is a cluster of risk factors that lead to microvascular dysfunction and chronic cerebral hypoperfusion (CCH). Long-standing reduction in oxygen and energy supply leads to brain hypoxia and protein misfolding, thereby linking CCH to Alzheimer's disease. Protein misfolding results in neurodegeneration as revealed by studying different experimental models of CCH. Regulating proteostasis network through pathways like the unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), chaperone-mediated autophagy (CMA), and macroautophagy emerges as a novel target for neuroprotection. Lipoxin A4 methyl ester, baclofen, URB597, N-stearoyl-L-tyrosine, and melatonin may pose potential neuroprotective agents for rebalancing the proteostasis network under CCH. Autophagy is one of the most studied pathways of proteostatic cell response against the decrease in blood supply to the brain though the role of the UPR-specific chaperones and the UPS system in CCH deserves further research. Pharmacotherapy targeting misfolded proteins at different stages in the proteostatic pathway might be promising in treating cognitive impairment following CCH.
Collapse
Affiliation(s)
- María I Herrera
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina.,Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Lucas D Udovin
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Nicolás Toro-Urrego
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Carlos F Kusnier
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Juan P Luaces
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina
| | - Francisco Capani
- Instituto de Investigaciones Cardiológicas (ININCA), Universidad de Buenos Aires (UBA-CONICET), Buenos Aires, Argentina.,Facultad de Medicina, Universidad Católica Argentina, Buenos Aires, Argentina.,Universidad Autónoma de Chile, Santiago de Chile, Chile
| |
Collapse
|
229
|
Bingol B. Autophagy and lysosomal pathways in nervous system disorders. Mol Cell Neurosci 2018; 91:167-208. [PMID: 29729319 DOI: 10.1016/j.mcn.2018.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an evolutionarily conserved pathway for delivering cytoplasmic cargo to lysosomes for degradation. In its classically studied form, autophagy is a stress response induced by starvation to recycle building blocks for essential cellular processes. In addition, autophagy maintains basal cellular homeostasis by degrading endogenous substrates such as cytoplasmic proteins, protein aggregates, damaged organelles, as well as exogenous substrates such as bacteria and viruses. Given their important role in homeostasis, autophagy and lysosomal machinery are genetically linked to multiple human disorders such as chronic inflammatory diseases, cardiomyopathies, cancer, and neurodegenerative diseases. Multiple targets within the autophagy and lysosomal pathways offer therapeutic opportunities to benefit patients with these disorders. Here, I will summarize the mechanisms of autophagy pathways, the evidence supporting a pathogenic role for disturbed autophagy and lysosomal degradation in nervous system disorders, and the therapeutic potential of autophagy modulators in the clinic.
Collapse
Affiliation(s)
- Baris Bingol
- Genentech, Inc., Department of Neuroscience, 1 DNA Way, South San Francisco 94080, United States.
| |
Collapse
|
230
|
Luo W, Tweedie D, Beedie SL, Vargesson N, Figg WD, Greig NH, Scerba MT. Design, synthesis and biological assessment of N-adamantyl, substituted adamantyl and noradamantyl phthalimidines for nitrite, TNF-α and angiogenesis inhibitory activities. Bioorg Med Chem 2018; 26:1547-1559. [PMID: 29472124 PMCID: PMC5891396 DOI: 10.1016/j.bmc.2018.01.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/22/2018] [Accepted: 01/31/2018] [Indexed: 02/07/2023]
Abstract
A library of 15 novel and heretofore uncharacterized adamantyl and noradamantyl phthalimidines was synthesized and evaluated for neuroprotective and anti-angiogenic properties. Phthalimidine treatment in LPS-challenged cells effected reductions in levels of secreted TNF-α and nitrite relative to basal amounts. The primary SAR suggests nitration of adamantyl phthalimidines has marginal effect on TNF-α activity but promotes anti-nitrite activity; thioamide congeners retain anti-nitrite activity but are less effective reducing TNF-α. Site-specific nitration and thioamidation provided phthalimidine 24, effecting an 88.5% drop in nitrite concurrent with only a 4% drop in TNF-α. Notable anti-angiogenesis activity was observed for 20, 21 and 22.
Collapse
Affiliation(s)
- Weiming Luo
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Shaunna L Beedie
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; Molecular Pharmacology Section, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - William D Figg
- Molecular Pharmacology Section, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Michael T Scerba
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
231
|
Cauda F, Nani A, Costa T, Palermo S, Tatu K, Manuello J, Duca S, Fox PT, Keller R. The morphometric co-atrophy networking of schizophrenia, autistic and obsessive spectrum disorders. Hum Brain Mapp 2018; 39:1898-1928. [PMID: 29349864 PMCID: PMC5895505 DOI: 10.1002/hbm.23952] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022] Open
Abstract
By means of a novel methodology that can statistically derive patterns of co-alterations distribution from voxel-based morphological data, this study analyzes the patterns of brain alterations of three important psychiatric spectra-that is, schizophrenia spectrum disorder (SCZD), autistic spectrum disorder (ASD), and obsessive-compulsive spectrum disorder (OCSD). Our analysis provides five important results. First, in SCZD, ASD, and OCSD brain alterations do not distribute randomly but, rather, follow network-like patterns of co-alteration. Second, the clusters of co-altered areas form a net of alterations that can be defined as morphometric co-alteration network or co-atrophy network (in the case of gray matter decreases). Third, within this network certain cerebral areas can be identified as pathoconnectivity hubs, the alteration of which is supposed to enhance the development of neuronal abnormalities. Fourth, within the morphometric co-atrophy network of SCZD, ASD, and OCSD, a subnetwork composed of eleven highly connected nodes can be distinguished. This subnetwork encompasses the anterior insulae, inferior frontal areas, left superior temporal areas, left parahippocampal regions, left thalamus and right precentral gyri. Fifth, the co-altered areas also exhibit a normal structural covariance pattern which overlaps, for some of these areas (like the insulae), the co-alteration pattern. These findings reveal that, similarly to neurodegenerative diseases, psychiatric disorders are characterized by anatomical alterations that distribute according to connectivity constraints so as to form identifiable morphometric co-atrophy patterns.
Collapse
Affiliation(s)
- Franco Cauda
- GCS‐FMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- Focus Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Andrea Nani
- GCS‐FMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- Focus Lab, Department of PsychologyUniversity of TurinTurinItaly
- Michael Trimble Neuropsychiatry Research Group, University of Birmingham and BSMHFTBirminghamUK
| | - Tommaso Costa
- GCS‐FMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- Focus Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Sara Palermo
- Department of NeuroscienceUniversity of TurinTurinItaly
| | - Karina Tatu
- GCS‐FMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- Focus Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Jordi Manuello
- GCS‐FMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
- Focus Lab, Department of PsychologyUniversity of TurinTurinItaly
| | - Sergio Duca
- GCS‐FMRI, Koelliker Hospital and Department of PsychologyUniversity of TurinTurinItaly
| | - Peter T. Fox
- Research Imaging Institute, University of Texas Health Science Center At San AntonioSan AntonioTexas
- South Texas Veterans Health Care SystemSan AntonioTexas
| | - Roberto Keller
- Adult Autism Center, DSM Local Health Unit ASL Citta’ Di TorinoTurinItaly
| |
Collapse
|
232
|
Pennuto M, Rinaldi C. From gene to therapy in spinal and bulbar muscular atrophy: Are we there yet? Mol Cell Endocrinol 2018; 465:113-121. [PMID: 28688959 DOI: 10.1016/j.mce.2017.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 01/12/2023]
Abstract
Abnormal polyglutamine expansions in the androgen receptor (AR) cause a muscular condition, known as Kennedy's disease or spinal and bulbar muscular atrophy (SBMA). The disease is transmitted in an X-linked fashion and is clinically characterized by weakness, atrophy and fasciculations of the limb and bulbar muscles as a result of a toxic gain-of-function of the mutant protein. Notably, affected males also show signs of androgen insensitivity, such as gynaecomastia and reduced fertility. The characterization of the natural history of the disease, the increasing understanding of the mechanism of pathogenesis and the elucidation of the functions of normal and mutant AR have offered a momentum for developing a rational therapeutic strategy for this disease. In this special issue on androgens and AR functions, we will review the molecular, biochemical, and cellular mechanisms underlying the pathogenesis of SBMA. We will discuss recent advances on therapeutic approaches and opportunities for this yet incurable disease, ranging from androgen deprivation, to gene silencing, to an expanding repertoire of peripheral targets, including muscle. With the advancement of these strategies into the clinic, it can be reasonably anticipated that the landscape of treatment options for SBMA and other neuromuscular conditions will change rapidly in the near future.
Collapse
Affiliation(s)
- Maria Pennuto
- Dulbecco Telethon Institute, Centre for Integrative Biology, University of Trento, 38123 Trento, Italy; Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy.
| | - Carlo Rinaldi
- Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX Oxford, UK.
| |
Collapse
|
233
|
Kelley KW, Ben Haim L, Schirmer L, Tyzack GE, Tolman M, Miller JG, Tsai HH, Chang SM, Molofsky AV, Yang Y, Patani R, Lakatos A, Ullian EM, Rowitch DH. Kir4.1-Dependent Astrocyte-Fast Motor Neuron Interactions Are Required for Peak Strength. Neuron 2018; 98:306-319.e7. [PMID: 29606582 PMCID: PMC5919779 DOI: 10.1016/j.neuron.2018.03.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 11/08/2017] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
Diversified neurons are essential for sensorimotor function, but whether astrocytes become specialized to optimize circuit performance remains unclear. Large fast α-motor neurons (FαMNs) of spinal cord innervate fast-twitch muscles that generate peak strength. We report that ventral horn astrocytes express the inward-rectifying K+ channel Kir4.1 (a.k.a. Kcnj10) around MNs in a VGLUT1-dependent manner. Loss of astrocyte-encoded Kir4.1 selectively altered FαMN size and function and led to reduced peak strength. Overexpression of Kir4.1 in astrocytes was sufficient to increase MN size through activation of the PI3K/mTOR/pS6 pathway. Kir4.1 was downregulated cell autonomously in astrocytes derived from amyotrophic lateral sclerosis (ALS) patients with SOD1 mutation. However, astrocyte Kir4.1 was dispensable for FαMN survival even in the mutant SOD1 background. These findings show that astrocyte Kir4.1 is essential for maintenance of peak strength and suggest that Kir4.1 downregulation might uncouple symptoms of muscle weakness from MN cell death in diseases like ALS. Kir4.1 is upregulated in astrocytes around high-activity alpha motor neurons (MNs) Astrocyte Kir4.1 KO caused decreased peak strength without alpha MN loss ALS patient-derived astrocytes show cell-autonomous Kir4.1 downregulation Astrocyte Kir4.1 regulates MN size through PI3K/mTOR/pS6 activation
Collapse
Affiliation(s)
- Kevin W Kelley
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lucile Ben Haim
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lucas Schirmer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Giulia E Tyzack
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Michaela Tolman
- Sackler School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - John G Miller
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hui-Hsin Tsai
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sandra M Chang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anna V Molofsky
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yongjie Yang
- Sackler School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Rickie Patani
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Andras Lakatos
- John van Geest Centre for Brain Repair and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB20QQ, UK
| | - Erik M Ullian
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David H Rowitch
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Pediatrics and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Paediatrics and Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB20QQ, UK.
| |
Collapse
|
234
|
Gupta S, Yadav K, Mantri SS, Singhal NK, Ganesh S, Sandhir R. Evidence for Compromised Insulin Signaling and Neuronal Vulnerability in Experimental Model of Sporadic Alzheimer’s Disease. Mol Neurobiol 2018; 55:8916-8935. [DOI: 10.1007/s12035-018-0985-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/28/2018] [Indexed: 02/07/2023]
|
235
|
Azpurua J, Mahoney RE, Eaton BA. Transcriptomics of aged Drosophila motor neurons reveals a matrix metalloproteinase that impairs motor function. Aging Cell 2018; 17:e12729. [PMID: 29411505 PMCID: PMC5847883 DOI: 10.1111/acel.12729] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2017] [Indexed: 12/12/2022] Open
Abstract
The neuromuscular junction (NMJ) is responsible for transforming nervous system signals into motor behavior and locomotion. In the fruit fly Drosophila melanogaster, an age-dependent decline in motor function occurs, analogous to the decline experienced in mice, humans, and other mammals. The molecular and cellular underpinnings of this decline are still poorly understood. By specifically profiling the transcriptome of Drosophila motor neurons across age using custom microarrays, we found that the expression of the matrix metalloproteinase 1 (dMMP1) gene reproducibly increased in motor neurons in an age-dependent manner. Modulation of physiological aging also altered the rate of dMMP1 expression, validating dMMP1 expression as a bona fide aging biomarker for motor neurons. Temporally controlled overexpression of dMMP1 specifically in motor neurons was sufficient to induce deficits in climbing behavior and cause a decrease in neurotransmitter release at neuromuscular synapses. These deficits were reversible if the dMMP1 expression was shut off again immediately after the onset of motor dysfunction. Additionally, repression of dMMP1 enzymatic activity via overexpression of a tissue inhibitor of metalloproteinases delayed the onset of age-dependent motor dysfunction. MMPs are required for proper tissue architecture during development. Our results support the idea that matrix metalloproteinase 1 is acting as a downstream effector of antagonistic pleiotropy in motor neurons and is necessary for proper development, but deleterious when reactivated at an advanced age.
Collapse
Affiliation(s)
- Jorge Azpurua
- Department of AnesthesiologyStony Brook University School of MedicineStony BrookNYUSA
| | - Rebekah E. Mahoney
- Department of Cellular and Integrative PhysiologyUTHSCSASan AntonioTXUSA
- Barshop Institute for Longevity and Aging StudiesUTHSCSASan AntonioTXUSA
| | - Benjamin A. Eaton
- Department of Cellular and Integrative PhysiologyUTHSCSASan AntonioTXUSA
- Barshop Institute for Longevity and Aging StudiesUTHSCSASan AntonioTXUSA
| |
Collapse
|
236
|
Pérez M, Medina M, Hernández F, Avila J. Secretion of full-length Tau or Tau fragments in cell culture models. Propagation of Tau in vivo and in vitro. Biomol Concepts 2018; 9:1-11. [DOI: 10.1515/bmc-2018-0001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/25/2018] [Indexed: 01/18/2023] Open
Abstract
AbstractThe microtubule-associated protein Tau plays a crucial role in stabilizing neuronal microtubules. In Tauopathies, Tau loses its ability to bind microtubules, detach from them and forms intracellular aggregates. Increasing evidence in recent years supports the notion that Tau pathology spreading throughout the brain in AD and other Tauopathies is the consequence of the propagation of specific Tau species along neuroanatomically connected brain regions in a so-called “prion-like” manner. A number of steps are assumed to be involved in this process, including secretion, cellular uptake, transcellular transfer and/or seeding, although the precise mechanisms underlying propagation of Tau pathology are not fully understood yet. This review summarizes recent evidence on the nature of the specific Tau species that are propagated and the different mechanisms of Tau pathology spreading.
Collapse
Affiliation(s)
- Mar Pérez
- Departamento de Anatomía Histología y Neurociencia, Facultad de Medicina UAM, 28029Madrid, Spain
| | - Miguel Medina
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031Madrid, Spain
- CIEN Foundation, Carlos III Institute of Health, 28031Madrid, Spain
| | - Félix Hernández
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CSICUAM), 28049Madrid, Spain
| | - Jesús Avila
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CSICUAM), 28049Madrid, Spain
| |
Collapse
|
237
|
Astrocytic JWA deletion exacerbates dopaminergic neurodegeneration by decreasing glutamate transporters in mice. Cell Death Dis 2018; 9:352. [PMID: 29500411 PMCID: PMC5834463 DOI: 10.1038/s41419-018-0381-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/01/2018] [Accepted: 02/05/2018] [Indexed: 12/29/2022]
Abstract
Astrocytic JWA exerts neuroprotective roles by alleviating oxidative stress and inhibiting inflammation. However, the molecular mechanisms of how astrocytic JWA is involved in dopaminergic neurodegeneration in Parkinson's disease (PD) remain largely unknown. In this study, we found that astrocyte-specific JWA knockout mice (JWA CKO) exacerbated dopamine (DA) neuronal loss and motor dysfunction, and reduced the levels of DA and its metabolites in a 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine/probenecid (MPTP/p)-induced PD model. Astrocytic JWA deficiency repressed expression of excitatory amino-acid transporter 2 (GLT-1) and glutamate uptake both in vivo and in vitro. Further, the regulation of GLT-1 expression was involved in JWA-triggered activation of the MAPK and PI3K signaling pathways. JWA-increased GLT-1 expression was abolished by inhibitors of MEK and PI3K. Silencing CREB also abrogated JWA-increased GLT-1 expression and glutamate uptake. Additionally, JWA deficiency activated glial fibrillary acidic protein (GFAP), and increased the expression of STAT3. Similarly to the MPTP model, paraquat (PQ) exposure produced PD-like phenotypes in JWA CKO mice. Taken together, our findings provide novel insights into astrocytic JWA function in the pathogenesis of neurotoxin mouse models of PD.
Collapse
|
238
|
Chen H, Kankel MW, Su SC, Han SWS, Ofengeim D. Exploring the genetics and non-cell autonomous mechanisms underlying ALS/FTLD. Cell Death Differ 2018; 25:648-662. [PMID: 29459769 DOI: 10.1038/s41418-018-0060-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022] Open
Abstract
Although amyotrophic lateral sclerosis (ALS), also known as Lou Gehrig's disease, was first described in 1874, a flurry of genetic discoveries in the last 10 years has markedly increased our understanding of this disease. These findings have not only enhanced our knowledge of mechanisms leading to ALS, but also have revealed that ALS shares many genetic causes with another neurodegenerative disease, frontotemporal lobar dementia (FTLD). In this review, we survey how recent genetic studies have bridged our mechanistic understanding of these two related diseases and how the genetics behind ALS and FTLD point to complex disorders, implicating non-neuronal cell types in disease pathophysiology. The involvement of non-neuronal cell types is consistent with a non-cell autonomous component in these diseases. This is further supported by studies that identified a critical role of immune-associated genes within ALS/FTLD and other neurodegenerative disorders. The molecular functions of these genes support an emerging concept that various non-autonomous functions are involved in neurodegeneration. Further insights into such a mechanism(s) will ultimately lead to a better understanding of potential routes of therapeutic intervention. Facts ALS and FTLD are severe neurodegenerative disorders on the same disease spectrum. Multiple cellular processes including dysregulation of RNA homeostasis, imbalance of proteostasis, contribute to ALS/FTLD pathogenesis. Aberrant function in non-neuronal cell types, including microglia, contributes to ALS/FTLD. Strong neuroimmune and neuroinflammatory components are associated with ALS/FTLD patients. Open Questions Why can patients with similar mutations have different disease manifestations, i.e., why do C9ORF72 mutations lead to motor neuron loss in some patients while others exhibit loss of neurons in the frontotemporal lobe? Do ALS causal mutations result in microglial dysfunction and contribute to ALS/FTLD pathology? How do microglia normally act to mitigate neurodegeneration in ALS/FTLD? To what extent do cellular signaling pathways mediate non-cell autonomous communications between distinct central nervous system (CNS) cell types during disease? Is it possible to therapeutically target specific cell types in the CNS?
Collapse
Affiliation(s)
- Hongbo Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA, 02115, USA
| | - Mark W Kankel
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Susan C Su
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Steve W S Han
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,GSK, Upper Providence, PA, 19426, USA
| | - Dimitry Ofengeim
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA. .,Sanofi Neuroscience, Framingham, MA, USA.
| |
Collapse
|
239
|
Abstract
The decline of cognitive function occurs with aging, but the mechanisms responsible are unknown. Astrocytes instruct the formation, maturation, and elimination of synapses, and impairment of these functions has been implicated in many diseases. These findings raise the question of whether astrocyte dysfunction could contribute to cognitive decline in aging. We used the Bac-Trap method to perform RNA sequencing of astrocytes from different brain regions across the lifespan of the mouse. We found that astrocytes have region-specific transcriptional identities that change with age in a region-dependent manner. We validated our findings using fluorescence in situ hybridization and quantitative PCR. Detailed analysis of the differentially expressed genes in aging revealed that aged astrocytes take on a reactive phenotype of neuroinflammatory A1-like reactive astrocytes. Hippocampal and striatal astrocytes up-regulated a greater number of reactive astrocyte genes compared with cortical astrocytes. Moreover, aged brains formed many more A1 reactive astrocytes in response to the neuroinflammation inducer lipopolysaccharide. We found that the aging-induced up-regulation of reactive astrocyte genes was significantly reduced in mice lacking the microglial-secreted cytokines (IL-1α, TNF, and C1q) known to induce A1 reactive astrocyte formation, indicating that microglia promote astrocyte activation in aging. Since A1 reactive astrocytes lose the ability to carry out their normal functions, produce complement components, and release a toxic factor which kills neurons and oligodendrocytes, the aging-induced up-regulation of reactive genes by astrocytes could contribute to the cognitive decline in vulnerable brain regions in normal aging and contribute to the greater vulnerability of the aged brain to injury.
Collapse
|
240
|
Strickland MR, Koller EJ, Deng DZ, Ceballos-Diaz C, Golde TE, Chakrabarty P. Ifngr1 and Stat1 mediated canonical Ifn-γ signaling drives nigrostriatal degeneration. Neurobiol Dis 2018; 110:133-141. [PMID: 29196213 PMCID: PMC5748010 DOI: 10.1016/j.nbd.2017.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/20/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022] Open
Abstract
Brain expression of AAV-Ifn-γ leads to reactive gliosis, nigrostriatal degeneration and midbrain calcification in wild type mice. This mouse model phenocopies idiopathic basal ganglia calcification which is associated with Parkinsonian symptoms. To understand how the nigro-striatal pathway is selectively vulnerable to Ifn-γ, we determined if the phenotype is driven by canonical signaling intermediates, Ifngr1 and Stat1. Using focused bioinformatic analysis and rotarod testing, we show that neuroinflammation and motor abnormalities precede the appearance of midbrain neuropathologies in the brains of Ifn-γ mouse model. To test whether canonical Ifn-γ signaling is a key driver of progressive nigrostriatal degeneration, we overexpressed Ifn-γ in the brains of Ifngr1-/- and Stat1-/- mice. Expression of Ifn-γ in Ifngr1-/- mice did not result in any neuroinflammation, midbrain calcinosis or nigrostriatal degenerative pathology. Interestingly, in Stat1-/- mice, Ifn-γ expression resulted in gliosis without recapitulating the neurodegenerative phenotype. Overall, our data shows that canonical Ifn-γ signaling triggers midbrain calcinosis and nigrostriatal neurodegeneration, providing mechanistic insights into cytokine-driven selective neuronal vulnerability. Our study establishes the broader relevance of inflammatory signaling in neurodegenerative diseases and can potentially identify novel immunological targets for Parkinsonian syndromes.
Collapse
Affiliation(s)
- Michael R Strickland
- Center for Translational Research in Neurodegenerative Diseases, University of Florida, Gainesville, FL 32610, United States
| | - Emily J Koller
- Center for Translational Research in Neurodegenerative Diseases, University of Florida, Gainesville, FL 32610, United States; Department of Neuroscience, University of Florida, Gainesville, FL 32610, United States
| | - Doris Z Deng
- Center for Translational Research in Neurodegenerative Diseases, University of Florida, Gainesville, FL 32610, United States
| | - Carolina Ceballos-Diaz
- Center for Translational Research in Neurodegenerative Diseases, University of Florida, Gainesville, FL 32610, United States; Department of Neuroscience, University of Florida, Gainesville, FL 32610, United States
| | - Todd E Golde
- Center for Translational Research in Neurodegenerative Diseases, University of Florida, Gainesville, FL 32610, United States; Department of Neuroscience, University of Florida, Gainesville, FL 32610, United States; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Diseases, University of Florida, Gainesville, FL 32610, United States; Department of Neuroscience, University of Florida, Gainesville, FL 32610, United States; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
241
|
Cope TE, Rittman T, Borchert RJ, Jones PS, Vatansever D, Allinson K, Passamonti L, Vazquez Rodriguez P, Bevan-Jones WR, O'Brien JT, Rowe JB. Tau burden and the functional connectome in Alzheimer's disease and progressive supranuclear palsy. Brain 2018; 141:550-567. [PMID: 29293892 PMCID: PMC5837359 DOI: 10.1093/brain/awx347] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/25/2017] [Accepted: 10/29/2017] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease and progressive supranuclear palsy (PSP) represent neurodegenerative tauopathies with predominantly cortical versus subcortical disease burden. In Alzheimer's disease, neuropathology and atrophy preferentially affect 'hub' brain regions that are densely connected. It was unclear whether hubs are differentially affected by neurodegeneration because they are more likely to receive pathological proteins that propagate trans-neuronally, in a prion-like manner, or whether they are selectively vulnerable due to a lack of local trophic factors, higher metabolic demands, or differential gene expression. We assessed the relationship between tau burden and brain functional connectivity, by combining in vivo PET imaging using the ligand AV-1451, and graph theoretic measures of resting state functional MRI in 17 patients with Alzheimer's disease, 17 patients with PSP, and 12 controls. Strongly connected nodes displayed more tau pathology in Alzheimer's disease, independently of intrinsic connectivity network, validating the predictions of theories of trans-neuronal spread but not supporting a role for metabolic demands or deficient trophic support in tau accumulation. This was not a compensatory phenomenon, as the functional consequence of increasing tau burden in Alzheimer's disease was a progressive weakening of the connectivity of these same nodes, reducing weighted degree and local efficiency and resulting in weaker 'small-world' properties. Conversely, in PSP, unlike in Alzheimer's disease, those nodes that accrued pathological tau were those that displayed graph metric properties associated with increased metabolic demand and a lack of trophic support rather than strong functional connectivity. Together, these findings go some way towards explaining why Alzheimer's disease affects large scale connectivity networks throughout cortex while neuropathology in PSP is concentrated in a small number of subcortical structures. Further, we demonstrate that in PSP increasing tau burden in midbrain and deep nuclei was associated with strengthened cortico-cortical functional connectivity. Disrupted cortico-subcortical and cortico-brainstem interactions meant that information transfer took less direct paths, passing through a larger number of cortical nodes, reducing closeness centrality and eigenvector centrality in PSP, while increasing weighted degree, clustering, betweenness centrality and local efficiency. Our results have wide-ranging implications, from the validation of models of tau trafficking in humans to understanding the relationship between regional tau burden and brain functional reorganization.
Collapse
Affiliation(s)
- Thomas E Cope
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Robin J Borchert
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Deniz Vatansever
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Psychology, University of York, York, UK
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Kieren Allinson
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - W Richard Bevan-Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, Cambridge, UK
| |
Collapse
|
242
|
Bhatt PC, Pathak S, Kumar V, Panda BP. Attenuation of neurobehavioral and neurochemical abnormalities in animal model of cognitive deficits of Alzheimer's disease by fermented soybean nanonutraceutical. Inflammopharmacology 2018; 26:105-118. [PMID: 28791538 DOI: 10.1007/s10787-017-0381-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/22/2017] [Indexed: 11/28/2022]
Abstract
The present study was performed to evaluate the efficacy of nanonutraceuticals (NN) for attenuation of neurobehavioral and neurochemical abnormalities in Alzheimer's disease. Solid-state fermentation of soybean with Bacillus subtilis was performed to produce different metabolites (nattokinase, daidzin, genistin and glycitin and menaquinone-7). Intoxication of rats with colchicine caused impairment in learning and memory which was demonstrated in neurobehavioral paradigms (Morris water maze and passive avoidance) linked with decreased activity of acetylcholinesterase (AChE). NN treatment led to a significant increase in TLT in the retention trials as compared to acquisition trial TLT suggesting an improved learning and memory in rats. Further, treatment of NN caused an increase in the activity of AChE (42%), accompanied with a reduced activity of glutathione (42%), superoxide dismutase (43%) and catalase (41%). It also decreased the level of lipid peroxidation (28%) and protein carbonyl contents (30%) in hippocampus as compared to those treated with colchicine alone, suggesting a possible neuroprotective efficacy of NN. Interestingly, in silico studies also demonstrated an effective amyloid-β and BACE-1 inhibition activity. These findings clearly indicated that NN reversed colchicine-induced behavioral and neurochemical alterations through potent antioxidant activity and could possibly impart beneficial effects in cognitive defects associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Prakash Chandra Bhatt
- Microbial and Pharmaceutical Biotechnology Laboratory, Centre for Advanced Research in Pharmaceutical Sciences, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Shruti Pathak
- Microbial and Pharmaceutical Biotechnology Laboratory, Centre for Advanced Research in Pharmaceutical Sciences, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Allahabad, Uttar Pradesh, 211007, India
| | - Bibhu Prasad Panda
- Microbial and Pharmaceutical Biotechnology Laboratory, Centre for Advanced Research in Pharmaceutical Sciences, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
243
|
Abstract
Metabolic changes are hallmarks of aging and genetic and pharmacologic alterations of relevant pathways can extend life span. In this review, we will outline how cellular biochemistry and energy homeostasis change during aging. We will highlight protein quality control, mitochondria, epigenetics, nutrient-sensing pathways, as well as the interplay between these systems with respect to their impact on cellular health.
Collapse
Affiliation(s)
- Andre Catic
- Huffington Center on Aging, Stem Cells and Regenerative Medicine Center, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
244
|
Ohta Y, Soucy G, Phaneuf D, Audet JN, Gros-Louis F, Rouleau GA, Blasco H, Corcia P, Andersen PM, Nordin F, Yamashita T, Abe K, Julien JP. Sex-dependent effects of chromogranin B P413L allelic variant as disease modifier in amyotrophic lateral sclerosis. Hum Mol Genet 2018; 25:4771-4786. [PMID: 28175304 PMCID: PMC5418737 DOI: 10.1093/hmg/ddw304] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/28/2016] [Accepted: 08/25/2016] [Indexed: 11/14/2022] Open
Abstract
Recent genetic studies yielded conflicting results regarding a role for the variant chromogranin B (CHGB)P413L allele as a disease modifier in ALS. Moreover, potential deleterious effects of the CHGBP413L variant in ALS pathology have not been investigated. Here we report that in transfected cultured cells, the variant CHGBL413 protein exhibited aberrant properties including mislocalization, failure to interact with mutant superoxide dismutase 1 (SOD1) and defective secretion. The CHGBL413 transgene in SOD1G37R mice precipitated disease onset and pathological changes related to misfolded SOD1 specifically in female mice. However, the CHGBL413 variant also slowed down disease progression in SOD1G37R mice, which is in line with a very slow disease progression that we report for a Swedish woman with ALS who is carrier of two mutant SOD1D90A alleles and two variant CHGBP413L and CHGBR458Q alleles. In contrast, overexpression of the common CHGBP413 allele in SOD1G37R mice did not affect disease onset but significantly accelerated disease progression and pathological changes. As in transgenic mice, the CHGBP413L allele conferred an earlier ALS disease onset in women of Japanese and French Canadian origins with less effect in men. Evidence is presented that the sex-dependent effects of CHGBL413 allelic variant in ALS may arise from enhanced neuronal expression of CHGB in females because of a sex-determining region Y element in the gene promoter. Thus, our results suggest that CHGB variants may act as modifiers of onset and progression in some ALS populations and especially in females because of higher expression levels compared to males.
Collapse
Affiliation(s)
- Yasuyuki Ohta
- Research Centre of Institut universitaire en santé mentale de Québec, Québec, QC, Canada,Department of Psychiatry and Neuroscience, Laval University, Québec, QC, Canada
| | - Genevieve Soucy
- Research Centre of Institut universitaire en santé mentale de Québec, Québec, QC, Canada,Department of Psychiatry and Neuroscience, Laval University, Québec, QC, Canada
| | - Daniel Phaneuf
- Research Centre of Institut universitaire en santé mentale de Québec, Québec, QC, Canada,Department of Psychiatry and Neuroscience, Laval University, Québec, QC, Canada
| | - Jean-Nicolas Audet
- Research Centre of Institut universitaire en santé mentale de Québec, Québec, QC, Canada,Department of Psychiatry and Neuroscience, Laval University, Québec, QC, Canada
| | - François Gros-Louis
- CHU de Québec Research Center, LOEX Hôpital de l'Enfant-Jésus, Quebec, QC, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Hélène Blasco
- Centre de Ressources et Compétences SLA (CRCSLA), Federation des CRCSLA Tours-Limoges LITORALS, INSERM U 930, Université François-Rabelais de Tours, France
| | - Philippe Corcia
- Centre de Ressources et Compétences SLA (CRCSLA), Federation des CRCSLA Tours-Limoges LITORALS, INSERM U 930, Université François-Rabelais de Tours, France
| | - Peter M Andersen
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden,Department of Neurology, Ulm University, Ulm, Germany
| | - Frida Nordin
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jean-Pierre Julien
- Research Centre of Institut universitaire en santé mentale de Québec, Québec, QC, Canada,Department of Psychiatry and Neuroscience, Laval University, Québec, QC, Canada
| |
Collapse
|
245
|
Manuello J, Nani A, Premi E, Borroni B, Costa T, Tatu K, Liloia D, Duca S, Cauda F. The Pathoconnectivity Profile of Alzheimer's Disease: A Morphometric Coalteration Network Analysis. Front Neurol 2018; 8:739. [PMID: 29472885 PMCID: PMC5810291 DOI: 10.3389/fneur.2017.00739] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/21/2017] [Indexed: 01/18/2023] Open
Abstract
Gray matter alterations are typical features of brain disorders. However, they do not impact on the brain randomly. Indeed, it has been suggested that neuropathological processes can selectively affect certain assemblies of neurons, which typically are at the center of crucial functional networks. Because of their topological centrality, these areas form a core set that is more likely to be affected by neuropathological processes. In order to identify and study the pattern formed by brain alterations in patients’ with Alzheimer’s disease (AD), we devised an innovative meta-analytic method for analyzing voxel-based morphometry data. This methodology enabled us to discover that in AD gray matter alterations do not occur randomly across the brain but, on the contrary, follow identifiable patterns of distribution. This alteration pattern exhibits a network-like structure composed of coaltered areas that can be defined as coatrophy network. Within the coatrophy network of AD, we were able to further identify a core subnetwork of coaltered areas that includes the left hippocampus, left and right amygdalae, right parahippocampal gyrus, and right temporal inferior gyrus. In virtue of their network centrality, these brain areas can be thought of as pathoconnectivity hubs.
Collapse
Affiliation(s)
- Jordi Manuello
- GCS-fMRI, Department of Psychology, Koelliker Hospital, University of Turin, Turin, Italy.,FOCUS Laboratory, Department of Psychology, University of Turin, Turin, Italy
| | - Andrea Nani
- GCS-fMRI, Department of Psychology, Koelliker Hospital, University of Turin, Turin, Italy.,FOCUS Laboratory, Department of Psychology, University of Turin, Turin, Italy.,Michael Trimble Neuropsychiatry Research Group, Birmingham and Solihull Mental Health NHS Foundation Trust, Birmingham, United Kingdom
| | - Enrico Premi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Tommaso Costa
- GCS-fMRI, Department of Psychology, Koelliker Hospital, University of Turin, Turin, Italy.,FOCUS Laboratory, Department of Psychology, University of Turin, Turin, Italy
| | - Karina Tatu
- GCS-fMRI, Department of Psychology, Koelliker Hospital, University of Turin, Turin, Italy.,FOCUS Laboratory, Department of Psychology, University of Turin, Turin, Italy
| | - Donato Liloia
- FOCUS Laboratory, Department of Psychology, University of Turin, Turin, Italy
| | - Sergio Duca
- GCS-fMRI, Department of Psychology, Koelliker Hospital, University of Turin, Turin, Italy
| | - Franco Cauda
- GCS-fMRI, Department of Psychology, Koelliker Hospital, University of Turin, Turin, Italy.,FOCUS Laboratory, Department of Psychology, University of Turin, Turin, Italy
| |
Collapse
|
246
|
Penke B, Bogár F, Crul T, Sántha M, Tóth ME, Vígh L. Heat Shock Proteins and Autophagy Pathways in Neuroprotection: from Molecular Bases to Pharmacological Interventions. Int J Mol Sci 2018; 19:E325. [PMID: 29361800 PMCID: PMC5796267 DOI: 10.3390/ijms19010325] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases (NDDs) such as Alzheimer's disease, Parkinson's disease and Huntington's disease (HD), amyotrophic lateral sclerosis, and prion diseases are all characterized by the accumulation of protein aggregates (amyloids) into inclusions and/or plaques. The ubiquitous presence of amyloids in NDDs suggests the involvement of disturbed protein homeostasis (proteostasis) in the underlying pathomechanisms. This review summarizes specific mechanisms that maintain proteostasis, including molecular chaperons, the ubiquitin-proteasome system (UPS), endoplasmic reticulum associated degradation (ERAD), and different autophagic pathways (chaperon mediated-, micro-, and macro-autophagy). The role of heat shock proteins (Hsps) in cellular quality control and degradation of pathogenic proteins is reviewed. Finally, putative therapeutic strategies for efficient removal of cytotoxic proteins from neurons and design of new therapeutic targets against the progression of NDDs are discussed.
Collapse
Affiliation(s)
- Botond Penke
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Dóm Square 8, Hungary.
| | - Ferenc Bogár
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Dóm Square 8, Hungary.
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, H-6720 Szeged, Dóm Square 8, Hungary.
| | - Tim Crul
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| | - László Vígh
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| |
Collapse
|
247
|
Aluminum-Induced Neural Cell Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1091:129-160. [DOI: 10.1007/978-981-13-1370-7_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
248
|
Sehgal SA, Hammad MA, Tahir RA, Akram HN, Ahmad F. Current Therapeutic Molecules and Targets in Neurodegenerative Diseases Based on in silico Drug Design. Curr Neuropharmacol 2018; 16:649-663. [PMID: 29542412 PMCID: PMC6080102 DOI: 10.2174/1570159x16666180315142137] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 01/01/2018] [Accepted: 03/02/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND As the number of elderly persons increases, neurodegenerative diseases are becoming ubiquitous. There is currently a great need for knowledge concerning management of oldage neurodegenerative diseases; the most important of which are: Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis, and Huntington's disease. OBJECTIVE To summarize the potential of computationally predicted molecules and targets against neurodegenerative diseases. METHOD Review of literature published since 1997 against neurodegenerative diseases, utilizing as keywords: in silico, Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis ALS, and Huntington's disease was conducted. RESULTS AND CONCLUSION Due to the costs associated with experimentation and current ethical law, performing experiments directly on living organisms has become much more difficult. In this scenario, in silico techniques have been successful and have become powerful tools in the search to cure disease. Researchers use the Computer Aided Drug Design pipeline which: 1) generates 3- dimensional structures of target proteins through homology modeling 2) achieves stabilization through molecular dynamics simulation, and 3) exploits molecular docking through large compound libraries. Next generation sequencing is continually producing enormous amounts of raw sequence data while neuroimaging is producing a multitude of raw image data. To solve such pressing problems, these new tools and algorithms are required. This review elaborates precise in silico tools and techniques for drug targets, active molecules, and molecular docking studies, together with future prospects and challenges concerning possible breakthroughs in Alzheimer's, Parkinson's, Amyotrophic Lateral Sclerosis, and Huntington's disease.
Collapse
Affiliation(s)
- Sheikh Arslan Sehgal
- Address correspondence to this author at the State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences; Beijing, China; E-mail:
| | | | | | | | | |
Collapse
|
249
|
Somayaji MR, Przekwas AJ, Gupta RK. Combination Therapy for Multi-Target Manipulation of Secondary Brain Injury Mechanisms. Curr Neuropharmacol 2018; 16:484-504. [PMID: 28847295 PMCID: PMC6018188 DOI: 10.2174/1570159x15666170828165711] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a major healthcare problem that affects millions of people worldwide. Despite advances in understanding and developing preventative and treatment strategies using preclinical animal models, clinical trials to date have failed, and a 'magic bullet' for effectively treating TBI-induced damage does not exist. Thus, novel pharmacological strategies to effectively manipulate the complex and heterogeneous pathophysiology of secondary injury mechanisms are needed. Given that goal, this paper discusses the relevance and advantages of combination therapies (COMTs) for 'multi-target manipulation' of the secondary injury cascade by administering multiple drugs to achieve an optimal therapeutic window of opportunity (e.g., temporally broad window) and compares these regimens to monotherapies that manipulate a single target with a single drug at a given time. Furthermore, we posit that integrated mechanistic multiscale models that combine primary injury biomechanics, secondary injury mechanobiology/neurobiology, physiology, pharmacology and mathematical programming techniques could account for vast differences in the biological space and time scales and help to accelerate drug development, to optimize pharmacological COMT protocols and to improve treatment outcomes.
Collapse
Affiliation(s)
| | | | - Raj K. Gupta
- Department of Defense Blast Injury Research Program Coordinating Office, U.S. Army Medical Research and Materiel Command, Fort Detrick, MD, USA
| |
Collapse
|
250
|
How do morphological alterations caused by chronic pain distribute across the brain? A meta-analytic co-alteration study. NEUROIMAGE-CLINICAL 2017; 18:15-30. [PMID: 30023166 PMCID: PMC5987668 DOI: 10.1016/j.nicl.2017.12.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/19/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023]
Abstract
•In chronic pain, gray matter (GM) alterations are not distributed randomly across the brain.•The pattern of co-alterations resembles that of brain connectivity.•The alterations' distribution partly rely on the pathways of functional connectivity.•This method allows us to identify tendencies in the distribution of GM co-alteration related to chronic pain.
Collapse
|