Combined metabolic activators improve cognitive functions in Alzheimer’s disease patients: a randomised, double-blinded, placebo-controlled phase-II trial

[1]  J. Borén,et al.  Combined metabolic activators improve metabolic functions in the animal models of neurodegenerative diseases. , 2022, Life sciences.

[2]  M. Vyhnálek,et al.  Compromised autophagy and mitophagy in brain ageing and Alzheimer’s diseases , 2022, Aging brain.

[3]  G. Landreth,et al.  The niacin receptor HCAR2 modulates microglial response and limits disease progression in a mouse model of Alzheimer’s disease , 2022, Science Translational Medicine.

[4]  K. Caldwell,et al.  Amelioration of Alzheimer’s disease pathology by mitophagy inducers identified via machine learning and a cross-species workflow , 2022, Nature Biomedical Engineering.

[5]  J. Nielsen,et al.  Combined metabolic activators therapy ameliorates liver fat in nonalcoholic fatty liver disease patients , 2021, Molecular systems biology.

[6]  M. Mattson,et al.  NAD+ supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS–STING , 2021, Proceedings of the National Academy of Sciences.

[7]  J. Nielsen,et al.  Combined Metabolic Activators Accelerates Recovery in Mild‐to‐Moderate COVID‐19 , 2021, Advanced science.

[8]  J. Nielsen,et al.  Combined Metabolic Activators Reduces Liver Fat in Nonalcoholic Fatty Liver Disease Patients , 2021, medRxiv.

[9]  Wenzhe Li,et al.  The significance of sialylation on the pathogenesis of Alzheimer’s disease , 2021, Brain Research Bulletin.

[10]  N. Kurbatova,et al.  Urinary metabolic phenotyping for Alzheimer’s disease , 2020, Scientific Reports.

[11]  P. Wong,et al.  Metabolic Profiling of Female Tg2576 Mouse Brains Provides Novel Evidence Supporting Intranasal Low-Dose Pioglitazone for Long-Term Treatment at an Early Stage of Alzheimer’s Disease , 2020, Biomedicines.

[12]  A. Nunomura,et al.  RNA and Oxidative Stress in Alzheimer's Disease: Focus on microRNAs , 2020, Oxidative medicine and cellular longevity.

[13]  Angus M. Brown,et al.  Universal Glia to Neurone Lactate Transfer in the Nervous System: Physiological Functions and Pathological Consequences , 2020, Biosensors.

[14]  J. Błaszczyk Energy Metabolism Decline in the Aging Brain—Pathogenesis of Neurodegenerative Disorders , 2020, Metabolites.

[15]  S. Chojnowska,et al.  Preventive Role of L-Carnitine and Balanced Diet in Alzheimer’s Disease , 2020, Nutrients.

[16]  Dana E. Feldman,et al.  Brain Network Segregation and Glucose Energy Utilization: Relevance for Age-Related Differences in Cognitive Function. , 2020, Cerebral cortex.

[17]  A. Fonteh,et al.  Involvement of Lipids in Alzheimer’s Disease Pathology and Potential Therapies , 2020, Frontiers in Physiology.

[18]  Wenzhang Wang,et al.  Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances , 2020, Molecular Neurodegeneration.

[19]  J. Nielsen,et al.  A systems biology approach for studying neurodegenerative diseases. , 2020, Drug discovery today.

[20]  H. Chui,et al.  Urine dicarboxylic acids change in pre-symptomatic Alzheimer’s disease and reflect loss of energy capacity and hippocampal volume , 2020, PloS one.

[21]  J. Nielsen,et al.  The acute effect of metabolic cofactor supplementation: a potential therapeutic strategy against non‐alcoholic fatty liver disease , 2020, Molecular systems biology.

[22]  M. Sabbagh,et al.  Alzheimer’s Disease Drug Development Pipeline 2020 , 2020, The Journal of Prevention of Alzheimer's Disease.

[23]  Michel E. Vandenberghe,et al.  Impairment of Glycolysis-Derived L-Serine Production in Astrocytes Contributes to Cognitive Deficits in Alzheimer's Disease , 2019, SSRN Electronic Journal.

[24]  A. Bryll,et al.  Oxidative-Antioxidant Imbalance and Impaired Glucose Metabolism in Schizophrenia , 2020, Biomolecules.

[25]  B. Kuehn In Alzheimer Research, Glucose Metabolism Moves to Center Stage. , 2020, JAMA.

[26]  H. Braak,et al.  Longitudinal brain atrophy distribution in advanced Parkinson's disease: What makes the difference in “cognitive status” converters? , 2019, Human brain mapping.

[27]  M. M. Gromiha,et al.  Investigating the energy crisis in Alzheimer disease using transcriptome study , 2019, Scientific Reports.

[28]  M. Delorenzi,et al.  Systemic and central nervous system metabolic alterations in Alzheimer’s disease , 2019, Alzheimer's Research & Therapy.

[29]  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 , 2019, bioRxiv.

[30]  M. Mattson,et al.  NAD+ in Brain Aging and Neurodegenerative Disorders. , 2019, Cell metabolism.

[31]  D. Perani,et al.  TAM Receptor Pathways at the Crossroads of Neuroinflammation and Neurodegeneration , 2019, Disease markers.

[32]  D. Ryu,et al.  Plasma Klotho concentration is associated with the presence, burden and progression of cerebral small vessel disease in patients with acute ischaemic stroke , 2019, PloS one.

[33]  G. Brewer,et al.  Global Metabolic Shifts in Age and Alzheimer’s Disease Mouse Brains Pivot at NAD+/NADH Redox Sites , 2019, Journal of Alzheimer's disease : JAD.

[34]  J. Borén,et al.  The Potential Use of Metabolic Cofactors in Treatment of NAFLD , 2019, Nutrients.

[35]  L. Hood,et al.  iNetModels 2.0: an interactive visualization and database of multi-omics data , 2019, bioRxiv.

[36]  R. Taliyan,et al.  Therapeutic approaches to Alzheimer's type of dementia: A focus on FGF21 mediated neuroprotection. , 2019, Current pharmaceutical design.

[37]  Ying Han,et al.  Trajectories of the Hippocampal Subfields Atrophy in the Alzheimer’s Disease: A Structural Imaging Study , 2019, Front. Neuroinform..

[38]  R. Swerdlow,et al.  Mitochondrial dysfunction in Alzheimer's disease: Role in pathogenesis and novel therapeutic opportunities , 2019, British journal of pharmacology.

[39]  M. Z. Cader,et al.  Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease , 2019, Nature Neuroscience.

[40]  E. Mazzon,et al.  Overview on the Effects of N-Acetylcysteine in Neurodegenerative Diseases , 2018, Molecules.

[41]  M. Herrero,et al.  Effect of NAC treatment and physical activity on neuroinflammation in subchronic Parkinsonism; is physical activity essential? , 2018, Journal of Neuroinflammation.

[42]  Yong Shen,et al.  A Novel Perspective Linkage Between Kidney Function and Alzheimer’s Disease , 2018, Front. Cell. Neurosci..

[43]  S. Heckers,et al.  Regionally specific volume deficits along the hippocampal long axis in early and chronic psychosis , 2018, NeuroImage: Clinical.

[44]  W. Jagust Imaging the evolution and pathophysiology of Alzheimer disease , 2018, Nature Reviews Neuroscience.

[45]  C. Forsblom,et al.  Metabolomic Profile Predicts Development of Microalbuminuria in Individuals with Type 1 Diabetes , 2018, Scientific Reports.

[46]  G. Sancesario,et al.  Alzheimer's disease in the omics era. , 2018, Clinical biochemistry.

[47]  N. Resnick,et al.  Functional connectivity of the brain in older women with urgency urinary incontinence , 2018, Neurourology and urodynamics.

[48]  F. Panza,et al.  Klotho at the Edge of Alzheimer’s Disease and Senile Depression , 2018, Molecular Neurobiology.

[49]  S. Makin The amyloid hypothesis on trial , 2018, Nature.

[50]  A. Whitworth,et al.  The NAD+ Precursor Nicotinamide Riboside Rescues Mitochondrial Defects and Neuronal Loss in iPSC and Fly Models of Parkinson's Disease. , 2018, Cell reports.

[51]  A. Passaro,et al.  Lower Plasma Klotho Concentrations Are Associated with Vascular Dementia but Not Late-Onset Alzheimer’s Disease , 2018, Gerontology.

[52]  J. Nielsen,et al.  Systems biology in hepatology: approaches and applications , 2018, Nature Reviews Gastroenterology & Hepatology.

[53]  Tiangang Li,et al.  Bile acids regulate cysteine catabolism and glutathione regeneration to modulate hepatic sensitivity to oxidative injury. , 2018, JCI insight.

[54]  Hee-Sun Kim,et al.  Sulforaphane rescues amyloid-β peptide-mediated decrease in MerTK expression through its anti-inflammatory effect in human THP-1 macrophages , 2018, Journal of Neuroinflammation.

[55]  Jens Nielsen,et al.  An Integrated Understanding of the Rapid Metabolic Benefits of a Carbohydrate-Restricted Diet on Hepatic Steatosis in Humans. , 2018, Cell metabolism.

[56]  J. Wiśniewski,et al.  Aging‐associated changes in hippocampal glycogen metabolism in mice. Evidence for and against astrocyte‐to‐neuron lactate shuttle , 2018, Glia.

[57]  M. Mattson,et al.  NAD+ supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency , 2018, Proceedings of the National Academy of Sciences.

[58]  Steven E. Arnold,et al.  Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums , 2018, Nature Reviews Neurology.

[59]  L. Vécsei,et al.  Mitochondria, Oxidative Stress and the Kynurenine System, with a Focus on Ageing and Neuroprotection , 2018, Molecules.

[60]  Qiong Yang,et al.  Association of amine biomarkers with incident dementia and Alzheimer's disease in the Framingham Study , 2017, Alzheimer's & Dementia.

[61]  J. Auwerx,et al.  Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity , 2017, Nature.

[62]  M. Mattson,et al.  NAD+ in Aging: Molecular Mechanisms and Translational Implications. , 2017, Trends in molecular medicine.

[63]  Michael W. Weiner,et al.  Metabolic network failures in Alzheimer's disease: A biochemical road map , 2017, Alzheimer's & Dementia.

[64]  A. Lusis,et al.  Multi-omics approaches to disease , 2017, Genome Biology.

[65]  F. Nicoletti,et al.  Vasorelaxing Action of the Kynurenine Metabolite, Xanthurenic Acid: The Missing Link in Endotoxin-Induced Hypotension? , 2017, Front. Pharmacol..

[66]  C. Rowe,et al.  Altered levels of blood proteins in Alzheimer's disease longitudinal study: Results from Australian Imaging Biomarkers Lifestyle Study of Ageing cohort , 2017, Alzheimer's & dementia.

[67]  Muke Zhou,et al.  L-carnitine for cognitive enhancement in people without cognitive impairment. , 2017, The Cochrane database of systematic reviews.

[68]  R. Palmer,et al.  δ scores predict mild cognitive impairment and Alzheimer's disease conversions from nondemented states , 2017, Alzheimer's & dementia.

[69]  M. Z. Cader,et al.  Mitophagy and Alzheimer’s Disease: Cellular and Molecular Mechanisms , 2017, Trends in Neurosciences.

[70]  J. Griffin,et al.  Amino Acid Catabolism in Alzheimer's Disease Brain: Friend or Foe? , 2017, Oxidative medicine and cellular longevity.

[71]  K. O'Farrell,et al.  Stress-related regulation of the kynurenine pathway: Relevance to neuropsychiatric and degenerative disorders , 2017, Neuropharmacology.

[72]  N. Câmara,et al.  Soluble Uric Acid Activates the NLRP3 Inflammasome , 2017, Scientific Reports.

[73]  E. Boerwinkle,et al.  Prospective associations of plasma phospholipids and mild cognitive impairment/dementia among African Americans in the ARIC Neurocognitive Study , 2016, Alzheimer's & dementia.

[74]  C. Humpel,et al.  Alpha-Smooth Muscle Actin mRNA and Protein Are Increased in Isolated Brain Vessel Extracts of Alzheimer Mice , 2016, Pharmacology.

[75]  F. Piras,et al.  Medium-chain plasma acylcarnitines, ketone levels, cognition, and gray matter volumes in healthy elderly, mildly cognitively impaired, or Alzheimer's disease subjects , 2016, Neurobiology of Aging.

[76]  M. Kraut,et al.  Blood metabolite markers of cognitive performance and brain function in aging , 2016, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[77]  G. Corso,et al.  Serum Levels of Acyl-Carnitines along the Continuum from Normal to Alzheimer's Dementia , 2016, PloS one.

[78]  E. Boerwinkle,et al.  Plasma phospholipids and prevalence of mild cognitive impairment and/or dementia in the ARIC Neurocognitive Study (ARIC-NCS) , 2016, Alzheimer's & dementia.

[79]  Heng Du,et al.  Deregulation of mitochondrial F1FO-ATP synthase via OSCP in Alzheimer’s disease , 2016, Nature Communications.

[80]  Bernadette McGuinness,et al.  Alzheimer's disease–like pathology has transient effects on the brain and blood metabolome , 2016, Neurobiology of Aging.

[81]  A. Salminen,et al.  Hypoxia and GABA shunt activation in the pathogenesis of Alzheimer's disease , 2016, Neurochemistry International.

[82]  M. Keller,et al.  Unbiased Metabolomic Investigation of Alzheimer's Disease Brain Points to Dysregulation of Mitochondrial Aspartate Metabolism. , 2016, Journal of Proteome Research.

[83]  N. Inestrosa,et al.  Is L-methionine a trigger factor for Alzheimer’s-like neurodegeneration?: Changes in Aβ oligomers, tau phosphorylation, synaptic proteins, Wnt signaling and behavioral impairment in wild-type mice , 2015, Molecular Neurodegeneration.

[84]  D. Bassett,et al.  Dynamic reconfiguration of frontal brain networks during executive cognition in humans , 2015, Proceedings of the National Academy of Sciences.

[85]  Y. Liu,et al.  Ribosylation triggering Alzheimer’s disease-like Tau hyperphosphorylation via activation of CaMKII , 2015, Aging cell.

[86]  M. Zou,et al.  Tryptophan-kynurenine pathway is dysregulated in inflammation, and immune activation. , 2015, Frontiers in bioscience.

[87]  Koenraad Van Leemput,et al.  A computational atlas of the hippocampal formation using ex vivo, ultra-high resolution MRI: Application to adaptive segmentation of in vivo MRI , 2015, NeuroImage.

[88]  C. Hölscher,et al.  Untargeted Metabolomic Analysis of Human Plasma Indicates Differentially Affected Polyamine and L-Arginine Metabolism in Mild Cognitive Impairment Subjects Converting to Alzheimer’s Disease , 2015, PloS one.

[89]  L. Mosca,et al.  PARP-1 involvement in neurodegeneration: A focus on Alzheimer’s and Parkinson’s diseases , 2015, Mechanisms of Ageing and Development.

[90]  W. Xu,et al.  Plasma metabolite profiles of Alzheimer's disease and mild cognitive impairment. , 2014, Journal of proteome research.

[91]  H. Soininen,et al.  Evidence of altered phosphatidylcholine metabolism in Alzheimer's disease , 2014, Neurobiology of Aging.

[92]  M. Uhlén,et al.  Genome-scale metabolic modelling of hepatocytes reveals serine deficiency in patients with non-alcoholic fatty liver disease , 2014, Nature Communications.

[93]  A. Alexandrov,et al.  N-acetylcysteine (NAC) in neurological disorders: mechanisms of action and therapeutic opportunities , 2014, Brain and behavior.

[94]  J. Moffett,et al.  N-Acetylaspartate reductions in brain injury: impact on post-injury neuroenergetics, lipid synthesis, and protein acetylation , 2013, Front. Neuroenergetics.

[95]  A. Gutiérrez,et al.  In vivo modification of Abeta plaque toxicity as a novel neuroprotective lithium-mediated therapy for Alzheimer’s disease pathology , 2013, Acta neuropathologica communications.

[96]  Zhichun Chen,et al.  Decoding Alzheimer's disease from perturbed cerebral glucose metabolism: Implications for diagnostic and therapeutic strategies , 2013, Progress in Neurobiology.

[97]  M. Mattson,et al.  Nicotinamide forestalls pathology and cognitive decline in Alzheimer mice: evidence for improved neuronal bioenergetics and autophagy procession , 2013, Neurobiology of Aging.

[98]  Gilles J. Guillemin,et al.  Expression of Tryptophan 2,3-Dioxygenase and Production of Kynurenine Pathway Metabolites in Triple Transgenic Mice and Human Alzheimer's Disease Brain , 2013, PloS one.

[99]  Mert R. Sabuncu,et al.  Statistical analysis of longitudinal neuroimage data with Linear Mixed Effects models , 2013, NeuroImage.

[100]  A. Castelnuovo,et al.  Effects of long-term treatment with pioglitazone on cognition and glucose metabolism of PS1-KI, 3xTg-AD, and wild-type mice , 2012, Cell Death and Disease.

[101]  Xinsheng Yao,et al.  Epidermal growth factor receptor is a preferred target for treating Amyloid-β–induced memory loss , 2012, Proceedings of the National Academy of Sciences.

[102]  Bruce Fischl,et al.  Within-subject template estimation for unbiased longitudinal image analysis , 2012, NeuroImage.

[103]  R. Nagele,et al.  Neuronal PAD4 expression and protein citrullination: possible role in production of autoantibodies associated with neurodegenerative disease. , 2012, Journal of autoimmunity.

[104]  P. Dutar,et al.  Reversal of age‐related oxidative stress prevents hippocampal synaptic plasticity deficits by protecting d‐serine‐dependent NMDA receptor activation , 2012, Aging cell.

[105]  Vidhi Singh,et al.  Meta-Analysis of the Efficacy of Adjunctive NMDA Receptor Modulators in Chronic Schizophrenia , 2011, CNS drugs.

[106]  Xianlin Han,et al.  Metabolomics in Early Alzheimer's Disease: Identification of Altered Plasma Sphingolipidome Using Shotgun Lipidomics , 2011, PloS one.

[107]  Shunichiro Shinagawa,et al.  Association between executive dysfunction and hippocampal volume in Alzheimer's disease , 2010, International Psychogeriatrics.

[108]  K. A. Wollen,et al.  Alzheimer's disease: the pros and cons of pharmaceutical, nutritional, botanical, and stimulatory therapies, with a discussion of treatment strategies from the perspective of patients and practitioners. , 2010, Alternative medicine review : a journal of clinical therapeutic.

[109]  R. Killiany,et al.  Subregions of the inferior parietal lobule are affected in the progression to Alzheimer's disease , 2010, Neurobiology of Aging.

[110]  Q. Garrett,et al.  Role of carnitine in disease , 2010, Nutrition & metabolism.

[111]  Y. Zhong,et al.  PI3 kinase signaling is involved in Aβ-induced memory loss in Drosophila , 2010, Proceedings of the National Academy of Sciences.

[112]  G. Guillemin,et al.  Kynurenine Pathway Metabolites in Humans: Disease and Healthy States , 2009, International journal of tryptophan research : IJTR.

[113]  Winnie S. Liang,et al.  Alzheimer's disease is associated with reduced expression of energy metabolism genes in posterior cingulate neurons , 2008, Proceedings of the National Academy of Sciences.

[114]  B. Brew,et al.  The involvement of astrocytes and kynurenine pathway in Alzheimer’s disease , 2007, Neurotoxicity Research.

[115]  T. Kudo,et al.  Hematopoietic Prostaglandin D Synthase and DP1 Receptor Are Selectively Upregulated in Microglia and Astrocytes Within Senile Plaques From Human Patients and in a Mouse Model of Alzheimer Disease , 2007, Journal of neuropathology and experimental neurology.

[116]  M. Beal,et al.  Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases , 2006, Nature.

[117]  T. Olsson,et al.  Neuroactive steroid effects on cognitive functions with a focus on the serotonin and GABA systems , 2006, Brain Research Reviews.

[118]  Xue-wen Chen,et al.  High intrinsic oxidative stress may underlie selective vulnerability of the hippocampal CA1 region. , 2005, Brain research. Molecular brain research.

[119]  Yu Wang,et al.  A biochemical and functional characterization of diet‐induced brain insulin resistance , 2005, Journal of neurochemistry.

[120]  E. Otomo,et al.  Association of a polymorphism of the transforming growth factor-β1 gene with cerebral amyloid angiopathy , 2005, Journal of Neurology, Neurosurgery & Psychiatry.

[121]  K. Nave,et al.  A hematopoietic growth factor, thrombopoietin, has a proapoptotic role in the brain. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[122]  D. Lahiri,et al.  Role of the APP Promoter in Alzheimer's Disease: Cell Type‐Specific Expression of the β‐Amyloid Precursor Protein , 2004, Annals of the New York Academy of Sciences.

[123]  B. Brew,et al.  A&bgr;1-42 induces production of quinolinic acid by human macrophages and microglia , 2003, Neuroreport.

[124]  Anders Wallin,et al.  Increased intrathecal levels of the angiogenic factors VEGF and TGF-β in Alzheimer’s disease and vascular dementia , 2002, Neurobiology of Aging.

[125]  Sudha Seshadri,et al.  Plasma Homocysteine as a Risk Factor for Dementia and Alzheimer's Disease , 2002 .

[126]  A. Dale,et al.  Whole Brain Segmentation Automated Labeling of Neuroanatomical Structures in the Human Brain , 2002, Neuron.

[127]  M Schulzer,et al.  Expectation and Dopamine Release: Mechanism of the Placebo Effect in Parkinson's Disease , 2001, Science.

[128]  A. Convit,et al.  Hippocampal formation glucose metabolism and volume losses in MCI and AD , 2001, Neurobiology of Aging.

[129]  S. DeKosky,et al.  Analysis of genetic polymorphisms in the transforming growth factor-β1 gene and the risk of Alzheimer's disease , 2000, Human Genetics.

[130]  Anders M. Dale,et al.  Cortical Surface-Based Analysis I. Segmentation and Surface Reconstruction , 1999, NeuroImage.

[131]  P. Heinrich,et al.  Oncostatin M and the Interleukin-6 and Soluble Interleukin-6 Receptor Complex Regulate α1-Antichymotrypsin Expression in Human Cortical Astrocytes* , 1998, The Journal of Biological Chemistry.

[132]  T. Owens,et al.  Inflammatory cytokines in the brain: does the CNS shape immune responses? , 1994, Immunology today.

[133]  P. Peterson,et al.  Serum cytokine levels in patients with Alzheimer's disease. , 1994, Clinical and diagnostic laboratory immunology.

[134]  G Rotilio,et al.  Saturated dicarboxylic acids as products of unsaturated fatty acid oxidation. , 1993, Biochimica et biophysica acta.

[135]  D. Selkoe The molecular pathology of Alzheimer's disease , 1991, Neuron.

[136]  P. Heinrich,et al.  Interleukin‐6 is the major regulator of acute phase protein synthesis in adult human hepatocytes , 1989, FEBS letters.

[137]  D. Selkoe,et al.  Immunochemical identification of the serine protease inhibitor α 1-antichymotrypsin in the brain amyloid deposits of Alzheimer's disease , 1988, Cell.

[138]  H. Fillit,et al.  Evaluation of the Neuroprotective Potential of N-Acetylcysteine for Prevention and Treatment of Cognitive Aging and Dementia. , 2017, The journal of prevention of Alzheimer's disease.

[139]  C. Humpel,et al.  Targeted Metabolomic Analysis of Soluble Lysates from Platelets of Patients with Mild Cognitive Impairment and Alzheimer's Disease Compared to Healthy Controls: Is PC aeC40:4 a Promising Diagnostic Tool? , 2017, Journal of Alzheimer's disease : JAD.

[140]  A. Fuso,et al.  Bioenergetic Impairment in Animal and Cellular Models of Alzheimer's Disease: PARP-1 Inhibition Rescues Metabolic Dysfunctions. , 2016, Journal of Alzheimer's disease : JAD.

[141]  L. Kotra,et al.  Peptidyl Arginine Deiminases and Neurodegenerative Diseases. , 2016, Current Medicinal Chemistry.

[142]  K. Blennow,et al.  Increased Cerebrospinal Fluid Level of Insulin-like Growth Factor-II in Male Patients with Alzheimer's Disease. , 2015, Journal of Alzheimer's disease : JAD.

[143]  S. Lidstone Great expectations: the placebo effect in Parkinson's disease. , 2014, Handbook of experimental pharmacology.

[144]  Jon Neville,et al.  Understanding placebo responses in Alzheimer's disease clinical trials from the literature meta-data and CAMD database. , 2013, Journal of Alzheimer's disease : JAD.

[145]  S. Gavrilova,et al.  [Acetyl-L-carnitine (carnicetine) in the treatment of early stages of Alzheimer's disease and vascular dementia]. , 2011, Zhurnal nevrologii i psikhiatrii imeni S.S. Korsakova.

[146]  C. Lyketsos,et al.  Plasma sphingomyelins are associated with cognitive progression in Alzheimer's disease. , 2011, Journal of Alzheimer's disease : JAD.

[147]  S. DeKosky,et al.  Is the urea cycle involved in Alzheimer's disease? , 2010, Journal of Alzheimer's Disease.

[148]  E. Mackenzie,et al.  Transforming growth factor-beta 1 potentiates amyloid-beta generation in astrocytes and in transgenic mice. , 2003, The Journal of biological chemistry.

[149]  B. Liang,et al.  Transforming growth factor-beta-induced transcription of the Alzheimer beta-amyloid precursor protein gene involves interaction between the CTCF-complex and Smads. , 2002, Biochemical and biophysical research communications.