Emerging Links between Nonalcoholic Fatty Liver Disease and Neurodegeneration

The association between liver and brain health has gained attention as biomarkers of liver function have been revealed to predict neurodegeneration. The liver is a central regulator in metabolic homeostasis. However, in nonalcoholic fatty liver disease (NAFLD), homeostasis is disrupted which can result in extrahepatic organ pathologies. Emerging literature provides insight into the mechanisms behind the liver-brain health axis. These include the increased production of liver-derived factors that promote insulin resistance and loss of neuroprotective factors under conditions of NAFLD that increase insulin resistance in the central nervous system. In addition, elevated proinflammatory cytokines linked to NAFLD negatively impact the blood-brain barrier and increase neuroinflammation. Furthermore, exacerbated dyslipidemia associated with NAFLD and hepatic dysfunction can promote altered brain bioenergetics and oxidative stress. In this review, we summarize the current knowledge of the crosstalk between liver and brain as it relates to the pathophysiology between NAFLD and neurodegeneration, with an emphasis on Alzheimer's disease. We also highlight knowledge gaps and future areas for investigation to strengthen the potential link between NAFLD and neurodegeneration.

[1]  Zhiyong Zhou,et al.  Non‐alcoholic fatty liver disease and the risk of dementia , 2022, Liver international (Print).

[2]  R. Swerdlow,et al.  Mitochondrial function and Aβ in Alzheimer's disease postmortem brain , 2022, Neurobiology of Disease.

[3]  Sang Min Park,et al.  Association of non-alcoholic fatty liver disease with incident dementia later in life among elder adults , 2022, Clinical and molecular hepatology.

[4]  Elena Simona Micu,et al.  Systemic and adipose tissue inflammation in NASH: correlations with histopathological aspects , 2021, Romanian journal of morphology and embryology = Revue roumaine de morphologie et embryologie.

[5]  L. Velloso,et al.  Hippocampal Function Is Impaired by a Short-Term High-Fat Diet in Mice: Increased Blood–Brain Barrier Permeability and Neuroinflammation as Triggering Events , 2021, Frontiers in Neuroscience.

[6]  E. D. Khilazheva,et al.  NLRP3 Inflammasome Blocking as a Potential Treatment of Central Insulin Resistance in Early-Stage Alzheimer’s Disease , 2021, International journal of molecular sciences.

[7]  B. Honoré,et al.  Shotgun-based proteomics of extracellular vesicles in Alzheimer’s disease reveals biomarkers involved in immunological and coagulation pathways , 2021, Scientific Reports.

[8]  P. Iozzo,et al.  Brain-gut-liver interactions across the spectrum of insulin resistance in metabolic fatty liver disease , 2021, World journal of gastroenterology.

[9]  R. Goyal,et al.  Molecular and pathobiological involvement of fetuin-A in the pathogenesis of NAFLD , 2021, Inflammopharmacology.

[10]  B. Železná,et al.  Aging and high-fat diet feeding lead to peripheral insulin resistance and sex-dependent changes in brain of mouse model of tau pathology THY-Tau22 , 2021, Journal of Neuroinflammation.

[11]  J. Ali,et al.  Linagliptin, a DPP-4 inhibitor, ameliorates Aβ (1−42) peptides induced neurodegeneration and brain insulin resistance (BIR) via insulin receptor substrate-1 (IRS-1) in rat model of Alzheimer's disease , 2021, Neuropharmacology.

[12]  F. Crews,et al.  TRAIL Mediates Neuronal Death in AUD: A Link between Neuroinflammation and Neurodegeneration , 2021, International journal of molecular sciences.

[13]  H. Vilstrup,et al.  Cognitive Dysfunction in Non-Alcoholic Fatty Liver Disease—Current Knowledge, Mechanisms and Perspectives , 2021, Journal of clinical medicine.

[14]  F. Fu,et al.  Escin ameliorates the impairments of neurological function and blood brain barrier by inhibiting systemic inflammation in intracerebral hemorrhagic mice , 2020, Experimental Neurology.

[15]  R. Hultcrantz,et al.  Non-alcoholic fatty liver disease does not increase dementia risk although histology data might improve risk prediction , 2020, JHEP reports : innovation in hepatology.

[16]  Divya P Kumar,et al.  Extracellular Vesicles as Inflammatory Drivers in NAFLD , 2021, Frontiers in Immunology.

[17]  C. Glass,et al.  Epigenetic Regulation of Kupffer Cell Function in Health and Disease , 2021, Frontiers in Immunology.

[18]  P. Gual,et al.  Chronic Inflammation in Non-Alcoholic Steatohepatitis: Molecular Mechanisms and Therapeutic Strategies , 2020, Frontiers in Endocrinology.

[19]  M. Calzada,et al.  Time-dependent dual effect of NLRP3 inflammasome in brain ischemia , 2020, bioRxiv.

[20]  M. Selenica,et al.  TDP-43 mediated blood-brain barrier permeability and leukocyte infiltration promote neurodegeneration in a low-grade systemic inflammation mouse model , 2020, Journal of Neuroinflammation.

[21]  M. Selenica,et al.  TDP-43 mediated blood-brain barrier permeability and leukocyte infiltration promote neurodegeneration in a low-grade systemic inflammation mouse model , 2020, Journal of neuroinflammation.

[22]  A. D. de Bem,et al.  High Cholesterol Diet Exacerbates Blood-Brain Barrier Disruption in LDLr–/– Mice: Impact on Cognitive Function , 2020, Journal of Alzheimer's disease : JAD.

[23]  E. S. Abdel-Reheim,et al.  Evaluation of insulin resistance induced brain tissue dysfunction in obese dams and their neonates: Role of ipriflavone amelioration. , 2020, Combinatorial chemistry & high throughput screening.

[24]  D. Burrin,et al.  Neurodegeneration in juvenile Iberian pigs with diet-induced non-alcoholic fatty liver disease. , 2020, American journal of physiology. Endocrinology and metabolism.

[25]  Shu Zheng,et al.  Application of exosomes as liquid biopsy in clinical diagnosis , 2020, Signal Transduction and Targeted Therapy.

[26]  M. Nagarkatti,et al.  Lipocalin 2 induces neuroinflammation and blood-brain barrier dysfunction through liver-brain axis in murine model of nonalcoholic steatohepatitis , 2020, Journal of Neuroinflammation.

[27]  S. Caprio,et al.  Intrahepatic fat, irrespective of ethnicity, is associated with reduced endogenous insulin clearance and hepatic insulin resistance in obese youths: a cross-sectional and longitudinal study from the Yale Pediatric NAFLD cohort. , 2020, Diabetes, obesity & metabolism.

[28]  R. Karbalaei,et al.  Showing NAFLD, as a key connector disease between Alzheimer’s disease and diabetes via analysis of systems biology , 2020, Gastroenterology and hepatology from bed to bench.

[29]  Y. Tachibana,et al.  Dual microglia effects on blood brain barrier permeability induced by systemic inflammation , 2019, Nature Communications.

[30]  R. Loomba,et al.  The 20% Rule of NASH Progression: The Natural History of Advanced Fibrosis and Cirrhosis Caused by NASH , 2019, Hepatology.

[31]  Jonathan D. Smith,et al.  HDL Flux is Higher in Patients with Nonalcoholic Fatty Liver Disease. , 2019, American journal of physiology. Endocrinology and metabolism.

[32]  M. Watt,et al.  The liver as an endocrine organ - linking NAFLD and insulin resistance. , 2019, Endocrine reviews.

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

[34]  A. Beiser,et al.  NON-ALCOHOLIC FATTY LIVER DISEASE, LIVER FIBROSIS SCORE AND COGNITIVE FUNCTION IN MIDDLE-AGED ADULTS: THE FRAMINGHAM STUDY , 2019, Alzheimer's & Dementia.

[35]  Xianlin Han,et al.  Association of Altered Liver Enzymes With Alzheimer Disease Diagnosis, Cognition, Neuroimaging Measures, and Cerebrospinal Fluid Biomarkers , 2019, JAMA network open.

[36]  B. Miller,et al.  Association of Early-Onset Alzheimer Disease With Elevated Low-density Lipoprotein Cholesterol Levels and Rare Genetic Coding Variants of APOB. , 2019, JAMA neurology.

[37]  F. Lesage,et al.  Non-Alcoholic Fatty Liver Disease, and the Underlying Altered Fatty Acid Metabolism, Reveals Brain Hypoperfusion and Contributes to the Cognitive Decline in APP/PS1 Mice , 2019, Metabolites.

[38]  R. Takahashi,et al.  The Dipeptidyl Peptidase-4 Inhibitor Linagliptin Ameliorates High-fat Induced Cognitive Decline in Tauopathy Model Mice , 2019, International journal of molecular sciences.

[39]  Silvia Maioli,et al.  Alterations in cholesterol metabolism as a risk factor for developing Alzheimer’s disease: Potential novel targets for treatment , 2019, The Journal of Steroid Biochemistry and Molecular Biology.

[40]  T. Mocan,et al.  Extracellular Vesicles in Liver Diseases: Diagnostic, Prognostic, and Therapeutic Application , 2019, Seminars in Liver Disease.

[41]  Nicola J. Ray,et al.  Neuroinflammation in mild cognitive impairment and Alzheimer’s disease: A meta-analysis , 2019, Ageing Research Reviews.

[42]  D. Belsham,et al.  Tumour necrosis factor α induces neuroinflammation and insulin resistance in immortalised hypothalamic neurones through independent pathways , 2019, Journal of neuroendocrinology.

[43]  G. Svegliati-Baroni,et al.  Nonalcoholic Fatty Liver Disease: Basic Pathogenetic Mechanisms in the Progression From NAFLD to NASH , 2019, Transplantation.

[44]  F. Cubero,et al.  Extracellular vesicles in liver disease and beyond , 2018, World journal of gastroenterology.

[45]  M. Manns,et al.  Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016-2030. , 2018, Journal of hepatology.

[46]  K. Daffner,et al.  Dementia. , 2018, The American journal of medicine.

[47]  H. Reeves,et al.  Liquid biopsy for liver diseases , 2018, Gut.

[48]  E. Yang,et al.  Relationship between Liver Pathology and Disease Progression in a Murine Model of Amyotrophic Lateral Sclerosis , 2018, Neurodegenerative Diseases.

[49]  V. Felipo,et al.  Histological Features of Cerebellar Neuropathology in Patients With Alcoholic and Nonalcoholic Steatohepatitis , 2018, Journal of neuropathology and experimental neurology.

[50]  Ming Lu,et al.  Inhibition of the hepatic Nlrp3 protects dopaminergic neurons via attenuating systemic inflammation in a MPTP/p mouse model of Parkinson’s disease , 2018, Journal of Neuroinflammation.

[51]  Z. Niu,et al.  Serum retinol binding protein 4 and galectin-3 binding protein as novel markers for postmenopausal nonalcoholic fatty liver disease. , 2018, Clinical biochemistry.

[52]  M. Altamimi,et al.  Could Autism Be Associated With Nutritional Status in the Palestinian population? The Outcomes of the Palestinian Micronutrient Survey , 2018, Nutrition and metabolic insights.

[53]  A. Feldstein,et al.  Triggering and resolution of inflammation in NASH , 2018, Nature Reviews Gastroenterology & Hepatology.

[54]  F. Schick,et al.  The hepatokines fetuin-A and fetuin-B are upregulated in the state of hepatic steatosis and may differently impact on glucose homeostasis in humans. , 2018, American journal of physiology. Endocrinology and metabolism.

[55]  W. Freeman,et al.  Insulin-like growth factor receptor signaling regulates working memory, mitochondrial metabolism, and amyloid-β uptake in astrocytes , 2018, Molecular metabolism.

[56]  S. Craft,et al.  Triglycerides cross the blood–brain barrier and induce central leptin and insulin receptor resistance , 2017, International Journal of Obesity.

[57]  R. Vasan,et al.  Association of Nonalcoholic Fatty Liver Disease With Lower Brain Volume in Healthy Middle-aged Adults in the Framingham Study , 2018, JAMA neurology.

[58]  A. Holmgren,et al.  Redox Signaling Mediated by Thioredoxin and Glutathione Systems in the Central Nervous System. , 2017, Antioxidants & redox signaling.

[59]  N. Chattipakorn,et al.  SGLT2‐inhibitor and DPP‐4 inhibitor improve brain function via attenuating mitochondrial dysfunction, insulin resistance, inflammation, and apoptosis in HFD‐induced obese rats , 2017, Toxicology and applied pharmacology.

[60]  Miguel López,et al.  Brain Ceramide Metabolism in the Control of Energy Balance , 2017, Front. Physiol..

[61]  G. Reiser,et al.  Brain energy metabolism spurns fatty acids as fuel due to their inherent mitotoxicity and potential capacity to unleash neurodegeneration , 2017, Neurochemistry International.

[62]  S. Mukhopadhyay,et al.  Palmitate induced Fetuin-A secretion from pancreatic β-cells adversely affects its function and elicits inflammation. , 2017, Biochemical and biophysical research communications.

[63]  N. Stefan,et al.  Elevated hepatic DPP4 activity promotes insulin resistance and non-alcoholic fatty liver disease , 2017, Molecular Metabolism.

[64]  G. Gores,et al.  TRAIL deletion prevents liver inflammation but not adipose tissue inflammation during murine diet‐induced obesity , 2017, Hepatology communications.

[65]  M. Watt,et al.  Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance , 2017, Nature Reviews Endocrinology.

[66]  S. Friedman,et al.  Mechanisms of hepatic stellate cell activation , 2017, Nature Reviews Gastroenterology &Hepatology.

[67]  W. Ling,et al.  Retinol Binding Protein-4 Levels and Non-alcoholic Fatty Liver Disease: A community-based cross-sectional study , 2017, Scientific Reports.

[68]  Jun Li,et al.  Emerging role of exosomes in liver physiology and pathology , 2017, Hepatology research : the official journal of the Japan Society of Hepatology.

[69]  J. Rutledge,et al.  Triglyceride-rich lipoprotein lipolysis products increase blood-brain barrier transfer coefficient and induce astrocyte lipid droplets and cell stress. , 2017, American journal of physiology. Cell physiology.

[70]  A. Ahmadiani,et al.  Mitochondrial Dysfunction and Biogenesis in Neurodegenerative diseases: Pathogenesis and Treatment , 2017, CNS neuroscience & therapeutics.

[71]  M. Bredella,et al.  The Association Between IGF-1 Levels and the Histologic Severity of Nonalcoholic Fatty Liver Disease , 2017, Clinical and Translational Gastroenterology.

[72]  Zhi-Sheng Jiang,et al.  Hyperlipidemia-induced apoptosis of hippocampal neurons in apoE(−/−) mice may be associated with increased PCSK9 expression , 2016, Molecular medicine reports.

[73]  R. Swerdlow,et al.  Mitochondria, Cybrids, Aging, and Alzheimer's Disease. , 2017, Progress in molecular biology and translational science.

[74]  R. Carr,et al.  Nonalcoholic Fatty Liver Disease: Pathophysiology and Management. , 2016, Gastroenterology clinics of North America.

[75]  M. Beekman,et al.  Fibroblast growth factor 21 reflects liver fat accumulation and dysregulation of signalling pathways in the liver of C57BL/6J mice , 2016, Scientific Reports.

[76]  S. Glaser,et al.  Exosomes in liver pathology. , 2016, Journal of hepatology.

[77]  M. Orešič,et al.  Ceramides Dissociate Steatosis and Insulin Resistance in the Human Liver in Non-Alcoholic Fatty Liver Disease Short title : Ceramides in Human Non-Alcoholic Fatty Liver Disease , 2016 .

[78]  G. Gores,et al.  Lipid-Induced Signaling Causes Release of Inflammatory Extracellular Vesicles From Hepatocytes. , 2016, Gastroenterology.

[79]  Jeanne M. Clark,et al.  Nonalcoholic fatty liver disease is associated with cognitive function in adults , 2016, Neurology.

[80]  G. Gores,et al.  Mixed lineage kinase 3 mediates release of C‐X‐C motif ligand 10–bearing chemotactic extracellular vesicles from lipotoxic hepatocytes , 2016, Hepatology.

[81]  R. Coffman,et al.  Hepatocyte mitochondrial DNA drives nonalcoholic steatohepatitis by activation of TLR9. , 2016, The Journal of clinical investigation.

[82]  S. Mclennan,et al.  Molecular Sciences Extracellular Vesicles: a New Frontier in Biomarker Discovery for Non-alcoholic Fatty Liver Disease , 2022 .

[83]  M. Yin,et al.  Hepatocytes release ceramide-enriched pro-inflammatory extracellular vesicles in an IRE1α-dependent manner[S] , 2016, Journal of Lipid Research.

[84]  Leon E. Toussaint,et al.  Non-alcoholic fatty liver disease induces signs of Alzheimer’s disease (AD) in wild-type mice and accelerates pathological signs of AD in an AD model , 2016, Journal of Neuroinflammation.

[85]  P. Chaurand,et al.  Aberrant Lipid Metabolism in the Forebrain Niche Suppresses Adult Neural Stem Cell Proliferation in an Animal Model of Alzheimer's Disease. , 2015, Cell stem cell.

[86]  P. Acton,et al.  Peripheral Administration of Tumor Necrosis Factor-Alpha Induces Neuroinflammation and Sickness but Not Depressive-Like Behavior in Mice , 2015, BioMed research international.

[87]  H. Neumann,et al.  CXCL10 Triggers Early Microglial Activation in the Cuprizone Model , 2015, The Journal of Immunology.

[88]  G. Gores,et al.  Death Receptor-Mediated Cell Death and Proinflammatory Signaling in Nonalcoholic Steatohepatitis , 2014, Cellular and molecular gastroenterology and hepatology.

[89]  A. Wree,et al.  Circulating Extracellular Vesicles with Specific Proteome and Liver MicroRNAs Are Potential Biomarkers for Liver Injury in Experimental Fatty Liver Disease , 2014, PloS one.

[90]  J. Trepanowski,et al.  Fetuin-A: a novel link between obesity and related complications , 2014, International Journal of Obesity.

[91]  A. Wree,et al.  NLRP3 inflammasome activation is required for fibrosis development in NAFLD , 2014, Journal of Molecular Medicine.

[92]  S. M. de la Monte,et al.  Brain metabolic dysfunction at the core of Alzheimer's disease. , 2014, Biochemical pharmacology.

[93]  T. Saibara,et al.  Type 2 diabetes mellitus is associated with the fibrosis severity in patients with nonalcoholic fatty liver disease in a large retrospective cohort of Japanese patients , 2014, Journal of Gastroenterology.

[94]  M. Synofzik,et al.  Evidence for altered transport of insulin across the blood–brain barrier in insulin-resistant humans , 2014, Acta Diabetologica.

[95]  Hulun Li,et al.  Accumulation of natural killer cells in ischemic brain tissues and the chemotactic effect of IP-10 , 2014, Journal of Neuroinflammation.

[96]  Q. Anstee,et al.  Non-alcoholic fatty liver disease: a practical approach to diagnosis and staging , 2013, Frontline Gastroenterology.

[97]  R. Schwabe,et al.  Fate-tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its etiology , 2013, Nature Communications.

[98]  E. Fisher,et al.  Lipoprotein Metabolism, Dyslipidemia, and Nonalcoholic Fatty Liver Disease , 2013, Seminars in Liver Disease.

[99]  Andreas Meisel,et al.  Sugar for the brain: the role of glucose in physiological and pathological brain function , 2013, Trends in Neurosciences.

[100]  J. Loeffler,et al.  Fatting the brain: a brief of recent research , 2013, Front. Cell. Neurosci..

[101]  A. Goustin,et al.  Ahsg-fetuin blocks the metabolic arm of insulin action through its interaction with the 95-kD β-subunit of the insulin receptor. , 2013, Cellular signalling.

[102]  Laura J. Dixon,et al.  Kupffer cells in the liver. , 2013, Comprehensive Physiology.

[103]  N. Chattipakorn,et al.  DPP4‐inhibitor improves neuronal insulin receptor function, brain mitochondrial function and cognitive function in rats with insulin resistance induced by high‐fat diet consumption , 2013, The European journal of neuroscience.

[104]  V. Adam,et al.  Redox status expressed as GSH:GSSG ratio as a marker for oxidative stress in paediatric tumour patients. , 2012, Oncology letters.

[105]  R. Palmiter,et al.  Mitochondrion-derived reactive oxygen species lead to enhanced amyloid beta formation. , 2012, Antioxidants & redox signaling.

[106]  J. Kaye,et al.  Dyslipidemia and Blood-Brain Barrier Integrity in Alzheimer's Disease , 2012, Current gerontology and geriatrics research.

[107]  Eduardo D. Martín,et al.  IRS-2 Deficiency Impairs NMDA Receptor-Dependent Long-term Potentiation , 2011, Cerebral cortex.

[108]  D. Ghareeb,et al.  Non-alcoholic fatty liver induces insulin resistance and metabolic disorders with development of brain damage and dysfunction , 2011, Metabolic Brain Disease.

[109]  J. Wands,et al.  Weight loss amelioration of non‐alcoholic steatohepatitis linked to shifts in hepatic ceramide expression and serum ceramide levels , 2011, Hepatology research : the official journal of the Japan Society of Hepatology.

[110]  A. Dolganiuc,et al.  Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells , 2011, Hepatology.

[111]  G. Reiser,et al.  Non‐esterified polyunsaturated fatty acids distinctly modulate the mitochondrial and cellular ROS production in normoxia and hypoxia , 2011, Journal of neurochemistry.

[112]  B. Platt,et al.  Susceptibility to diet-induced obesity and glucose intolerance in the APPSWE/PSEN1A246E mouse model of Alzheimer’s disease is associated with increased brain levels of protein tyrosine phosphatase 1B (PTP1B) and retinol-binding protein 4 (RBP4), and basal phosphorylation of S6 ribosomal protein , 2011, Diabetologia.

[113]  Joel Z Stengel,et al.  Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: a prospective study. , 2011, Gastroenterology.

[114]  J. Féher,et al.  Serum Dipeptidyl Peptidase-4 Activity in Insulin Resistant Patients with Non-Alcoholic Fatty Liver Disease: A Novel Liver Disease Biomarker , 2010, PloS one.

[115]  Martin D. Brand,et al.  The sites and topology of mitochondrial superoxide production , 2010, Experimental Gerontology.

[116]  B. Garner Lipids and Alzheimer's disease. , 2010, Biochimica et biophysica acta.

[117]  L. Ferrucci,et al.  Relationship between low levels of high-density lipoprotein cholesterol and dementia in the elderly. The InChianti study. , 2010, The journals of gerontology. Series A, Biological sciences and medical sciences.

[118]  Rosemary O’Connor,et al.  Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer's disease indicate possible resistance to IGF-1 and insulin signalling , 2010, Neurobiology of Aging.

[119]  J. Wands,et al.  Ceramide-mediated insulin resistance and impairment of cognitive-motor functions. , 2010, Journal of Alzheimer's disease : JAD.

[120]  L. Lesmana,et al.  Diagnostic value of a group of biochemical markers of liver fibrosis in patients with non‐alcoholic steatohepatitis , 2009, Journal of digestive diseases.

[121]  M. Swain,et al.  Cerebral Microglia Recruit Monocytes into the Brain in Response to Tumor Necrosis Factorα Signaling during Peripheral Organ Inflammation , 2009, The Journal of Neuroscience.

[122]  Z Walker,et al.  Microglial activation and amyloid deposition in mild cognitive impairment , 2009, Neurology.

[123]  J. Wands,et al.  Hepatic ceramide may mediate brain insulin resistance and neurodegeneration in type 2 diabetes and non-alcoholic steatohepatitis. , 2009, Journal of Alzheimer's disease : JAD.

[124]  M. Mahmoudi,et al.  Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer. , 2009, Recent patents on inflammation & allergy drug discovery.

[125]  A. Passaro,et al.  Markers of endothelial dysfunction in older subjects with late onset Alzheimer's disease or vascular dementia , 2008, Journal of the Neurological Sciences.

[126]  H. Scheich,et al.  Overexpression of human apolipoprotein B-100 induces severe neurodegeneration in transgenic mice. , 2008, Journal of proteome research.

[127]  K. Xiang,et al.  Serum retinol binding protein 4 and nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. , 2008, Diabetes research and clinical practice.

[128]  B. S. Mohammed,et al.  Alterations in adipose tissue and hepatic lipid kinetics in obese men and women with nonalcoholic fatty liver disease. , 2008, Gastroenterology.

[129]  V. Haroutunian,et al.  Gene Expression Alterations in the Sphingolipid Metabolism Pathways during Progression of Dementia and Alzheimer’s Disease: A Shift Toward Ceramide Accumulation at the Earliest Recognizable Stages of Alzheimer’s Disease? , 2007, Neurochemical Research.

[130]  Y. Le Marchand-Brustel,et al.  Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression. , 2007, Endocrinology.

[131]  G. Wilcock,et al.  The metabolic syndrome and Alzheimer disease. , 2007, Archives of neurology.

[132]  S. Summers,et al.  Ceramides in insulin resistance and lipotoxicity. , 2006, Progress in lipid research.

[133]  E. Parks,et al.  Spillover of dietary fatty acids and use of serum nonesterified fatty acids for the synthesis of VLDL-triacylglycerol under two different feeding regimens. , 2005, Diabetes.

[134]  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.

[135]  I. Santana,et al.  Mitochondria dysfunction of Alzheimer's disease cybrids enhances Aβ toxicity , 2004, Journal of neurochemistry.

[136]  Y. Masui,et al.  Functional and morphometric study of the liver in motor neuron disease , 2004, Journal of Neurology.

[137]  J. Kushner,et al.  Insulin Receptor Substrate-2 Deficiency Impairs Brain Growth and Promotes Tau Phosphorylation , 2003, The Journal of Neuroscience.

[138]  I A Silver,et al.  Tissue oxygen tension and brain sensitivity to hypoxia. , 2001, Respiration physiology.

[139]  H. Lei,et al.  Tumour necrosis factor-α causes an increase in blood-brain barrier permeability during sepsis , 2001 .

[140]  H. Lei,et al.  Tumour necrosis factor-alpha causes an increase in blood-brain barrier permeability during sepsis. , 2001, Journal of medical microbiology.

[141]  R. Swerdlow,et al.  Alzheimer's disease cybrids replicate β‐amyloid abnormalities through cell death pathways , 2000 .

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

[143]  W. Banks,et al.  Murine tumor necrosis factor alpha is transported from blood to brain in the mouse , 1993, Journal of Neuroimmunology.

[144]  D. Porte,et al.  Insulin binding to brain capillaries is reduced in genetically obese, hyperinsulinemic Zucker rats , 1990, Peptides.

[145]  K. Shirai,et al.  Existence of lipoprotein lipase in rat brain microvessels. , 1986, The Tohoku journal of experimental medicine.

[146]  A. Bernsmeier,et al.  [The glucose consumption of the brain & its dependence on the liver]. , 1958, Archiv fur Psychiatrie und Nervenkrankheiten, vereinigt mit Zeitschrift fur die gesamte Neurologie und Psychiatrie.

[147]  D. Harman Aging: a theory based on free radical and radiation chemistry. , 1956, Journal of gerontology.