Hallmarks of neurodegenerative disease: A systems pharmacology perspective

Age‐related central neurodegenerative diseases, such as Alzheimer's and Parkinson's disease, are a rising public health concern and have been plagued by repeated drug development failures. The complex nature and poor mechanistic understanding of the etiology of neurodegenerative diseases has hindered the discovery and development of effective disease‐modifying therapeutics. Quantitative systems pharmacology models of neurodegeneration diseases may be useful tools to enhance the understanding of pharmacological intervention strategies and to reduce drug attrition rates. Due to the similarities in pathophysiological mechanisms across neurodegenerative diseases, especially at the cellular and molecular levels, we envision the possibility of structural components that are conserved across models of neurodegenerative diseases. Conserved structural submodels can be viewed as building blocks that are pieced together alongside unique disease components to construct quantitative systems pharmacology (QSP) models of neurodegenerative diseases. Model parameterization would likely be different between the different types of neurodegenerative diseases as well as individual patients. Formulating our mechanistic understanding of neurodegenerative pathophysiology as a mathematical model could aid in the identification and prioritization of drug targets and combinatorial treatment strategies, evaluate the role of patient characteristics on disease progression and therapeutic response, and serve as a central repository of knowledge. Here, we provide a background on neurodegenerative diseases, highlight hallmarks of neurodegeneration, and summarize previous QSP models of neurodegenerative diseases.

[1]  Houeto Jean-Luc [Parkinson's disease]. , 2022, La Revue du praticien.

[2]  Michiel A. van der Vlag,et al.  Brain simulation as a cloud service: The Virtual Brain on EBRAINS , 2022, NeuroImage.

[3]  J. Burke,et al.  Quantitative systems pharmacology model for Alzheimer’s disease to predict the effect of aducanumab on brain amyloid , 2022, CPT: pharmacometrics & systems pharmacology.

[4]  B. Xiao,et al.  Microglial Phenotypic Transition: Signaling Pathways and Influencing Modulators Involved in Regulation in Central Nervous System Diseases , 2021, Frontiers in Cellular Neuroscience.

[5]  K. Blennow,et al.  Plasma phosphorylated tau 217 and phosphorylated tau 181 as biomarkers in Alzheimer's disease and frontotemporal lobar degeneration: a retrospective diagnostic performance study , 2021, The Lancet Neurology.

[6]  Eleanor Catherine Sams Oligodendrocytes in the aging brain , 2021, Neuronal signaling.

[7]  V. Alexaki The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives , 2021, Cells.

[8]  D. Lauffenburger,et al.  Systematic in silico analysis of clinically tested drugs for reducing amyloid‐beta plaque accumulation in Alzheimer's disease , 2021, Alzheimer's & dementia : the journal of the Alzheimer's Association.

[9]  H. Geerts,et al.  Exploring the relation between BOLD fMRI and cognitive performance using a computer-based quantitative systems pharmacology model: Applications to the COMTVAL158MET genotype and ketamine , 2021, European Neuropsychopharmacology.

[10]  B. Hyman,et al.  Acetylated tau inhibits chaperone-mediated autophagy and promotes tau pathology propagation in mice , 2021, Nature Communications.

[11]  S. Muldoon,et al.  Quantitative systems pharmacology in neuroscience: Novel methodologies and technologies , 2021, CPT: pharmacometrics & systems pharmacology.

[12]  Kira E. Poskanzer,et al.  Reactive astrocyte nomenclature, definitions, and future directions , 2021, Nature Neuroscience.

[13]  K. Borges,et al.  CNS glucose metabolism in Amyotrophic Lateral Sclerosis: a therapeutic target? , 2021, Cell & Bioscience.

[14]  A. Regev,et al.  Single cell RNA sequencing of human microglia uncovers a subset associated with Alzheimer’s disease , 2020, Nature Communications.

[15]  F. Nekka,et al.  An integrative model of Parkinson’s disease treatment including levodopa pharmacokinetics, dopamine kinetics, basal ganglia neurotransmission and motor action throughout disease progression , 2020, Journal of pharmacokinetics and pharmacodynamics.

[16]  A. Brickman,et al.  Metabolic syndrome and its components in relation to in vivo brain amyloid and neurodegeneration in late middle age , 2020, Neurobiology of Aging.

[17]  Rajanikanth Vadigepalli,et al.  Credible practice of modeling and simulation in healthcare: ten rules from a multidisciplinary perspective , 2020, Journal of Translational Medicine.

[18]  J. Cummings,et al.  Repurposed agents in the Alzheimer’s disease drug development pipeline , 2020, Alzheimer's Research & Therapy.

[19]  J. Suckling,et al.  Evidence-based prevention of Alzheimer's disease: systematic review and meta-analysis of 243 observational prospective studies and 153 randomised controlled trials , 2020, Journal of Neurology, Neurosurgery, and Psychiatry.

[20]  Marie P. Schützmann,et al.  Endo-lysosomal Aβ concentration and pH trigger formation of Aβ oligomers that potently induce Tau missorting , 2020, Nature Communications.

[21]  Derek H. Oakley,et al.  Tau molecular diversity contributes to clinical heterogeneity in Alzheimer’s disease , 2020, Nature Medicine.

[22]  Bin Zhang,et al.  Large-scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation , 2020, Nature Medicine.

[23]  H. Geerts,et al.  Learning from amyloid trials in Alzheimer's disease. A virtual patient analysis using a quantitative systems pharmacology approach , 2020, Alzheimer's & dementia : the journal of the Alzheimer's Association.

[24]  V. Sansone,et al.  Emerging Drugs for the Treatment of Amyotrophic Lateral Sclerosis: A Focus on Recent Phase 2 Trials , 2020, Expert opinion on emerging drugs.

[25]  Dean P. Jones,et al.  Reactive oxygen species (ROS) as pleiotropic physiological signalling agents , 2020, Nature Reviews Molecular Cell Biology.

[26]  R. Borisyuk,et al.  A Population Model of Deep Brain Stimulation in Movement Disorders From Circuits to Cells , 2020, Frontiers in Human Neuroscience.

[27]  N. Mizushima,et al.  Lysosome biology in autophagy , 2020, Cell Discovery.

[28]  Yahiya Y. Syed Sodium Oligomannate: First Approval , 2020, Drugs.

[29]  R. Sankowski,et al.  Microglia Heterogeneity in the Single-Cell Era. , 2020, Cell reports.

[30]  Andrew B Singleton,et al.  The genetic architecture of Parkinson's disease , 2020, The Lancet Neurology.

[31]  S. Wilton,et al.  ALS Genetics, Mechanisms, and Therapeutics: Where Are We Now? , 2019, Front. Neurosci..

[32]  Ulrike Winkler,et al.  Heterogeneity of Astrocytes in Grey and White Matter , 2019, Neurochemical Research.

[33]  L. Goldstein,et al.  Amyloid-β-independent regulators of tau pathology in Alzheimer disease , 2019, Nature Reviews Neuroscience.

[34]  S. Hasselbalch,et al.  Ageing as a risk factor for neurodegenerative disease , 2019, Nature Reviews Neurology.

[35]  D. Holtzman,et al.  The microbiome: A target for Alzheimer disease? , 2019, Cell Research.

[36]  Jian Ding,et al.  Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression , 2019, Cell Research.

[37]  Sung Hoon Baik,et al.  A Breakdown in Metabolic Reprogramming Causes Microglia Dysfunction in Alzheimer's Disease. , 2019, Cell metabolism.

[38]  Nikhil Panicker,et al.  Transneuronal Propagation of Pathologic α-Synuclein from the Gut to the Brain Models Parkinson’s Disease , 2019, Neuron.

[39]  M. Prinz,et al.  Macrophages at CNS interfaces: ontogeny and function in health and disease , 2019, Nature Reviews Neuroscience.

[40]  Theodore W. Berger,et al.  Pathogenic Processes Underlying Alzheimer’s Disease: Modeling the Effects of Amyloid Beta on Synaptic Transmission , 2019, 2019 41st Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC).

[41]  Shuai Jiang,et al.  Mitochondrial electron transport chain, ROS generation and uncoupling (Review) , 2019, International journal of molecular medicine.

[42]  M. Kitazawa,et al.  Metal Toxicity Links to Alzheimer's Disease and Neuroinflammation. , 2019, Journal of molecular biology.

[43]  W. Teo,et al.  Parkinson's Disease and the Environment , 2019, Front. Neurol..

[44]  D. Allan Butterfield,et al.  Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease , 2019, Nature Reviews Neuroscience.

[45]  E. Kuhl,et al.  Prion-like spreading of Alzheimer’s disease within the brain’s connectome , 2019, bioRxiv.

[46]  G. Logroscino,et al.  A critical appraisal of amyloid-β-targeting therapies for Alzheimer disease , 2019, Nature Reviews Neurology.

[47]  J. Cummings,et al.  Alzheimer's disease drug development pipeline: 2019 , 2019, Alzheimer's & dementia.

[48]  Vijayalakshmi Chelliah,et al.  Mathematical Biology Models of Parkinson's Disease , 2018, CPT: pharmacometrics & systems pharmacology.

[49]  Neil Benson,et al.  Quantitative Systems Pharmacology Model for Alzheimer Disease Indicates Targeting Sphingolipid Dysregulation as Potential Treatment Option , 2018, CPT: pharmacometrics & systems pharmacology.

[50]  S. Hickman,et al.  Microglia in neurodegeneration , 2018, Nature Neuroscience.

[51]  S. Studenski,et al.  Metabolic Syndrome and Amyloid Accumulation in the Aging Brain. , 2018, Journal of Alzheimer's disease : JAD.

[52]  M. Jaiswal,et al.  Riluzole and edaravone: A tale of two amyotrophic lateral sclerosis drugs , 2018, Medicinal research reviews.

[53]  E. Sigurdsson,et al.  Tau-targeting therapies for Alzheimer disease , 2018, Nature Reviews Neurology.

[54]  Alberto Pupi,et al.  Mediterranean diet and 3-year Alzheimer brain biomarker changes in middle-aged adults , 2018, Neurology.

[55]  J. Belleroche,et al.  Amyotrophic lateral sclerosis (ALS) and Alzheimer’s disease (AD) are characterised by differential activation of ER stress pathways: focus on UPR target genes , 2018, Cell Stress and Chaperones.

[56]  P. Tariot,et al.  Randomized Trial of Verubecestat for Mild‐to‐Moderate Alzheimer's Disease , 2018, The New England journal of medicine.

[57]  Alberto J Espay,et al.  Disease Modification in Parkinson's Disease: Current Approaches, Challenges, and Future Considerations , 2018, Movement disorders : official journal of the Movement Disorder Society.

[58]  Yan-Qin Shen,et al.  Dysbiosis of gut microbiota and microbial metabolites in Parkinson’s Disease , 2018, Ageing Research Reviews.

[59]  Jie Zheng,et al.  Composite mathematical modeling of calcium signaling behind neuronal cell death in Alzheimer’s disease , 2018, BMC Systems Biology.

[60]  O. Demin,et al.  A mathematical model of multisite phosphorylation of tau protein , 2018, PloS one.

[61]  Hugo Geerts,et al.  Impact of amyloid-beta changes on cognitive outcomes in Alzheimer’s disease: analysis of clinical trials using a quantitative systems pharmacology model , 2018, Alzheimer's Research & Therapy.

[62]  Fan Wu,et al.  Preclinical QSP Modeling in the Pharmaceutical Industry: An IQ Consortium Survey Examining the Current Landscape , 2018, CPT: pharmacometrics & systems pharmacology.

[63]  Hongchun Li,et al.  Connecting Neuronal Cell Protective Pathways and Drug Combinations in a Huntington’s Disease Model through the Application of Quantitative Systems Pharmacology , 2017, Scientific Reports.

[64]  Sterling C. Johnson,et al.  Gut microbiome alterations in Alzheimer’s disease , 2017, Scientific Reports.

[65]  T. Nicholas,et al.  Studying the Progression of Amyloid Pathology and Its Therapy Using Translational Longitudinal Model of Accumulation and Distribution of Amyloid Beta , 2017, CPT: pharmacometrics & systems pharmacology.

[66]  T. Nicholas,et al.  A Translational Systems Pharmacology Model for Aβ Kinetics in Mouse, Monkey, and Human , 2017, CPT: pharmacometrics & systems pharmacology.

[67]  J. Cummings,et al.  Lessons Learned from Alzheimer Disease: Clinical Trials with Negative Outcomes , 2017, Clinical and translational science.

[68]  C. Hetz,et al.  ER stress and the unfolded protein response in neurodegeneration , 2017, Nature Reviews Neurology.

[69]  Matthew P Frosch,et al.  Enhanced Tau Aggregation in the Presence of Amyloid β. , 2017, The American journal of pathology.

[70]  D. Rubinsztein,et al.  Huntington's Disease: Mechanisms of Pathogenesis and Therapeutic Strategies. , 2017, Cold Spring Harbor perspectives in medicine.

[71]  I. Amit,et al.  A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease , 2017, Cell.

[72]  M. Goedert,et al.  Propagation of Tau aggregates , 2017, Molecular Brain.

[73]  Alan C. Evans,et al.  Multifactorial causal model of brain (dis)organization and therapeutic intervention: Application to Alzheimer’s disease , 2017, NeuroImage.

[74]  M. Prinz,et al.  Ontogeny and homeostasis of CNS myeloid cells , 2017, Nature Immunology.

[75]  G. Frisoni,et al.  Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly , 2017, Neurobiology of Aging.

[76]  Marco Prinz,et al.  The role of peripheral immune cells in the CNS in steady state and disease , 2017, Nature Neuroscience.

[77]  Roman Borisyuk,et al.  A Network Model of Local Field Potential Activity in Essential Tremor and the Impact of Deep Brain Stimulation , 2017, PLoS computational biology.

[78]  S. Herculano‐Houzel,et al.  The search for true numbers of neurons and glial cells in the human brain: A review of 150 years of cell counting , 2016, The Journal of comparative neurology.

[79]  H. Braak,et al.  Potential Pathways of Abnormal Tau and α-Synuclein Dissemination in Sporadic Alzheimer's and Parkinson's Diseases. , 2016, Cold Spring Harbor perspectives in biology.

[80]  Viktor K. Jirsa,et al.  Transcranial direct current stimulation changes resting state functional connectivity: A large-scale brain network modeling study , 2016, NeuroImage.

[81]  R. Ransohoff How neuroinflammation contributes to neurodegeneration , 2016, Science.

[82]  F. Heppner,et al.  High-fat diet-induced brain region-specific phenotypic spectrum of CNS resident microglia , 2016, Acta Neuropathologica.

[83]  M. Albert,et al.  Blood glucose levels and cortical thinning in cognitively normal, middle-aged adults , 2016, Journal of the Neurological Sciences.

[84]  T. Montine,et al.  Seed‐competent high‐molecular‐weight tau species accumulates in the cerebrospinal fluid of Alzheimer's disease mouse model and human patients , 2016, Annals of neurology.

[85]  D. Isaacson,et al.  Early Treatment Critical: Bexarotene Reduces Amyloid-Beta Burden In Silico , 2016, PloS one.

[86]  J. Hardy,et al.  The amyloid hypothesis of Alzheimer's disease at 25 years , 2016, EMBO molecular medicine.

[87]  Hugo Geerts,et al.  A Humanized Clinically Calibrated Quantitative Systems Pharmacology Model for Hypokinetic Motor Symptoms in Parkinson’s Disease , 2016, Front. Pharmacol..

[88]  Rodney W. Johnson,et al.  Neuro-immune dysfunction during brain aging: new insights in microglial cell regulation. , 2016, Current opinion in pharmacology.

[89]  A. Cuervo,et al.  Proteostasis and aging , 2015, Nature Network Boston.

[90]  Ali Elkamel,et al.  Modeling the Interaction between β-Amyloid Aggregates and Choline Acetyltransferase Activity and Its Relation with Cholinergic Dysfunction through Two-Enzyme/Two-Compartment Model , 2015, Comput. Math. Methods Medicine.

[91]  Danielle J. Harvey,et al.  The Alzheimer's Disease Neuroimaging Initiative phase 2: Increasing the length, breadth, and depth of our understanding , 2015, Alzheimer's & Dementia.

[92]  Elise M. Braatz,et al.  A mathematical model of insulin resistance in Parkinson's disease , 2015, Comput. Biol. Chem..

[93]  Sterling C. Johnson,et al.  Insulin resistance predicts brain amyloid deposition in late middle-aged adults , 2015, Alzheimer's & Dementia.

[94]  Alexander L Green,et al.  Tremor Reduction by Deep Brain Stimulation Is Associated With Gamma Power Suppression in Parkinson's Disease , 2015, Neuromodulation : journal of the International Neuromodulation Society.

[95]  W. M. van der Flier,et al.  The metabolic syndrome in a memory clinic population: Relation with clinical profile and prognosis , 2015, Journal of the Neurological Sciences.

[96]  S. Pedraza,et al.  Hypothalamic damage is associated with inflammatory markers and worse cognitive performance in obese subjects. , 2015, The Journal of clinical endocrinology and metabolism.

[97]  K. Guan,et al.  mTOR: a pharmacologic target for autophagy regulation. , 2015, The Journal of clinical investigation.

[98]  J. Grutzendler,et al.  Microglia constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques , 2014, Nature Communications.

[99]  Ivet Bahar,et al.  BalestraWeb: efficient online evaluation of drug–target interactions , 2014, Bioinform..

[100]  G. Wenk,et al.  Metabolic disturbances in diseases with neurological involvement. , 2014, Aging and disease.

[101]  Clifford R. Jack,et al.  Association of type 2 diabetes with brain atrophy and cognitive impairment , 2014, Neurology.

[102]  Lei Shi,et al.  Challenges for an enzymatic reaction kinetics database , 2014, The FEBS journal.

[103]  Míriam R. García,et al.  A single compartment model of pacemaking in dissasociated Substantia nigra neurons , 2013, Journal of Computational Neuroscience.

[104]  Z. Oltvai,et al.  Predicting Drug–Target Interactions Using Probabilistic Matrix Factorization , 2013, J. Chem. Inf. Model..

[105]  Leif Benner,et al.  Ketogenic Diet Improves Motor Performance but Not Cognition in Two Mouse Models of Alzheimer’s Pathology , 2013, PloS one.

[106]  U. Hofmann,et al.  An Interactive Channel Model of the Basal Ganglia: Bifurcation Analysis Under Healthy and Parkinsonian Conditions , 2013, Journal of mathematical neuroscience.

[107]  C. Cowan,et al.  Are Tau Aggregates Toxic or Protective in Tauopathies? , 2013, Front. Neurol..

[108]  W. Le,et al.  Genetics of amyotrophic lateral sclerosis: an update , 2013, Molecular Neurodegeneration.

[109]  P. Brown,et al.  Adaptive Deep Brain Stimulation In Advanced Parkinson Disease , 2013, Annals of neurology.

[110]  Michel Goedert,et al.  Tau pathology and neurodegeneration , 2013, The Lancet Neurology.

[111]  M. Mattson,et al.  A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer's disease , 2013, Neurobiology of Aging.

[112]  L. Kappos,et al.  Rapamycin Attenuates the Progression of Tau Pathology in P301S Tau Transgenic Mice , 2013, PloS one.

[113]  Matthew H. Bailey,et al.  GWAS of Cerebrospinal Fluid Tau Levels Identifies Risk Variants for Alzheimer’s Disease , 2013, Neuron.

[114]  Tomio Sasaki,et al.  Altered Expression of Diabetes-Related Genes in Alzheimer's Disease Brains: The Hisayama Study , 2013, Cerebral cortex.

[115]  H Geerts,et al.  A Quantitative System Pharmacology Computer Model for Cognitive Deficits in Schizophrenia , 2013, CPT: pharmacometrics & systems pharmacology.

[116]  I. Ferrer,et al.  Triheptanoin supplementation to ketogenic diet curbs cognitive impairment in APP/PS1 mice used as a model of familial Alzheimer's disease. , 2013, Current Alzheimer research.

[117]  King C. Li,et al.  A system mathematical model of a cell-cell communication network in amyotrophic lateral sclerosis. , 2013, Molecular bioSystems.

[118]  A. Convit,et al.  Obesity and Metabolic Syndrome and Functional and Structural Brain Impairments in Adolescence , 2012, Pediatrics.

[119]  Timothy D Verstynen,et al.  Increased Body Mass Index Is Associated With a Global and Distributed Decrease in White Matter Microstructural Integrity , 2012, Psychosomatic medicine.

[120]  D. Cai,et al.  IKKβ/NF-κB Disrupts Adult Hypothalamic Neural Stem Cells to Mediate Neurodegenerative Mechanism of Dietary Obesity and Pre-Diabetes , 2012, Nature Cell Biology.

[121]  Hugo Geerts,et al.  A Quantitative Systems Pharmacology Computer Model for Schizophrenia Efficacy and Extrapyramidal Side Effects , 2012 .

[122]  Domenico Consoli,et al.  Dementia is associated with Insulin Resistance in patients with Parkinson's Disease , 2012, Journal of the Neurological Sciences.

[123]  M. Weiner,et al.  A Network Diffusion Model of Disease Progression in Dementia , 2012, Neuron.

[124]  F. Panza,et al.  Metabolic syndrome as a risk factor for neurological disorders , 2012, Cellular and Molecular Life Sciences.

[125]  J. Flier,et al.  Remodeling of the arcuate nucleus energy-balance circuit is inhibited in obese mice. , 2012, The Journal of clinical investigation.

[126]  D. Rubinsztein,et al.  Autophagy and Aging , 2011, Cell.

[127]  B. Hyman,et al.  Neuropathological alterations in Alzheimer disease. , 2011, Cold Spring Harbor perspectives in medicine.

[128]  Pico Caroni,et al.  Selective Neuronal Vulnerability in Neurodegenerative Diseases: from Stressor Thresholds to Degeneration , 2011, Neuron.

[129]  A. Convit,et al.  Disinhibited Eating in Obese Adolescents Is Associated With Orbitofrontal Volume Reductions and Executive Dysfunction , 2011, Obesity.

[130]  H. Kettenmann,et al.  Physiology of microglia. , 2011, Physiological reviews.

[131]  M. Spillantini,et al.  Induction of the unfolded protein response by α‐synuclein in experimental models of Parkinson’s disease , 2011, Journal of neurochemistry.

[132]  B. Viollet,et al.  AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1 , 2011, Nature Cell Biology.

[133]  R. Roos,et al.  Huntington's disease: a clinical review , 2010, Orphanet journal of rare diseases.

[134]  T. Wyss-Coray,et al.  Beclin 1 complex in autophagy and Alzheimer disease. , 2010, Archives of neurology.

[135]  Hugo Geerts,et al.  Not all partial dopamine D2 receptor agonists are the same in treating schizophrenia. Exploring the effects of bifeprunox and aripiprazole using a computer model of a primate striatal dopaminergic synapse , 2010, Neuropsychiatric disease and treatment.

[136]  D. Westaway,et al.  Lysosomal Proteolysis and Autophagy Require Presenilin 1 and Are Disrupted by Alzheimer-Related PS1 Mutations , 2010, Cell.

[137]  Jayanta Debnath,et al.  Inhibition of mTOR by Rapamycin Abolishes Cognitive Deficits and Reduces Amyloid-β Levels in a Mouse Model of Alzheimer's Disease , 2010, PloS one.

[138]  D. Sulzer,et al.  CARGO RECOGNITION FAILURE IS RESPONSIBLE FOR INEFFICIENT AUTOPHAGY IN HUNTINGTON’S DISEASE , 2010, Nature Neuroscience.

[139]  S. Luo,et al.  Huntingtin Interacts with the Cue Domain of gp78 and Inhibits gp78 Binding to Ubiquitin and p97/VCP , 2010, PloS one.

[140]  Jennifer Goldman,et al.  Huntington disease , 2010, Principles and Practice of Movement Disorders.

[141]  M. Sofroniew,et al.  Astrocytes: biology and pathology , 2009, Acta Neuropathologica.

[142]  Eberhard O Voit,et al.  Computational analysis of determinants of dopamine (DA) dysfunction in DA nerve terminals , 2009, Synapse.

[143]  Hans Lassmann,et al.  Oligodendrocytes: biology and pathology , 2009, Acta Neuropathologica.

[144]  L. Calderón-Garcidueñas,et al.  Air pollution: mechanisms of neuroinflammation and CNS disease , 2009, Trends in Neurosciences.

[145]  Robert L. Green,et al.  A pragmatic approach to biochemical systems theory applied to an α-synuclein-based model of Parkinson's disease , 2009, Journal of Neuroscience Methods.

[146]  B. Barres The Mystery and Magic of Glia: A Perspective on Their Roles in Health and Disease , 2008, Neuron.

[147]  F. LaFerla,et al.  Aβ inhibits the proteasome and enhances amyloid and tau accumulation , 2008, Neurobiology of Aging.

[148]  Ralph A. Nixon,et al.  Autophagy Induction and Autophagosome Clearance in Neurons: Relationship to Autophagic Pathology in Alzheimer's Disease , 2008, The Journal of Neuroscience.

[149]  Eberhard O. Voit,et al.  Computational Systems Analysis of Dopamine Metabolism , 2008, PloS one.

[150]  H. Ichijo,et al.  ALS-linked mutant SOD1 induces ER stress- and ASK1-dependent motor neuron death by targeting Derlin-1. , 2008, Genes & development.

[151]  V. Kostic,et al.  Glucose homeostasis in Huntington disease: abnormalities in insulin sensitivity and early-phase insulin secretion. , 2008, Archives of neurology.

[152]  Peter T Lansbury,et al.  Dopamine-modified alpha-synuclein blocks chaperone-mediated autophagy. , 2008, The Journal of clinical investigation.

[153]  Nelson J. Trujillo-Barreto,et al.  Biophysical model for integrating neuronal activity, EEG, fMRI and metabolism , 2008, NeuroImage.

[154]  D. Selkoe,et al.  Soluble Aβ Inhibits Specific Signal Transduction Cascades Common to the Insulin Receptor Pathway* , 2007, Journal of Biological Chemistry.

[155]  C. Masters,et al.  Mitochondrial Oxidative Stress Causes Hyperphosphorylation of Tau , 2007, PloS one.

[156]  C. Geula,et al.  Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease , 2007, Nature Medicine.

[157]  J. Hay Faculty Opinions recommendation of Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. , 2007 .

[158]  M. Simons,et al.  Neuron-glia communication in the control of oligodendrocyte function and myelin biogenesis , 2006, Journal of Cell Science.

[159]  D. Butterfield,et al.  Protein oxidation and lipid peroxidation in brain of subjects with Alzheimer's disease: insights into mechanism of neurodegeneration from redox proteomics. , 2006, Antioxidants & redox signaling.

[160]  G. McKhann,et al.  Spinal cord endoplasmic reticulum stress associated with a microsomal accumulation of mutant superoxide dismutase-1 in an ALS model. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[161]  Ralph A. Nixon,et al.  Extensive Involvement of Autophagy in Alzheimer Disease: An Immuno-Electron Microscopy Study , 2005, Journal of neuropathology and experimental neurology.

[162]  T. Gillingwater,et al.  A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. , 2004, American journal of human genetics.

[163]  R. Nussbaum,et al.  Hereditary Early-Onset Parkinson's Disease Caused by Mutations in PINK1 , 2004, Science.

[164]  D. Bennett,et al.  Diabetes mellitus and risk of Alzheimer disease and decline in cognitive function. , 2004, Archives of neurology.

[165]  D. Calne,et al.  Presynaptic mechanisms of motor fluctuations in Parkinson's disease: a probabilistic model. , 2004, Brain : a journal of neurology.

[166]  Colin L. Masters,et al.  Neurodegenerative diseases and oxidative stress , 2004, Nature Reviews Drug Discovery.

[167]  Ronald C Petersen,et al.  Increased risk of type 2 diabetes in Alzheimer disease. , 2004, Diabetes.

[168]  Nobutaka Hattori,et al.  Parkin and Endoplasmic Reticulum Stress , 2003, Annals of the New York Academy of Sciences.

[169]  R. Nitsch,et al.  Proteasome inhibition by paired helical filament‐tau in brains of patients with Alzheimer's disease , 2003, Journal of neurochemistry.

[170]  Patrik Brundin,et al.  Pathogenesis of parkinson's disease: dopamine, vesicles and α-synuclein , 2002, Nature Reviews Neuroscience.

[171]  L. Launer,et al.  Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: The Honolulu-Asia Aging Study. , 2002, Diabetes.

[172]  H. Kitano Systems Biology: A Brief Overview , 2002, Science.

[173]  Patrick R. Hof,et al.  Tau protein isoforms, phosphorylation and role in neurodegenerative disorders 1 1 These authors contributed equally to this work. , 2000, Brain Research Reviews.

[174]  W. Markesbery,et al.  Decreased levels of proteasome activity and proteasome expression in aging spinal cord , 2000, Neuroscience.

[175]  J M Fuster,et al.  Dopamine-mediated stabilization of delay-period activity in a network model of prefrontal cortex. , 2000, Journal of neurophysiology.

[176]  A. Ott Risk of dementia: The Rotterdam Study , 1997 .

[177]  M. Hines,et al.  The NEURON Simulation Environment , 1997, Neural Computation.

[178]  G. Brown,et al.  Cellular energy utilization and molecular origin of standard metabolic rate in mammals. , 1997, Physiological reviews.

[179]  B. Juurlink,et al.  Low Glutathione and High Iron Govern the Susceptibility of Oligodendroglial Precursors to Oxidative Stress , 1996, Journal of neurochemistry.

[180]  A. Goldberg,et al.  Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class I molecules , 1994, Cell.

[181]  V. Meininger,et al.  A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group. , 1994, The New England journal of medicine.

[182]  Barry Halliwell,et al.  Reactive Oxygen Species and the Central Nervous System , 1992, Journal of neurochemistry.

[183]  B. Halliwell,et al.  Free radicals in biology and medicine , 1985 .

[184]  L. Wetterberg,et al.  Evidence for the presence of specific receptors for insulin-like growth factors 1 (IGF-1) and 2 (IGF-2) and insulin throughout the adult human brain , 1982, Neuroscience Letters.

[185]  M. Savageau Biochemical systems analysis. II. The steady-state solutions for an n-pool system using a power-law approximation. , 1969, Journal of theoretical biology.

[186]  R. Klausen,et al.  Selective , 2020, Encyclopedia of the UN Sustainable Development Goals.

[187]  C. Hetz,et al.  ER Stress and Neurodegenerative Disease: A Cause or Effect Relationship? , 2018, Current topics in microbiology and immunology.

[188]  Christopher G Schwarz,et al.  Widespread brain tau and its association with ageing, Braak stage and Alzheimer's dementia. , 2018, Brain : a journal of neurology.

[189]  J. Götz,et al.  Extracellular Vesicles Containing P301L Mutant Tau Accelerate Pathological Tau Phosphorylation and Oligomer Formation but Do Not Seed Mature Neurofibrillary Tangles in ALZ17 Mice. , 2016, Journal of Alzheimer's disease : JAD.

[190]  Alexandra K. Diem,et al.  Data for paper "A simulation model of periarterial clearance of amyloid-beta from the brain" , 2016 .

[191]  H. Geerts,et al.  Phenotypic screening of the Prestwick library for treatment of Parkinson's tremor symptoms using a humanized quantitative systems pharmacology platform. , 2013, Journal of Parkinson's disease.

[192]  G. Pandini,et al.  Insulin has multiple antiamyloidogenic effects on human neuronal cells. , 2013, Endocrinology.

[193]  V. Borutaite,et al.  There is no evidence that mitochondria are the main source of reactive oxygen species in mammalian cells. , 2012, Mitochondrion.

[194]  A. Ludolph,et al.  Amyotrophic lateral sclerosis. , 2012, Current opinion in neurology.

[195]  R. Castellani,et al.  Alzheimer disease. , 2010, Disease-a-month : DM.

[196]  J. Hardy,et al.  The Amyloid Hypothesis of Alzheimer ’ s Disease : Progress and Problems on the Road to Therapeutics , 2009 .

[197]  R. Mrak Alzheimer-type neuropathological changes in morbidly obese elderly individuals. , 2009, Clinical neuropathology.

[198]  V. Perry,et al.  What is immune privilege (not)? , 2007, Trends in immunology.

[199]  J. Wands,et al.  Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease--is this type 3 diabetes? , 2005, Journal of Alzheimer's disease : JAD.

[200]  Patrik Brundin,et al.  Pathogenesis of Parkinson's disease: dopamine, vesicles and alpha-synuclein. , 2002, Nature reviews. Neuroscience.

[201]  Edinburgh Research Explorer Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution , 2022 .