""139” 2‘! HR ‘\ :‘Iluiiu A x-cahje, : 91" ' a «I -u..- \. ,.....‘J - ‘.3 THE-I)" ‘15 2 LIBRARY Michigan State University This is to certify that the dissertation entitled INVOLVEMENT OF SATURATED FATTY ACIDS IN CAUSING PATHOPHYSIOLOGICAL AND METABOLIC CHANGES ASSOCIATED WITH ALZHEIMER’S DISEASE presented by SACHIN PATIL has been accepted towards fulfillment of the requirements for the PhD. degree in Chemiml EngineerinL Major Professor‘s Signature 9/24 o7 Date MSU is an aflin'native-action, equal-opportunity employer i l PLACE IN RETURN BOX to remove this checkout from your record. f TO AVOID FINES return on or before date due. ' MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE JAN 2 8 2009 1- ’flf" 45:ng 2 3 ZKJKHH 6/07 p:/CIRC/Date0ue.indd-p.1 INVOLVEMENT 0F SATURATED FATTY ACIDS IN CAUSING PATHOPHYSIOLOGICAL AND METABOLIC CHANGES ASSOCIATED WITH ALZHEIMER’S DISEASE By SACHIN PATIL A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Chemical Engineering and Materials Science 2007 ABSTRACT INVOLVEMENT OF SATURATED FATTY ACIDS IN CAUSING PATHOPHYSIOLOGICAL AND METABOLIC CHANGES ASSOCIATED WITH ALZHEIMER’S DISEASE By SACHIN PATIL Alzheimer’s disease (AD) is a very complex, age-related neurodegenerative disorder. Pathologically, AD brain is characterized by extracellular deposits of amyloid beta (AB) protein and intracellular accumulation of neurofibrillary tangles (NFTS) composed of hyperphosphorylated tau protein. The present studies were undertaken to investigate the possible causal role of saturated free fatty acids (FFAS) in the pathogenesis of AD with the primary focus on establishing the underlying mechanism, which may prove vital in developing novel therapeutic strategies for AD. We found that saturated FFAS had no direct effect on primary rat cortical neurons in terms of causing both the pathophysiological (AB production and tau hyperphosphorylation) as well as metabolic (glucose metabolism) abnormalities. In contrast, saturated FFAS significantly increased AB production and tau hyperphosphorylation in neurons through astroglial mediation. The conditioned media from FFA-treated astroglia induced increased production of reactive oxygen species (ROS) in neurons and co-treatment of neurons with N-acetyl cysteine, an anti-oxidant, inhibited FFA-astroglia-induced AB production and tau hyperphosphorylation, suggesting a central role of astroglia-mediated oxidative stress in the FFA-induced pathophysiological abnormalities in neurons. Furthermore, saturated FFAS significantly decreased the level of astroglial glucose transporter (GLUTl) and downregulated glucose uptake and lactate release by astroglia. By using a powerful mathematical technique, metabolic flux analysis (MFA), we found that de novo synthesis of ceramide in PA- treated astroglia was significantly higher as compared to controls. The treatment of astroglia with L-cycloserine inhibited FFA-induced de novo synthesis of ceramide in astroglia and in turn, ROS production, AB production and tau hyperphosphorylation in neurons. The data suggest that astroglial ceramide may play a central role in FFA- induced, AD-associated pathophysiological changes in neurons. To conclude, our results establish an underlying mechanism by which saturated FFAS induce AD-associated pathophysiological as well as metabolic changes, placing “astroglial FFA metabolism” at the center of the pathological cascade of AD. Further understanding of astroglial FFA metabolism, both in vitro and in vivo, may help in uncovering new aspects of AD pathogenesis that may be translated into potential targets for therapeutic intervention in AD. © COPYRIGHT SACHIN PATIL 2007 DEDICATED TO MY FAMILY ACKNOWLEDGMENTS No Matter How Hard You Work, You have Only One Brain and Two Handr, Great Work Demandr Sac/z Numerou: Once, W/ziclt You Find In Your Friends: At this juncture of successful completion of my dissertation, I wish to convey my deepest sense of gratitude towards those colleagues and friends who have helped me to achieve my goal. First and foremost, I would like to thank my advisor Dr. Christina Chan, for her guidance, constant inspiration and complete intellectual freedom throughout my research tenure. I am also thankful to all the members of my Ph.D. advisory committee-Dr. Bill Atchison, Dr. Worden and Dr. Walton. Their support and interest in my work, together with the critical evaluation of the work, helped me tremendously to improve the quality of the present dissertation. Words fall short to express my heartfelt feelings and gratitude towards these great personalities. I am proud that I got a chance to work with them. I would also like to thank my M.S. advisor, my GURU, Professor G. D. Yadav (UICT, Mumbai, India). His inspiring guidance has carved a niche in my heart. He imbibed in me the qualities of confidence, perseverance and unwavering commitment towards one’s work, which I am sure would definitely help me throughout my life. If “a friend in need is a true friend indeed”, then all my colleagues are my friends, in true sense. I wouldn’t be able to justify my thankful feelings by mere words towards my labmates (from Drs. Chan and Walton labs) who have always been there whenever I needed their help. My special thanks my senior colleague, Dr. Shireesh Srivastava who vi helped me throughout my tenure in the lab with constant encouragement and valuable suggestions. The mathematical work (MFA) that I performed for dissertation wouldn’t have been possible without his kind help. I am also thankful to my friends for their constant, unwavering encouragement- Dr. Mehul (Netherlands) and his wife, Dr. Sameer (Netherlands), Dr. Lek (Scotland) and his wife Heena, Dr. Ambareesh (Texas), Dr. Navin (MSU), Dr. Zheng Li (Boston), Dr. Srivatsan (Harvard), Sachin Injal (Lawyer), Dhanu, Adam, Nandu, Priti, Lufang, Chisa, Tanmay, Susan, Katie, Dana, Tao, Alison, Bahareh, John, Ketan, Hemant, Joe, Mike, Shengnan, Linxia, Deebika and Xureui. My special thanks also to Shirley Owens, for her help in terms of confocal microscopy. I was also very fortunate to have hard-working, sincere undergraduates assisting me- Alexis (Shell), Robert (U of M) and Joe. Their youthful exuberance was pleasantly encouraging. I would also like to thank the Department of Chemical Engineering and Materials Science, the College of Engineering, Quantitative and Biological Modeling Initiative (QBMI) at MSU, the office of international student and scholars, council of graduate studies (COGS) and the graduate school for the financial support in the form of Food, Nutrition and Chronic Disease Fellowship and travel fellowships. Their contributions helped me present my research at the national and international scientific meetings. Also, my apartment at Student living Center (SLC) was my home-away-from-home and my roomies made it that way with whom I will always share the deepest friendship- Sarfaraz (India), Arda (Turkey), Jorge (Peru), Kensuke (Japan), Qiang (China) and Paul (USA). It was truly an international experience. I have learned so many things from them vii that I cannot repay them in any way. Here, I would also like to express my gratitude towards all my ward mates at SLC for their true friendship- Stephanie, Tiffany, Kelly, Kerry, Emily, Conrad, Justin, Don, Dan, Ben, Josh, Gary. My special thanks to Cindy, Vern, Nicole and President Hinkle. Also thanks to the Elders who taught me how to tie a double knot. My sincere thanks to Amit and his wife Deboleena, for inviting me over for countless lunches and dinners. But, obviously it’s much more than food that is and will be keeping us in the bond of friendship! Last but not the least, it was the confidence laid in me by my Mom, Dad, my sisters and brother-in-laws, which enabled me to reach this pinnacle of success. I owe a debt of gratitude for their constant encouragement for higher education and unflinching, altruistic help during this work. viii -“fi‘fi'va- , . .- . *- 7 TABLE OF CONTENTS LIST OF TABLES xii LIST OF FIGURES xiii LIST OF ABBREVIATIONS xviii CHAPTER 1. INTRODUCTION 1 1.1 ALZHEIMER’S DISEASE .................................................................................... 1 1.2 THE CURRENT HYPOTHESES OF AD ................................................. 4 1.2.1 The Amyloid Cascade Hypothesis ................................................................... 4 1.2.2 The Cholesterol Metabolism and AD .............................................................. 6 1.2.3 Oxidative Stress and AD ................................................................................. 7 1.2.4 The Cholnergic Hypothesis of AD .................................................................. 8 1.2.5 Homocysteine and AD .................................................................................... 9 1.2.6 The Pathogen Hypothesis of AD ................................................................... 10 1.3 POTENTIAL INVOLVEMENT OF SATURATED FATTY ACIDS IN THE PATHOGENESIS OF AD ......................................................................................... 11 1.4 GOALS OF THE PRESENT STUDY .................................................................. 12 1.5 THESIS OUTLINE .............................................................................................. 12 CHAPTER 2. SATURATED FATTY ACID-INDUCED HYPERPHOSPHORYLATION OF TAU IN PRIMARY RAT CORTICAL NEURONS--- - - - 14 2.1 INTRODUCTION ............................................................................................... 14 2.2 MATERIALS AND METHODS ......................................................................... 18 2.2.1 Isolation and Culture of Primary Rat Cortical Neurons and Astroglia ............ 18 2.2.2 Lactate Dehydrogenase (LDH) Assay ........................................................... 18 2.2.3 Western Blot Analysis ................................................................................... 19 2.2.4 Immunofluorescence Analysis of Neurons and Astroglia ............................... 20 2.2.5 Immunostaining of Reactive oxygen Species ................................................. 20 2.2.6 Data Analyses ............................................................................................... 21 2.3 RESULTS AND DISCUSSION ........................................................................... 21 2.3.] Direct Treatment of Neurons with Saturated Fatty Acids ............................... 21 2.3.2 Involvement of Saturated F FAS in Tau Hyperphosphorylation Through Astroglial Mediation .............................................................................................. 22 2.3.3 Involvement of Oxidative Stress in FFA-Astroglia-Induced Tau Hyperphopshorylation in Neurons .......................................................................... 26 2.4 CONCLUSIONS ................................................................................................. 27 ix CHAPTER 3. SATURATED FATTY ACID-INDUCED AMYLOIDOGENIC PROCESSING OF APP IN PRIMARY RAT CORTICAL N EURONS ................... 28 3.1 INTRODUCTION ............................................................................................... 28 3.2 MATERIALS AND METHODS ......................................................................... 33 3.2.1 Isolation and Culture of Primary Rat Cortical Neurons and Astroglia ............ 33 3.2.2 Western Blot Analysis ................................................................................... 34 3.2.3 Immunofluorescence Analysis of BACEl in Neurons ................................... 34 3.2.4 Data Analyses ............................................................................................... 35 3.3 RESULTS ........................................................................................................... 35 3.3.1 Direct Treatment of Neurons with Saturated Fatty acid (Palmitic Acid) ......... 35 3.3.2 Involvement of Saturated FF As in BACEl Up-regulation and Amyloidogenic Processing of APP Through Astroglial Mediation .................................................. 36 3.3.3 Involvement of Oxidative Stress in FFA-Astroglia-Induced BACEl Up- regulation and Amyloidogenic Processing of APP in Primary Neurons .................. 39 3.4 CONCLUSIONS ................................................................................................. 41 CHAPTER 4. SATURATED FATTY ACID-INDUCED ABNORMAL METABOLIC CHANGES ASSOCIATED WITH ALZHEIMER'S DISEASE ....... 42 4.1 INTRODUCTION ............................................................................................... 42 4.2 MATERIALS AND METHODS ......................................................................... 45 4.2.1 Isolation and Culture of Primary Rat Cortical Neurons and Astroglia ............ 45 4.2.2 Western Blot Analysis ................................................................................... 46 4.2.3 Biochemical Measurements of Cellular Metabolites ...................................... 47 4.2.4 Metabolic Flux Analysis (MFA) .................................................................... 48 4.2.5 Measurement of Intracellular ATP in Astroglia ............................................. 54 4.2.6 Measurement of Intracellular Ceramide in Astroglia ..................................... 55 4.2.7 Data Analyses ............................................................................................... 56 4.3 RESULTS AND DISCUSSION ........................................................................... 56 4.3.1 FFA-Induced Abnormal Glucose Metabolism ............................................... 56 4.3.2 Cellular Mechanism of FF A-Induced Abnormal Glucose Metabolism in Astroglia ................................................................................................................ 59 4.3.3 FFA-Induced Global Metabolic Changes in Astroglia ................................... 61 4.3.3.1 Verification of the psueudo-steady state assumption and linearity of fluxes over 24 hours ...................................................................................... 62 4.3.3.2 MFA Analysis ................................................................................... 64 4.4 CONCLUSIONS ................................................................................................. 69 CHAPTER 5. INVOLVEMENT OF CERAMIDE IN FFA-ASTROGLIA- INDUCED PATHOPHYSIOLOGICAL ABNORMALITIES ASSOCIATED WITH AD -- 70 5.1 INTRODUCTION ............................................................................................... 70 5.1 MATERIALS AND METHODS ......................................................................... 72 5.2.1 Isolation and Culture of Primary Neurons and Astroglia from Rat Cortex and Cerebellum ............................................................................................................ 72 5.2.21mmunostaining of Reactive Oxygen Species (ROS) ..................................... 74 5.2.3 Western Blot Analysis ................................................................................... 74 5.2.4 ELISA measurements of AB4O and AB42 ..................................................... 76 5.2.5 Data Analyses ............................................................................................... 76 5.3 RESULTS AND DISCUSSION ........................................................................... 76 5.3.1. Involvement of Astroglial Ceramide in F FA-Astroglia-Induced ROS production in Neurons ........................................................................................... 76 5.3.2 Involvement of astroglial ceramide in FFA-astroglia-induced amyloidogenesis and tau hyperphoshorylation in neurons ................................................................. 78 5.3.3 A possible explanation for the region-specific and cell type—specific damage observed in AD ...................................................................................................... 84 5.4 CONCLUSIONS ................................................................................................. 87 CHAPTER 6 CONCLUSIONS--- -- - ------- - 88 APPENDIX- - - 96 1. Brain cells from older animals ............................................................................... 96 2. Trans-well experiments .......................................................................................... 96 3. Exogenous addition of ceramide as a positive control ............................................ 97 LIST OF PUBLICATIONS -- - - - -- - 99 BIBLIOGRAPHY -- ----- - -------------- - ------------- 100 xi LIST OF TABLES Table 4.1 List of the cellular metabolic reactions ............................................... 43 Table 4.2 List of intracellular metabolites ....................................................... 46 Table 4.3 List of measured fluxes ................................................................. 47 Table 4.4 Ratio of intracellular to extracellular lactate levels ................................. 57 Table 4.5 Metabolic flux values calculated by MFA ........................................... 58 xii Images in this dissertation are presented colored. LIST OF FIGURES Figure 1.1. The shrinkage of brain in Alzheimer’s disease. The brain regions involved in cognitive functions (frontal and temporal lobes, left and lower part of brain, respectively) show significant shrinkage in AD brain as compared to normal brain ........ 1 Figure 1.2. The pathophysiological lesions in AD brain. (A) Amyloid beta (AB) plaque and (B) Neurofibrillary tangle (NFT) .............................................................. 2 Figure 1.3. The decreased glucose metabolism in Alzheimer’s disease. The positron emission tomography (PET) images Show significant decline in cerebral glucose metabolism in AD brain as compared to normal brain; red and yellow-high metabolism, blue-low metabolism ................................................................................. 2 Figure 2.1. The physiology and pathology of tau. (A) In healthy neurons, tau stabilizes microtubules. (B) In AD, tau hyperphosphorylates and detaches from microtubules leading to tangle formation .......................................................................... 8 Figure 2.2. Direct treatment of neurons with saturated FFAS. Primary rat cortical neurons were treated for 24 hours with 0.2mM of either palmitic acid (PA) or stearic acid (SA) or with 5% bovine serum albumin (BSA), vehicle for FFAS (control). Detergent cell lysates from fatty acid-treated and control cells were immunoblotted with PHF-l and AT8 antibodies, which recognize phosphorylated tau. B-actin is shown as a marker for protein loading. ..................................................................................... 15 Figure 2.3. MAP-2, GFAP and AT8 immunostaining. (A) Neurons treated directly with FFAS for 24 hours. (B) Astrocytes treated for 12 hours with 0.2mM of either palmitic acid (PA) or stearic acid (SA) or 5% BSA (control). (C) Neurons treated for 24 hours with conditioned media from fatty acid-treated or control astrocytes. (D) Immunofluorescence labeling of phosphorylated tau with AT8 antibody in neurons treated with conditioned media from fatty acid-treated or untreated astrocytes (control). Images were obtained with confocal fluorescence microscopy. (Objective lens magnification- 40X for MAP-2 and GFAP, 63X for AT8) .................................... 16 Figure 2.4. Measurement of LDH release from astroglia treated with PA. 24h treatment with 0.2mM PA failed to liberate LDH from astroglia as compared to controls. In contrast, 1hr treatment of astroglia with 300mM H202 (positive control) induced significant LDH liberation after 24h as compared to both control and PA-treated cells. Data are taken from 3 different experiments and are expressed as mean :1: SD. One-way xiii ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control ............................................ 17 Figure 2.5. Astroglia-mediated, fatty acid-induced hyperphosphorylation of tau in neurons. (A) Western blot analysis of hyperphosphorylated tau was performed using phospho-specific antibodies PHF-l and AT8. (B) Histograms corresponding to PHF-l and AT8 blots represent quantitative determinations of intensities of the relevant bands. Data represent mean :1: SD. of 3 independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control; #, p<0.05 compared with fatty acid treatment ............................................................................................. 18 Figure 2.6. Intracellular accumulation of ROS in neurons. (A) Astroglia and (B) neurons were stained with CM-H2DCFDA for intracellular ROS detection and examined with confocal fluorescence microscopy (Zeiss LSM 5 Pascal). (Objective lens magnification, 63X) ................................................................................. 19 Figure 3.1. Processing of amyloid precursor protein (APP). (A) The non- amyloidogenic pathway catalyzed by a- and y-secretase. (B) The amyloidogenic pathway catalyzed by B- and y-secretase ................................................................... 23 Figure 3.2. Direct treatment of neurons with palmitic acid. Primary rat cortical neurons were treated for 24 hours with 0.2mM of palmitic acid (PA) or with 4% bovine serum albumin (BSA), vehicle for PA (control). Detergent cell lysates from PA-treated and control cells were immunoblotted with BACEl antibody. B-actin is shown as a marker for protein loading. The blots are representative of 3 independent experiments..29 Figure 3.3. BACEl immunostaining. Immunofluorescence labelling of BACEl in neurons treated for 24 hours with conditioned media from PA-treated (for 24 hours) or untreated astrocytes (control). Images were obtained with confocal fluorescence microscopy (objective lens magnification- 40X) ................................................ 30 Figure 3.4. Astroglia-mediated, PA-induced BACEI upregulation in neurons. Astrocytes were treated for 12 and 24 hours with 0.2mM of PA or 4% BSA (control), followed by transfer of the astrocytes-conditioned media to neurons (24 hours treatment). (A) Western blot analysis of BACEl protein levels in neurons. B-actin is shown as a marker for protein loading. (B) Histograms represent quantitative determinations of intensities of the relative bands. Data represent mean 1 SD. of three independent experiments. Student’s t-test was used for analyzing differences between different treatment groups. *, p<0.05 compared with respective control ............................... 31 Figure 3.5. Immunoblot analysis of presenilin-l (PSI) levels in neurons treated with astrocytes-conditioned media. Astrocytes were treated for 24 hours with 0.2mM PA or 5% BSA (control), followed by transfer of the astrocytes-conditioned media to neurons (24 hours treatment), with or without lOmM DMU. Detergent cell lysates from PA— xiv treated and control cells were immunoblotted for PS] and PSl-CTF levels. The immunoblot is representative of 3 independent experiments .................................. 32 Figure 3.6. Oxidative stress involved in astroglia-mediated, PA-induced elevations in BACEl and C99 levels in neurons. Astrocytes were treated for 24 hours with 0.2mM PA or 5% BSA (control), followed by transfer of the astrocytes-conditioned media to neurons (24 hours treatment), with or without lOmM DMU. (A) Western blot analysis of BACEl, APP and C99 protein levels in neurons. (B) Histograms represent quantitative determinations of intensities of the relative bands. Data represent mean at SE. of three independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control; ** p<0.05 compared with PA treatment ............................................................. 33 Figure 4.1. Conventional view of cerebral glucose metabolism ........................... 37 Figure 4.2. Cerebral glucose metabolism based on the novel astrocyte-neuron lactate shuttle hypothesis. ................................................................................ 38 Figure 4.3. Astroglial metabolic network. Boxes represent extracellular metabolites, while ovals represent intracellular metabolites. The direction of reaction assumed in the model is indicated by arrows. ..................................................................... 45 Figure 4.4. PA downregulates glucose uptake and lactate release by astroglia. The cortical neurons and astroglia were treated for 24 hours with 0.2mM of PA or 4% BSA. (A) In neurons, PA treatment did not change glucose uptake and lactate production. (B) PA treatment Significantly decreased glucose uptake and lactate production by astroglia. AMPK-activator AICAR did not inhibit PA-induced downregulation in glucose metabolism. Data represent mean :i: SD. of Six experiments. Student’s t-test was used for analyzing the differences between the two treatment groups. *, p<0.05 compared with respective control. ................................................................................... 50 Figure 4.5. Measurement of intracellular ATP in astroglia. The cortical astroglia were treated for 24 hours with 0.2mM of PA or 4% BSA. PA treatment increased cellular ATP production in astroglia. Data represent mean :1: SD. of 3 experiments. Student’s t—test was used for analyzing the differences between the two treatment groups. *, p<0.05 compared with respective control. ............................................................................. 51 Figure 4.6. PA downregulates GLUTI level in astroglia. Astroglia were treated for 24 hours with 0.2mM of PA or 4% BSA. The immunoblot analysis shows that PA-treatment significantly decreased the levels of GLUTl as compared to the untreated ones. B-actin is shown as a marker for protein loading. Histogram represents quantitative determinations of intensities of the relative bands normalized with actin. Data represent mean :t SD. of three independent experiments. Student’s t-test was used for analyzing the differences between the two treatment groups. *, p<0.05 compared with respective control ........... 53 Figure 4.7. FFA—metabolizing pathways involved in cellular ROS production ......... 54 XV Figure 4.8A. Time-dependent measurements of intracellular lactate levels. Astroglia were treated for 6, 12 and 24 hours with 0.2mM of PA or 4% BSA. The cells were trypsinized, washed with TBS and lyzed by using 0.7% perchloric acid. The lactate levels in cell lysate were measured by enzymatic assay. Data represent mean 3: SD. of three independent experiments .................................................................. 56 Figure 4.8B. Time-dependent measurements of extracellular lactate levels. Astroglia were treated for 6, 12 and 24 hours with 0.2mM of PA or 4% BSA. The conditioned media were collected and the lactate levels in the media were measured by enzymatic assay. Data represent mean :t SD. of three independent experiments ....................... 57 Figure 4.9. PA-induced, de novo synthesis of ceramide in astroglia. Astroglia were treated for 24h with 0.2mM PA or 4% BSA (control), afier which cellular lipids were extracted for ceramide determination by HPLC. PA significantly increased ceramide synthesis in astroglia, which was completely inhibited by treatment of astroglia with 2mM L-CS, an inhibitor of de novo synthesis of ceramide. Data are taken from 3 different experiments and are expressed as mean i S.D. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control; #, p<0.05 compared with PA treatment ..................... 61 Figure 5.1. Cellular ceramide production. Ceramide is produced in cells by de novo synthesis from serine and palmitoyl-COA or by breakdown of membrane sphingomyelins ...................................................................................... 64 Figure 5.2. Involvement of astroglial ceramide in PA-astroglia-induced ROS production in neurons. Co-treatment of astroglia with 2mM L-CS inhibited PA- astroglia-induced ROS production in neurons. The neurons were stained with CM- H2DCFDA for intracellular ROS detection and examined with confocal fluorescence microscopy (Zeiss LSM 5 Pascal). (Objective lens magnification, 40X) .................... 70 Figure 5.3. The expression of iNOS and IL-6 in astroglia. The expression of iNOS, but not IL-6 was increased by PA treatment in astroglia. Co-treatment of astroglia with 2mM L-CS inhibited PA-induced iNOS expression in astroglia. B-actin is shown as a marker for protein loading ................................................................................... 71 Figure 5.4. Involvement of astroglial ceramide in PA-astroglia—induced elevations in BACEl and amyloidogenic processing of APP in neurons. In neurons treated with conditioned media from PA-treated astroglia, the levels of (A) BACEl, (B) C99 and (C) AB40 and AB42 were found elevated. These PA-astroglia-induced tau abnormalities were blocked by inhibiting astroglial ceramide synthesis with 2mM L-CS. Data represent mean :1: SD. of three independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control; #, p<0.05 compared with PA treatment .............................. 72 Figure 5.5. Involvement of astroglial ceramide in PA-astroglia-induced tau hyperphosphorylation in neurons. In neurons treated with conditioned media from PA- xvi treated astroglia, tau was found pathologically hyperphosphorylated as shown by irnmunoblotting with PHF -1 and AT8 antibodies. Tau-l detects dephosphorylated tau, thus Showing decreased levels in PA-astroglia-treated neurons. These PA-astroglia- induced tau abnormalities were blocked by inhibiting astroglial ceramide synthesis with 2mM L-CS. Histograms corresponding to PHF-l and AT8 blots represent quantitative determinations of intensities of the relevant bands normalized with actin. Data represent mean i SD. of three independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control; #, p<0.05 compared with PA treatment .............................. 73 Figure 5.6. PA-astroglia-induced activation of AD-specific kinases in neurons is mediated by astroglial ceramide. Conditioned media from PA-treated astroglia activated (A) GSK-3 (increased levels of phosphorylated GSK-3) and (B) cdkS (increased cleavage of p35 to p25) but not (C) MAP- Erkl/2 (no change in the levels of phosphorylated MAP- Erkl/2). The treatment of astroglia with 2mM L-CS inhibited PA- astroglia-induced activation of both GSK-3 and cdk5. Data are representative of 3 different experiments ............................................................................... 75 Figure 5.7. GSK—3 is involved in PA-astroglia-induced tau hyperphosphorylation in neurons. Astroglia-conditioned media were transferred to neurons, with or without various kinase inhibitors, viz., lOmM LiC12 (GSK-3 inhibitor), lOmM Roscovitine (cdk- 5 inhibitor) and 30mM PD98059 (MAPK inhibitor). Immunoblot analysis with PHF-l and AT8 antibodies Show that only LiC12 inhibited the observed, PA-induced tau hyperphosphorylation in neurons. Histogram data represent mean :1: SD. of three independent experiments. One-way ANOVA and Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control; #, p<0.05 compared with PA treatment ............................................................. 77 Figure 5.8. Differential effects of cortical and cerebellar astroglia on cortical neurons. Cortical neurons (CTN) were treated with conditioned media from (A) cortical astroglia (CTA) and (B) cerebellar astroglia (CBA). Western blot analysis of hyperphosphorylated tau was performed using AT8 antibody. Cortical astroglia but not cerebellar astroglia were involved in PA-induced tau hyperphosphorylation in neurons. Data represent mean :l: SD. of three independent experiments. Student’s t-test was used for analyzing differences between different treatment groups. *, p<0.05 compared with control ................................................................................................ 79 Figure 6.1. The “FFA-AD” hypothesis. Proposed cellular mechanism by which astroglial FFA metabolism may play a central role in causing pathophysiological and metabolic changes associated with AD .......................................................... 83 xvii LIST OF ABBREVIATIONS AB: Amyloid beta AChE: Acetylcholine esterase AD: Alzheimer’s disease ADDLs: AB-derived diffusible ligands AMPK: AMP-activated protein kinase ApoE4: Apolipoprotein E4 APP: Amyloid precursor protein BACE: B-site APP cleaving enzyme BBB: Blood-brain barrier BSA: Bovine serim albumin cdk: cycline-dependent kinase CBA: Cerebellar astroglia CTA: Cortical astroglia CTN: Cortical neurons DAG: Diacyl glycerol DMU: 1,3-dimethyl urea ELISA: Enzyme-linked immunosorbent assay FACS: Fatty acyl-CoA synthetase FAD: Familial Alzheimer’s disease FFA: Free fatty acid GSK: Glycogen synthase kinase 4-HNE: 4-hydroxynonenal xviii ‘_ fl“..— m”...— HPLC: High performance liquid chromatography IL-6: Interleukin-6 iNOS: inducible nitric oxide synthase L-CS: L-cycloserine LDH: Lactate dehydrogenase MAPK: Mitogen-activated protein kinase MCI: Mild cognitive impairment MFA: Metabolic flux analysis NAC: N-acetyl cysteine NFTs: Neurofibrillary tangles N MDA: N-methyl D-aspartate NO: Nitric oxide PA: Palmitic acid PET: Positron emission tomography PHF: Paired helical filaments PKA: Protein kinase A PKC: Protein kinase C PS: Presenilin ROS: Reactive oxygen species SA: Stearic acid SAD: Sporadic Alzheimer’s disease SPT-l: Serine palmitoyltransferase-l TBARS: Thiobarbituric acid reactive substances xix CHAPTER 1. INTRODUCTION 1.1 ALZHEIMER’S DISEASE Dementia is one of the most complex and heterogeneous age-related disorders and the most common cause of dementia is Alzheimer’s disease (AD) '. AD is a progressive neurodegenerative disease clinically characterized by severe memory loss and cognitive impairment 2. As shown in Figure 1.1, AD brain exhibits significant shrinkage as compared to healthy controls 3. Pathologically, AD is characterized by extracellular deposits of amyloid beta (AB) protein and intracellular accumulation of neurofibrillary tangles (NFTS) composed of hyperphosphorylated tau protein (Figure 1.2) 3. In addition to these two classical neuropathological hallmarks, AD brain is also characterized by abnormal metabolic changes; abnormal cerebral glucose metabolism is one of the distinct characteristics of AD brain (Figure 1.3) 3'5. Healthy Brain AD Brain Figure 1.1. The shrinkage of brain in Alzheimer’s disease. The brain regions involved in wgnitive functions (frontal and temporal lobes, lefi and lower part of brain, respectively) show signifith shrinkage in AD brain as compared to normal brain (Figure taken from Ref. 3, with permission from Nature Publishing Group). Figure 1.2. The pathophysiological lesions in AD brain. (A) Amyloid beta (AB) plaque and (B) Neurofibrillary tangle (N FT) (Figures taken from www.alzheimers.org, 08-09-2007). Healthy Brain AD Brain Figure 1.3. The decreased glucose metabolism in Alzheimer's disease. The positron emission tomography (PET) images show significant decline in cereme glucose metabolism in AD brain as compared to normal brain; red and yellow-high metabolism, blue-low metabolism (Figure taken from Ref. 3, with permission from Nature Publishing Group). AD is classified into two categories- familial Alzheimer’s disease (FAD) and sporadic Alzheimer’s disease (SAD). FAD has been shown to be associated with the mutations in 2 APP, presenilin 1 and 2 (PSI and P82) genes on chromosome 21, 14 and 1, respectively 6‘9. Of all the AD cases only 5-10% are due to FAD mutations and the mutations in P8] are the most frequent of the FAD causes '0’ ”. Furthermore, apolipoprotein E4 (ApoE4) gene has been shown to cause slight predisposition to AD '2. On the other hand, SAD is the major form of AD and comprises 90-95% of all the cases 10. Unlike FAD, the etiology of SAD is not well understood and several possible risk factors in the development of SAD have been identified. Age is the most significant risk factor for the development of AD. Additional risk factors based upon the epidemiological studies are high fat diet, gender, head trauma and vascular risk factors such as diabetes, ischemia, hypertension, etc. ‘3. AD was first described in 1906 by the German psychiatrist Alois Alzheimer '4. Today, AD affects approximately 5-10% of the population over 65 years of age and more than 20% over the age of 80 years ’5. It represents 40-70% of all dementia cases ’5' 16. There are approximately 5 million AD patients in the US. alone and the total direct/indirect cost associated with the disease is estimated to be more than $140 billion annually [www.alz.org]. With the increasing aging population in the western world, AD has become one of the leading socio-economical challenges today. At the rate of 1 new patient every 72 seconds, the number of AD patients is expected to grow to 13 million by year 2050 and the associated cost will be more than the total US. budget [www.alz.org]. Currently, there is no cure for AD ’7. The available treatments include the use of acetylcholinesterase (AChE) inhibitors (donezepil, rivastigmine and galanthamine) and the N-methyl D-aspartate (NMDA) antagonist, memantine, which have beneficial, but short-lived effects on the symptoms of AD. This emphasizes a significant need for identifying novel targets for therapeutic intervention in AD. 1.2 THE CURRENT HYPOTHESES OF AD AD etiology is very complex and a large number of factors are hypothesized to play key roles in the pathogenesis of AD. Based on the extensive literature search, we present variety of such major scientific hypotheses about the pathogenesis of AD below. In case of each Of these hypotheses, we present supporting data in terms of epidemiological studies as well as various cell culture and animal models. In addition, the limitations associated with these hypotheses are also explained. 1.2.1 The Amyloid Cascade Hypothesis The amyloid hypothesis is the most widely studied hypothesis in AD research field. According to this hypothesis, the different gene defects can lead to altered expression or proteolytic processing of amyloid precursor protein (APP) leading to chronic imbalance between AB production and clearance. This results in the gradual accumulation of AB plaques, which in turn initiates a pathological cascade leading to the gliosis, inflammatory changes, synaptic change, neurofibrillary tangles and neurotransmitter loss”. Many studies support the amyloid cascade hypothesis. The brains of AD patients are characterized by the presence of AB plaques and their number far exceeds that found in the brains of age-matched healthy controls 19. Furthermore, the amount of AB plaques is highly correlated with the degree of cognitive impairment 20. In addition, all four genes associated with FAD have been shown to be involved in increased production of AB (APP, PSI and PS2) 2"28 and its aggregation (ApoE4) 29’ 3° 3'. ApoE4 leads to excessive deposition of AB in the brain long before AD symptoms occur 32. Down's syndrome patients who produce significantly higher amount of AB from birth and deposit AB plaques in their brains as early as age 12, inevitably develop AD by age the of 50 33. This further emphasizes the central role of AB in the pathogensis of AD. In addition, in various cell culture models AB fibrils have been shown to induce neuronal damage and activate inflammatory cells (microglia and astroglia) 34’ 35 36. Also transgenic animal models expressing human APP gene have been shown to develop AB plaques leading to the neuronal and microglial damage. These animal models reproduce most of the major features associated with the AD pathology ”'3’. Despite these data, the amyloid cascade hypothesis falls short in comprehensively interpreting AD pathology. In addition to the increased AB deposition, AD brain is also characterized by the formation of NFTS. In this context, although AB has been Shown to induce hyperphosphorylation of tau and its tangle formation in cell cultures, the APP transgenic mice do not show formation of NFTS in their brain 4°. Also, NFTS have been shown to occur independent of AB plaques“. In addition, it is not clear whether the behavioral deficits observed in APP transgenic animals are related exclusively to the increased AB production and deposition in these animals. Furthermore, the amyloid cascade hypothesis does not provide an explanation for the region-specific damage observed in AD. Finally, suspension of the AB vaccination clinical trial due to the development of encephalitis in a small percentage of individuals further undermined the amyloid cascade hypothesis 42. 1.2.2 The Cholesterol Metabolism and AD Recently, it has been hypothesized that the abnormalities in cholesterol homeostasis may play a central role in causing synaptic impairment, neuronal degeneration and other hallmarks associated with AD 43. The hypothesis is based on the observations that statins which inhibit cholesterol synthesis protect against age-associated dementia and AD 44’ 45. Also, the dietary hypercholesterolemia in APP transgenic mice and rabbits has been shown to accelerate AB deposition in their brains 46' 47. Various cell culture studies further support the involvement of abnormal cholesterol metabolism in AB production “8‘50. In addition to increased AB production, abnormal cholesterol metabolism may lead to neurite degeneration, neuronal cell death, cholinergic dysfunction, oxidative stress and behavioral impairment, thus suggesting its central role in AD pathogenesis 43. The main limitation of the cholesterol hypothesis is that the causal factor behind the abnormal brain cholesterol metabolism is not well established. Furthermore, It is not clear how dietary hypercholesterolemia affects brain cholesterol metabolism as it has been shown that the cholesterol pools in the plasma and the brain are independent of each other; in rats where hypercholesterolemia was induced by diet, all the brain cholesterol was synthesized in situ and did not come from circulation 5'. Furthermore, although cholesterol levels enhance AB production, tau hyperphosphorylation is caused by cellular cholesterol deficiency 52’ 53. Thus, how abnormal cholesterol metabolism may lead to the formation of both the pathophysiological changes associated with AD (AB production and tau hyperphosphorylation) is not clear. 1.2.3 Oxidative Stress and AD Age is the most important risk factor for AD and one of the most accepted theories of aging is increased oxidative stress 54. In this context, it has been hypothesized that the age-associated increase in oxidative stress may play a key role in the pathogenesis of AD. The increased oxidative stress in AD brain can also be attributed to various other factors such as increased levels of metal ions in the brain, inflammatory response from activated microglia and astroglia and increased levels of advanced glycation endproducts (AGES) associated with AD. Iron has been shown to be increased in NFTS as well as in AB plaques and involved in reactive oxygen species (ROS) production 55’ 56. Iron catalyzes the formation of hydroxyl radicals from H202 and thus may lead to lipid peroxidation and oxidation of cellular DNA and proteins. The iron-induced lipid peroxidation is further potentiated by aluminum 57, which also accumulates in neurofibn'llary tangle-containing neurons 58. The activated microglia surround the AB plaques 59 and are a source of NO and oxygen radicals 60, which can react to form peroxynitrite 6'. Furthermore, AGES in the presence of transition metals can undergo redox cycling with consequent ROS production 62‘“. The role of oxidative stress in AD pathogenesis is supported by various studies where free radicals were shown to be involved in increased AB production 65'7" as well as hyperphosphorylation of tau "’75, two hallmarks of AD. Here it is interesting to note that Amyloid-B itself has been directly implicated in ROS formation through peptidyl radicals 76'78. Furthermore, increased oxidative stress has been associated with many other neurodegenerative diseases such as Parkinson's disease and Huntington's disease, which have clinical and pathological features different than AD. Therefore, it is not clear whether oxidative stress plays a causal role in AD pathogenesis or merely a part of the complex AD etiology. 1.2.4 The Cholinergic Hypothesis of AD The cholinergic system in the brain is involved in controlling cerebral blood flow, cortical activity and sleep-wake cycle as well as in modulating cognitive function 79. The severe deficiency in the brain cholinergic system which is associated with the cognitive impairment has been proposed to be a central aspect of AD pathology 80. The strong correlation of clinical dementia ratings with the reductions in the cerebral cholinergic markers such as choline acetyltransferase and levels of acetylcholine support the association of cholinergic dysfunction with AD pathology 3" 82. The direct correlation of dysfunctional cholinergic system and AD pathology is supported by various cell culture and animal model studies that showed a central role of cholinergic system in regulating amyloidogenic processing of APP and hyperphopshorylation of tau, two main characteristics of AD. The activation of muscarinic acetylcholine receptor (M2-mAChR) in SH-SY5Y neuroblastoma cells significantly downregulated level of BACEl, which is involved in amyloidogenic processing of APP 83. Furthermore, selective lesion of basal forebrain cholinergic neurons in rat brain significantly increased AB production and deposition in cortical areas 84’ 85. Also, activation of nicotinic receptors (nAChR) has been 86. 87 shown to decrease amyloidogenic of APP both in cell culture and in vivo 88. In addition, the activation of mAChR has been shown to prevent tau phosphorylation 89‘ 9°. Despite these supporting data, the cholinergic hypothesis has some limitations. It is unclear if the cholinergic dysfunction leads to the AB production or AB leads to the death of cholinergic neurons associated with AD. Furthermore, basal forebrain cholinergic neuronal loss is not specific to AD and is also associated with many diseases e.g. Parkinson’s disease, Parkinsonism with dementing complex of Guam, Pick’s disease, Jakob-Creutzfeld disease etc. 79. 1.2.5 Homocysteine and AD The elevated plasma level of homcysteine (homocysteinuria) has been proposed to be an independent risk factor for the development of AD 9’. The level of total homocysteine is significantly higher in serum of AD patients compared to healthy subjects 92‘9” . The role of homocysteine in AD pathology is further emphasized by the cell culture studies where homocysteine caused oxidative stress, tau hyperphosphorylation and apoptosis in neurons 99' 100. There is a significant positive correlation between homocysteine and 4- hydroxynonenal (4-HNE), a lipid peroxidation product, in AD ’0'. Furthermore, BACEl and PS], two important enzymes involved in AB production have been shown to be regulated by methylation. In context to this, increased homocysteine levels (caused by the reduction of folate and vitamin BIZ in culture medium) cause a reduction of s- adenosylrnethionine, the universal methyl-group donor, thus consequently increasing PSI and BACEl levels '02. In this context, a positive correlation between elevated levels of homocysteine and AB40 has been established in AD '03. In addition to these supportive data, there are many studies that oppose any key role of homocysteine in AD. It has been observed that although plasma homocysteine levels were higher in AD cases than controls, this difference was not significant and homocysteine levels were not related to cognitive status ’04 . This was further supported by another study, which showed that high homocysteine levels were not associated with AD and were not related to a decrease in memory scores over time '05. Furthermore, homocysteine has been shown to potentiate copper-induced oxidative stress in primary mouse neurons, but homocysteine alone had no effect '“.Thus, it is clear that the area of homocysteine and AD needs to be further studied. We need additional, long-term studies using a variety of populations to determine if elevated homocysteine level is a significant and consistent risk factor for AD. 1.2.6 The Pathogen Hypothesis of AD The pathogen hypothesis proposes a potential role of microbes such as herpes simplex virus 1 (HSVl) and Chlamydophila pneumoniae (Cp) in the pathogenesis of AD. The pathogens have been accepted to play central role in causing many diseases which earlier had been thought to be non-infectious; Helicobacter pylori has been shown to cause duodenal ulcers and gastric cancers ’07, which previously were thought of as the result of stress, chemical irritants and genetic mutations. Also, human papillomavirus (HPV) virus has been accepted to cause virtually all cases of cervical cancer '08. Furthermore, C. pneumoniae has been recently suggested to play a role in atherosclerosis 109' ”0. In light of these data, proponents of the pathogen hypothesis suggest its serious consideration in the case of AD, given the fact that the dominant hypothesis in the AD field (the amyloid 10 cascade hypothesis) remains open to many questions. Interestingly, although various studies showed the presence of HSVl and Cp in brains of AD patients “H” , others failed to confirm these data “6"”. These contradictory results may be attributed to the fact that both HSVl and Cp are highly challenging to detect. Recently, an infection-based animal model showed that intranasal inoculation of mice with Cp results in the formation of amyloid plaques in their brain 120, thus further supporting the pathogen hypothesis of AD. However, these data await further independent replication. 1.3 POTENTIAL INVOLVEMENT OF STURATED FATTY ACIDS IN THE PATHOGENESIS OF AD Epidemiological studies suggest that high fat diets significantly increase the risk of AD and the degree of saturation of fatty acids is critical in determining the risk for AD ’2'. This notion is further supported by various in vivo studies where mice fed a western, high fat (21-40%)-high cholesterol (0.15-1%) diet developed AD—like pathophysiological changes in their brain ”242" . Diets rich in saturated fats may increase brain uptake of intact free fatty acids (FF As) from the plasma through the blood brain barrier (BBB) 125; the BBB is not a barrier for fatty acids ’26. Here, it is interesting to note that diabetes mellitus, which is a significant risk factor for AD 127 is characterized by elevated plasma levels of saturated FFAS '28. Due to the interaction between the FFA pools in the plasma and the brain 125’ ’26’ ’29, diabetes-associated increases in plasma FFAS may affect the level of F FAs in the brain and in turn increase the risk for AD. Likewise, traumatic brain injury, which has been established as an independent risk factor for AD '30 is associated with elevated levels of pahnitic, stearic and arachidonic acids in the brain '3’. Following 11 traumatic brain injury, palmitic acid increases from ~ 60 to 180 M and stearic acid from ~ 50 to 350 M ’32. In addition, the fatty acid profile of NFTS in AD brain has been shown to be rich in palmitic and stearic fatty acids ’33. Similarly, the white matter in AD brain is characterized by high fatty acid content ’34. Finally, apolipoprotein E4 (ApoE4) is an important genetic risk factor for AD and its risk may be further increased by a hyperlipidemic life-style 135. Despite these accumulating data, the basic mechanism of how elevated levels of fatty acids are involved in the pathogenesis of AD is unclear. 1.4 GOALS OF THE PRESENT STUDY The present study was undertaken with the following aims: I) investigate the possible involvement of saturated fatty acids in causing the pathophysiological and metabolic changes associated with AD; 2) establish the basic mechanism behind these potential, fatty acid-induced abnormalities; and 3) understand the causal interrelation between the neuropathological and metabolic changes. These data may be useful in identifying the possible causal role of elevated levels of saturated fatty acids in the pathogenesis of AD and may further help in identifying novel therapeutic targets. 1.5 THESIS OUTLINE In line with the main focus of the present work to investigate a possible causal link between saturated FFAS and AD-associated abnormalities, this dissertation is subdivided as follows: Chapter 2 presents results describing the involvement of saturated FFAS in causing oxidative stress and hyperphosphorylation of tan in neurons through astroglial mediation. Chapter 3 describes the role of saturated FFAS in causing amyloidogenic 12 processing of amyloid precursor protein (APP) through astroglia-mediated oxidative stress. In Chapter 4, FFA-induced abnormal glucose metabolism is studied. Metabolic flux analysis (MFA) is applied to obtain a comprehensive picture of the global metabolic changes caused by FFA treatment. This led to the identification of abnormal astroglial sphingolipid metabolism as a pathway of interest in relation to the FFA-induced pathophysiological changes observed in neurons. Chapter 5 further establishes the causal role of astroglial ceramide in the FFA-induced, AD-associated pathophysiological abnormalities in neurons. Chapter 6 presents the conclusions based on the present study and future research directions. 13 CHAPTER 2. SATURATED FATTY ACID-INDUCED HYPERPHOSPHORYLATION OF TAU IN PRIMARY RAT CORTICAL NEURONS 2.1 INTRODUCTION Neurofibrillary tangles (NFTS) are one of the classical neuropathological hallmarks of AD brain 3. NFTS have been suggested to play a central role in AD pathogenesis since they have been shown to correlate with the severity of dementia. According to a proposed model of pathological evolution of AD, NFTS appear first in the entorhinal cortex during the preclinical phase, spreading to the hippocampus in the middle phase and, eventually, to the neocortex during the late stage of AD ’36. This strong correlation between the number of NFTS and the disease severity was further supported by many other studies 137’ ’38. NFTS are composed of paired helical filaments of tau protein, which is hyperphosphorylated in AD '39. Physiologically, tan is one of the major microtubule- associated proteins that stabilize the microtubules, which play important structural and functional roles in the neurons (Figure 2.1A). Tau is involved in signal transduction 140’ 14', anchoring various protein kinases and phosphatases 142' ”3 and interacting with the actin cytoskeleton ”4. Six isoforrns of tau exist in the central nervous system, which are derived from a single gene and vary between having 3 or 4 microtubule-binding repeat domains and in the number and size of N-terminal inserts 145. Interactions between tau and microtubules are regulated by the length and more importantly, phosphorylation of the microtubule-binding repeat domains 146. Tau has about 30 possible phosphorylation sites '47 and in its phosphorylated form cannot stabilize microtubules 148. In this context, tan is phosphorylated to a degree of ~8 Pi/mol in AD as compared to ~2 Pi/mol for 14 normal tau and this hyperphosphorylation of tau in AD leads to the disruption of the cytoskeleton of neurons, which in turn leads to their degeneration, thereby playing an important role in AD pathology (Figure 2.13) 3. (A) Control Disintegrating Mierotubule :‘7 Diseased “i Neuron (B) AD Disintegrating Microtubules‘ ‘ .4 Figure 2.1. The physiology and pathology of tau. (A) In healthy neurons, tau silzes mcrules. (B) In AD, tau hyperphosphorylates and detaches from microtubules leading to tangle formation (figure taken from www.alz.org). The hyperphosphorylation of tau has been proposed to be critical in promoting aggregation of tau in NFTS 149. The hyperphosphorylation of tau has been shown to precede the formation of NFTS in degenerating neurons 150’ ’5'. The importance of hyperphosphorylation of tau in its aggregation is further emphasized by additional studies that showed that the aggregation of tau depends on the degree of its phosphorylation and in vitro dephosphorylation of phosphorylated-tau from AD brain prevents its aggregation ’52. Furthermore, in human neuroblastoma cells phosphorylated but not the unphosphorylated tau has been Shown to form tau filaments '53. The hyperphosphorylation of tau has been suggested to be due to the imbalance between the activities of protein kinases and phosphatases although the actual mechanism is still not well understood. The kinases involved in tau phosphorylation are divided into two groups, proline-directed protein kinases (PDPKs) and non-PDPKs 15". The PDPKs include glycogen synthase kinase 3 (GSK—3), cycline-dependent kinase 5 (cdk5) and mitogen activated protein kinases (MAPKS). The non-PDPKS include protein kinase C (PKC), protein kinase A (PKA) and Ca2+/calrnodulin-dependent kinase II (CaMKII). Different kinases may phosphorylate tau at different amino acid residues with some overlapping of sites. The phosphatases involved in dephosphorylation of tau are divided into two major groups, S or T site protein phosphatases and protein tyrosine phosphatases, of which PP-l, PP-2A and PP-2B dephosphorylate tau with a certain degree of overlap in sites ’5 5 '157. AD brain has been characterized by decreased activity of 158 phosphatases and increased activity of various kinases resulting in hyperphoshorylation of tau "’8’ ”9"“. Increased activity of these tau phosphorylating l6 kinases in AD has been suggested to be a direct result of increased oxidative stress '62. Increased oxidative stress, manifested by increased lipid peroxidation and protein 163,164 oxidation is one of the major and earliest characteristics of AD brain 73' ’65. Lipid peroxidation is an important aspect of AD brain and has been shown to be elevated in the regions of brain affected in AD '66' ‘67 . There is a strong correlation between thiobarbituric acid reactive substances (TBARS), an indicator of lipid peroxidation, and the presence of NFTS in AD brain ’66. Similarly, the levels of 4-hydroxynonenal (4-HNE) and acrolein have been shown to be elevated in AD brain "’8' ’69. Furthermore, protein oxidation is an inevitable aspect of aging and age-related neurodegenerative diseases 76' '70. Protein oxidation is Significantly increased in the cortex and hippocampus of AD brain as compared to age-matched healthy controls ”"172. In addition to increasing the activity of various stress-dependent kinases, which are involved in hyperphosphorylating tau thus leading to its aggregation, oxidative stress in itself has been shown to be involved in forming tau dimers, which are the building blocks of paired helical filaments (PHFs) in NFTS. The aggregation of tau is increased when tau molecules crosslink into dimers by an oxidized disulfide bond at Cys322 173’ ’74. Furthermore, 4-HNE, which has been shown to co-localize with NFTS in AD brain “’5’ ”’9' ’75, can act as an adduct to phosphorylated tau and enhance tau filament formation '53. In summary, oxidative stress and hyperphosphorylation are important pathological events in the formation of tau aggregates, which are one of the characteristics signatures of AD brain. Therefore, the aim of the current study was to investigate possible involvement of saturated fatty acids in causing oxidative stress and hyperphosphorylation of tau. l7 2.2 MATERIALS AND METHODS 2.2.1 Isolation and Culture of Primary Rat Cortical Neurons and Astroglia Primary cortical neurons were isolated from one-day-old Sprague-Dawley rat pups and cultured according to the published methods as described in Chandler et al. ’76. The cells were plated on poly-D-Lysine coated, 6-well plates at the concentration of 2 x 106 cells per well in fresh cortical medium [Dulbecco's Modified Eagle's Medium (DMEM and all other media are from Invitrogen, CA) supplemented with 10% horse serum (Sigma, MO), 25 mM glucose, 10 mM HEPES (Sigma), 2 mM glutamine (BioSource International, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycine]. Three days after incubation (37°C, 5% C02), the medium was subsequently replaced with 2 ml of cortical medium supplemented with 5 uM cytosine-B-arabinofuranoside (Arac, from Calbiochem, CA). After 2 days, the neuronal culture was switched back to cortical medium without Arac. The experiments were performed on 6-7 day old culture. To obtain primary cultures of astroglial cells, the cortical cells from one-day-old Sprague-Dawley rat pups were cultured in DMEM/Ham’s F12 medium (1:1), 10% fetal bovine serum (Biomeda, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycin 177. The cells were plated on poly-D- Lysine coated, 6-well plates at the concentration of 2 x 106 cells per well. Cells were grown for 8-10 days (37°C, 5% C02) and culture medium was changed every 2 days. 24 hours prior to treatment with fatty acids, the medium was changed to neuronal cell culture medium. 2.2.2 Lactate Dehydrogenase (LDH) Assay The secreted and intracellular levels of LDH were measured to determine the level of cell toxicity in astroglial cells by using cytotoxicity detection kit (Roche, IN, USA). The 18 cytotoxicity was determined as the fraction of LDH released into the medium, normalized to the total LDH (released + intracellular), as shown in the equation below-. LDH (medium) %LDH release = X 100 LDH (medium+intracellular) 2.3 Western Blot Analysis For western blot analysis, cells were washed three times with ice-cold TBS (25 mM Tris, pH 8.0, 140 mM NaCl, and 5 mM KCl) and lysed for 20 minutes by scraping into ice-cold radioimmunoprecipitation assay (RIPA) buffer [1% (v/v) Triton, 0.1% (w/v) SDS, 0.5% (w/v) deoxycholate, 20 mM Tris, pH 7.4, 150 mM NaCl, 100 mM NaF, 1 mM Na3VO4, 1 mM EDTA, 1 mM EGTA, and 1 mM PMSF, all chemicals from Sigma] ’7’. The total cell lysate was obtained by centrifugation at 12,000 rpm for 15 minutes at 4 0C. The total protein concentration was measured by BCA protein assay kit from Pierce (Rockford, IL). Equal amounts of total protein from each condition were run at 200 V on 10% SDS- PAGE gels (BioRad, CA) for phosphorylated tau and actin. The separated proteins were transferred to nitrocellulose membranes for 1 hour at 100 V and incubated at 4 0C overnight with the appropriate primary antibodies [1:200 PHF-l (from Dr. P. Davies, Albert Einstein, NY), 1:200 AT8 (Pierce Biotechnology, IL), 122000 actin (Sigma, MO)]. Blots were washed three times in PBS-Tween (PBS-T) and incubated with appropriate HRP-linked secondary antibodies (Pierce Biotechnology, IL) diluted in PBS-T for 1 hr. After an additional three washes in PBS-T, blots were developed with the Pierce SuperSignal West Femto Maximum Sensitivity Substrate (Pierce Biotechnology) and imaged with the BioRad ChemiDoc. Quantity One software from Bio-Rad was used to quantify the signal intensity of the protein bands. 19 2.2.4 Immunofluorescence Analysis of Neurons and Astroglia To perform confocal immunofluorescence microscopy study, neurons and astroglia cultures were fixed for 20 minutes in 4% paraforrnaldehyde and permeabilized with 0.1% Triton X-100 and 5% goat serum (Invitrogen) in PBS. Cells were then labeled overnight at 4 0C with appropriate primary antibodies [1:50 MAP-2 (Santa Cruz Biotechnology, CA) for neurons, 1:1000 GFAP (Dako, CA) for astroglia and 1:200 AT8 for hyperphosphorylated tan] in 5% goat serum in PBS. After three PBS washes, primary antibodies were detected with rhodamine conjugated (Chemicon, CA) or Alexa Flour 594 conjugated (Molecular Probes, OR) secondary antibodies. The cells were visualized with the confocal microscope Zeiss LSM 5 Pascal (Carl Zeiss, Jena, Germany) using a 40X (for MAP-2 and GFAP) or 63x (for AT8) oil-immersion objective lens. 2.2.5 Immunostaining of Reactive Oxygen Species (ROS) Intracellular reactive oxygen species (ROS) were detected by staining with the oxidant- sensitive dye 5-(6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate (CM- HzDCFDA, from Molecular Probes, OR). HzDCFDA is cleaved of its ester groups by intracellular esterases and converted into membrane impermeable, nonfluorescent derivative HzDCF. Oxidation of HzDCF by ROS results in highly fluorescent 2,7- dichlorofluorescein (DCF) '79. The cells were incubated for 30 minutes at 37 0C with 2 uM CM-HzDCFDA in Hanks’ Balanced Salt Solution without phenol red (Invitrogen). The cells were then washed three times with PBS and analyzed with confocal microscopy. A 63X oil-immersion objective lens was used for data acquisition. 20 2.2.6 Data Analyses Data are shown as means : SD. for indicated number of experiments. Student’s t-test and one-way ANOVA with Tukey’s post hoc method were used to evaluate statistical significances between different treatment groups. Statistical significance was set at p<0.05. 2.3 RESULTS AND DISCUSSION 2.3.11 Direct Treatment of Neurons with Saturated Fatty Acids Although tan is present in astroglia and oligodendrocytes in the brain and in some peripheral tissues, it is mainly synthesized by neurons "’0’ ’8'. Tau is mainly located in neuronal axons and hyperphosphorylated tau is deposited in dystrophic neurites and neuronal bodies in the form of NFTS. Therefore, to examine the possible involvement of saturated FFAS in the hyperphosphorylation of tau, primary rat cortical neurons were left untreated or treated with 0.2 mM of palmitic acid (PA) or stearic acid (SA) for 24 hours. After 24 hours, the cells were lysed and western blot analysis was performed to determine the cellular levels of hyperphosphorylated tau. There was no change in the levels of phosphorylated tan in rat cortical neurons treated directly with pahnitic or stearic acid, as compared to controls (Figure 2.2). Furthermore, the morphology of the neurons was not affected by the FFA treatment, as shown by MAP-2 immunostaining (Figure 2.3A). The observed lack of F FA effect on neurons may be attributed to the low capacity of primary neurons to take up and metabolize saturated fatty acids ’82. 21 Previously, primary rat cortical neurons have been shown to possess a very low capacity to take up PA and incorporate it into glycerolipids and sphingolipids '82. Control PA SA PHF-1 -~- AT8 it” “he—a In... Is-actin m Figure 2.2. Direct treatment of neurons with saturated FFAs. Primary rat cortical neurons were treated for 24 hours with 0.2 mM of palmitic acid (PA) or stearic acid (SA) or with 5% bovine serum albumin (BSA), vehicle for FFAS (control). Detergent cell lysates from fatty acid-treated and control cells were immunoblotted with PHF-l and AT8 antibodies, which recognize phosphorylated tau. B-actin is shown as a marker for protein loading. 2.3.2 Involvement of Saturated FFAS in Tau Hyperphosphorylation Through Astroglial Mediation Compared to primary neurons, primary astroglia possess a significantly higher capacity to 182 utilize saturated fatty acids . The uptake and incorporation of PA into glycerolipids and sphingolipids have been Shown to be more than 3 times higher in primary rat cortical astroglia as compared to primary rat cortical neurons '82. Therefore, we treated cortical astroglia with 0.2 mM of PA or SA for 12 hours and transferred the conditioned media to treat the cortical neurons for 24 hours. The morphologies of the fatty acid-treated astroglia and neurons are shown using GFAP and MAP-2 immunostaining, respectively (Figures 2.38 and C). No significant change in the cell morphologies was observed, except, the characteristic dotted MAP-2 labeling of the neurites was less significant in the 22 neurons treated with conditioned media from FFA-treated astroglia as compared to controls. (A) MAP-2 Figure 2.3. MAP-2, GFAP and AT8 immunostaining. (A) Neurons treated directly with FFAS l'or 24 hours (B) Astrocytes treated for 12 hours with 0.2 mM of either palmitic acid (PA) or stearic acid (SA) or 5% BSA (control). (C) Neurons treated for 24 hours with conditioned media from fatty acid-treated or control astrocytes. (D) Immunot‘luorcsccncc labeling of phosphorylated tau with AT8 antibody in neurons treated with conditioned media from fully acid-treated or untreated astrocytes (control). Images were obtained with confocal fluorescence microscopy. (Objective lens magnification- 40X l‘or MAP-2 and GFAP, 63X for AT8).. Furthermore, treatment of astroglia with 0.2mM of PA did not affect the astroglial cell viability compared to controls as measured by % LDH release (Figure 2.4). 300 IIM H202 treatment was used as a positive control and significantly increased LDH release 23 from astroglia (Figure 2.4), which is in accordance with a previous study in the literature 183 50 * 451 40- 35- 30- ’75.. 20- 151 10- 5.. 0+ %LDH released Control PA 300uM H202 Figure 2.4. Measurement of LDH release from astroglia treated with PA. 24hr treatment with 0.2mM PA failed to liberate LDH from astroglia as compared to controls. In contrast, 1hr treatment of astroglia with 300 uM H202 (positive control) induced significant LDH liberation after 24h as compared to both control and PA-treated cells. Data are taken from 3 different experiments and are expressed as mean i S.D. One-way ANOVA with T‘ukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control. The conditioned media from FFA-treated astroglia caused significant hyperphosphorylation of tan in cortical neurons, as observed by immunostaining with AT8 (Figure 2.3D) and immunoblotting with AT8 and PHF-l antibodies Gilgure 2.5). AT8 and PHF-l antibodies recognize tau protein hyperphosphorylated at AD-specific 184 phospho-epitopes; AT8 recognizes tau phosphorylated at Ser202 and Thr205 , while PHF-l is specific for tau phosphorylated at Ser396 and Ser404 ”‘5. Ser202, Ser396 and 24 Ser404 are three of the nine abnormal phosphorylation sites of the hyperphosphorylated tau associated with NFTS in AD '36. (A) C PA SA PA+NAC SA+NAC PHF-l - u ,- - AT8 “"“ ~ - — -1 Actin (B) Pbospho-Tau (% of control) PA+NAC SA+NAC 400‘ AT8 350. _ . m. Phospho—Tau 250 i (% of control) 200 1 # # 150 ‘ 100 . 50 J 0 C PA SA PA+NAC SA+NAC Figure 2.5. Astroglia-mediated, fatty acid-induced hyperphosphorylation of tau in neurons. (A) Western blot analysis of hyperphosphorylated tau was performed using phospho—specific antibodies PHF-l and AT8. (B) Histograms corresponding to PHF-l and AT8 blots represent quantitative determinations of intensities of the relevant bands. Data represent mean in SD. of 3 independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. ‘, p<0.05 compared with control; #, p<0.05 compared with fatty acid treatment. 25 2.3.3 Involvement of Oxidative Stress in FFA-Astroglia-Induced Tau Hyperphopshorylation in Neurons As shown in Figure 2.6A, treatment of astroglia with 0.2mM of PA or SA did not cause any ROS production in cortical astroglia. On the other hand, we found that intracellular levels of ROS were elevated in the neurons treated with conditioned media from FFA- treated astroglia as compared to controls (Figure 2.68). (A) Control (3) control PA+NAC Figure 2.6. Intracellular accumulation of ROS in neurons. (A) Astroglia and (B) neurons were stained with CM-HZDCFDA for intracellular ROS detection and examined with confocal fluorescence microscopy. (Objective lens magnification, 63X). 26 Previously, various in vitro and in vivo studies have implicated increased oxidative stress in the hyperphosphorylation of tau 7” 72. 4-HNE and acrolein, the lipid peroxidation products found elevated in AD brain 73, have been shown to induce hyperphosphorylation 74, 75 of tan in neurons . Furthermore, although acute administration of a high concentration of H202 (lmM for 1 hr) decreases tau phosphorylation '87 ; chronic exposure of a low concentration of H202 (10 [AM for 24, 48 or 72 hrs), more relevant to AD, increases tau phosphorylation in primary rat cortical neurons (Mark Smith and Xiongwei Zhu, personal communication). Therefore, we treated neurons with lOmM N- acetyl cysteine (NAC), an anti-oxidant. The co-treatment of neurons with NAC inhibited the observed FFA-astroglia-induced increase in ROS levels (Figure 2.68) as well as tau hyperphosphorylation in neurons (Figure 2.5). 2.4 CONCLUSIONS In conclusion, saturated F F As had no direct effect on the neuronal morphology and the level of phosphorylated tau in neurons. In addition, saturated FFAS had no effect on astroglial morphology and viability. However, the conditioned media from FFA-treated astroglia affected the classical dotted MAP-2 labeling of the neuronal axons and also increased the levels of phosphorylated tan in neurons. Furthermore, there was a significant increase in the ROS production in neurons treated with conditioned media from FFA-treated astroglia, without any increase in astroglial ROS levels. The elevated levels of ROS in the neurons were found to be involved in the observed, FFA-astroglia- induced hyperphosphorylation of tan in the neurons. Thus, the present results establish a central role of saturated FFAS in causing hyperphosphorylation of tau in neurons through astroglia-mediated oxidative stress. 27 CHAPTER 3. SATURATED FATTY ACID-INDUCED AMYLOIDOGENIC PROCESSING OF APP IN PRIMARY RAT CORTICAL NEURONS 3.1 INTRODUCTION The “amyloid cascade hypothesis”, which suggests the accumulation of aggregated amyloid beta (AB) in the brain as a main trigger for AD, has been extensively studied since the first characterization of AB deposits in 1984 “’8. According to this hypothesis, a chronic imbalance between the production and clearance of AB results in the formation of AB plaques, which leads to a multi-step cascade including reactive gliosis, inflammatory changes, synaptic change and transmitter loss 2" ”9"”. In AD brain, two major types of AB plaques are observed, diffuse plaques and neuritic plaques. Diffuse plaques mainly consist of nonfibrillar AB, while neuritic plaques are more developed consisting of dense AB fibrils together with degenerating dendrites and axons, serum amyloid P, (11- antichymotrypsin, al-antitrypsin, sulphated glycosaminoglycans, apolipoproteins E and D, and the neurotrophic factor midkine ”3"”. Recently, the soluble AB intermediates have been shown to play a more important role in AD pathogenesis as compared to the mature neuritic plaques '90' '96' '97. These soluble AB oligomers, AB protofibrils and AB- derived diffusible ligands (ADDLS) cause synaptic dysfunction, which have been suggested to be an early event in AD-associated memory loss '98. AB is generated from the proteolytic processing of amyloid precursor protein (APP). APP is an integral type I membrane glycoprotein of 110-120kDa in size and contains a large amino terminal extracellular domain and a small COOH-terrninal intracellular domain "’9‘ 28 20°. APP has three major isoforms containing 695, 751 or 770 amino acids. The APP 69, isoform is mostly present in neurons, while the others are present in peripheral and glial cells 20'. APP m and APP 770 have serine protease inhibitor domain called Kunitz protease inhibitor domain, while APP ,9, lacks this domain. The physiological functions of APP include neuronal survival, cell adhesion, axonal adhesion, neuritic outgrowth, synaptic plasticity and signaling "202. The proteolytic processing of APP takes place by sequential cleavage by proteases named 01-, B- and y-secretase (Figure 3.1A). The a-secretase is a member of the ADAM (a disintegrin and metalloprotease) family such as ADAM17 or TACE (tumor necrosis factor-a converting enzyme), ADAM 9, ADAMIO, MDC9 and an aspartyl protease BACE2 20’. The ct—secretase cleavage of APP may occur at the cell surface, within calveolae or in the trans-Golgi compartment "’0' 20". The a-secretase has been shown mainly to be active in non-raft regions of the membrane 2°" 20". The a-secretase cleaves APP within AB domain (shown as red) between residues Lysl6 and Leul7, thus avoiding the generation of intact AB peptides. It leads to the formation of a soluble domain (SAPPa), which is released into extracellular space and a lO-kDa C-tenninal fragment (C83), which remains within the cellular membrane 207. The B-secretase, also called BACE (B-site of APP cleaving enzyme), Asp-2 or memapsin-2 is a trans-membrane protein and an aspartic-acid protease 20“”. BACE contains aspartate residues in its extracellular protein domain which are involved in BACE activity 20”. 29 AMA MR c... / K APPSB M if" tit i Liideaft (A) (B) Flgu3re ”lProcmingo ofa amylororidpecurs pro i (APP)()Th n-aomylioesdge cawpth ay catalyz edbyu -andy- steecreas .e(B)Th eoioenamyldg icpatwh ayc atazely zedbyBoa an-sedy setacre 30 BACE] is a major B-secreatse involved in the amyloidogenic processing of APP in neurons 2”. Cleavage of APP by B-secretase may occur in endosomes and trans-Golgi compartments 212-214 , which provide an acidic environment that has been shown to be critical for maximal activity of BACE 2". The B-secretase has been shown mainly to be active in lipid raft regions of the membrane 206' 2'6' 2”. The B-secretase cleaves APP at the Asp+1 residue of the AB region and leads to the generation of a secreted soluble fragment (SAPPB) and a membrane-bound C-terminal fragment (C99). Both the a-secretase product of APP (C83) and the B-secretase product of APP (C99) act as immediate substrates for y-secretase. The y-secretase is a membrane-bound complex of at least four enzymes including components such as Presenilin 1 and 2 (PSI and PS2), Nicastiin (th), anterior pharynx-defective phenotype (APH-l) and Presenilin enhancer (PEN-2) 2". The PS1 C-terminal tail (PSl-CTF) is critical for y-secretase activity “9‘ 22°. The y —secretase activity resides in various cellular compartments such as the ER, late- Golgi/trans-Golgi, endosomes and the plasma membrane ”"22". Similar to the B- secretase, the y-secretase has been associated with lipid raft microdomains of the membrane 22’. The y-secretase cleavage of ‘C83 is a non-amyloidogenic pathway, which leads to the generation of a short peptide (p3) containing the C-terminal domain of the AB peptide. The physiological or pathological significance of p3 remains unclear. On the other hand, the y-secretase cleavage of C99 is an amyloidogenic pathway, which leads to the generation of a spectrum of AB peptides. The AB peptides containing 40 or 42 amino acids (AB40/42) are the two most common amyloidogenic AB peptides. Both AB40 and AB42 are produced during normal cellular metabolism but the production of AB42 is 31 considered to be elevated in AD 226' 227. AB42 is more prone to aggregation as compared to AB40 228‘ 229. AB42 initially forms non-filamentous, diffuse plaques onto which AB40 starts to aggregate, which leads to the mature, neuritic plaques. The importance of amyloidogenic processing of APP in AD is emphasized by the involvement of mutations in the APP, presenilin-l (PSI) and presenilin-2 (P82) genes, localized on chromosome 21, 14 and 1, respectively, in FAD ”0'23". These APP and PS mutations alter APP processing leading to the pathological increase in the production of total AB or AB42 which is highly fibrilogenic “'2" “’9. In APP mutations linked to FAD, clinical and pathological symptoms are identical to those of the late-onset sporadic AD, which consists of more than 95% of total AD cases. This strongly suggests that amyloidogenic processing of APP plays a central role not only in FAD but also in sporadic AD. Furthermore, ApoE4, a genetic risk factor for AD, has been shown to play an important role in the production and clearance of AB 2’7. This further suggests that amyloidogenic processing of APP is a central pathological event in AD pathology. In this context, the aim of the current study was to investigate possible involvement of saturated fatty acids in causing amyloidogenic processing of APP, potentially by affecting the levels and/or activities of B- and y-secretases. 32 3.2 MATERIALS AND METHODS 3.2.1 Isolation and Culture of Primary Rat Cortical Neurons and Astroglia Primary neurons and astroglia were isolated from the cortex of one-day-old Sprague— Dawley rat pups and cultured according to the methods as described in chapter 2. The cells were plated on poly-L-Lysine coated, 6-well plates at the concentration of 2 x 10'5 cells per well in fresh cortical medium [Dulbecco's Modified Eagle's Medium (DMEM and all other media are from Invitrogen, CA) supplemented with 10% horse serum (Sigma, MO), 25 mM glucose, 10 mM HEPES (Sigma), 2 mM glutamine (BioSource International, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycin] ’76. To obtain pure neuronal cell cultures, after 3 days of incubation (37°C, 5% C02) the medium was replaced with the cortical medium supplemented with 5 uM cytosine-B-arabinofuranoside (Arac, from Calbiochem, CA). After 2 days of Arac treatment, the neuronal culture was switched back to cortical medium without Arac. The neuronal cell culture of more than 95% was obtained by this procedure. The experiments were performed on 6-7 day old neuronal culture. To obtain primary cultures of astroglial cells, the cortical cells from one-day-old Sprague-Dawley rat pups were cultured in DMEM/Ham’s F12 medium (1:1), 10% fetal bovine serum (Biomeda, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycine '77. Cells were grown for 8-10 days (37°C, 5% C02) and culture medium was changed every 2 days. The astroglial cell culture of more than 95% was obtained by this procedure. 24 hours prior to treatment with fatty acids, the medium was changed to neuronal cell culture medium. 33 3.2.2 Western Blot Analysis For western blot analysis, cells were washed three times with ice-cold TBS (25 mM Tris, pH 8.0, 140 mM NaCl, and 5 mM KCl) and lysed for 20 minutes by scraping into scraping into ice-cold radioimmunoprecipitation assay (RIPA) buffer [1% (v/v) Nonidet P-40, 0.1% (w/v) SDS, 0.5% (w/v) deoxycholate, 50 mM Tris, pH 7.2, 150 mM NaCl, 1 mM Na3VO4 and lmM PMSF, all chemicals from Sigma] 238. The total cell lysate was obtained by centrifugation at 12,000 rpm for 15 minutes at 4 0C. The total protein concentration was measured by BCA protein assay kit from Pierce (Rockford, IL). Equal amounts of total protein from each condition were run at 200 V on 10% Tris-HCl gels (for BACEl , actin), 12% Tris-HCl gels (for PS1) and 10-20% Tris-Tricine gels (for APP, C99). The separated proteins were transferred to nitrocellulose membranes for 1 hour at 100 V and incubated at 4 0C overnight with the appropriate primary antibodies [1:1000 BACE] (chemicon, CA), 1:1000 PS1 (Calbiochem, CA), 1:2000 actin (Sigma, MO), 1:1000 APP/C99 (QED Bioscience Inc, CA)]. Blots were washed three times in PBS- Tween (PBS-T) and incubated with appropriate HRP-linked secondary antibodies (Pierce Biotechnology, IL) diluted in PBS-T for 1 hr. After an additional three washes in PBS-T, blots were developed with the Pierce SuperSignal West Femto Maximum Sensitivity Substrate (Pierce Biotechnology) and imaged with the BioRad ChemiDoc. Quantity One software from Bio-Rad was used to quantify the signal intensity of the protein bands. 3.2.3 Immunofluorescence Analysis of BACEl in Neurons To perform confocal immunofluorescence microscopy study, neuronal cultures were fixed for 20 min in 4% paraforrnaldehyde and permeabilized with O. 1% Triton X-100 and 34 5% goat serum (Invitrogen) in PBS. Cells were then labeled overnight at 4 0C with the primary antibody [1:100, BACEl] in 5% goat serum in PBS. After three PBS washes, primary antibody was detected with Alexa Flour 594 conjugated (Molecular Probes, OR) secondary antibody. The cells were visualized with confocal microscope Zeiss LSM 5 Pascal (Carl Zeiss, Jena, Germany) using a 40x oil-immersion objective lens. 3.2.4 Data Analyses Data are shown as means :9: SD. for indicated number of experiments. Student’s t-test and one-way ANOVA with Tukey’s post hoc method were used to evaluate statistical significances between different treatment groups. Statistical significance was set at p<0.05. ‘ 3.3 RESULTS 3.3.1 Direct Treatment of Neurons with Saturated Fatty acid (Palmitic Acid) The BACEl enzyme involved in the first step of amyloidogenic processing of APP has been localized in the brain, mainly in the neurons, thus suggesting that neurons are the prominent source of AB peptides in the brain 239. The BACEl levels have been shown to 240. Therefore, to be significantly elevated in AD brain as compared to healthy controls examine the possible effect of saturated FFAS on the BACE] levels, primary rat cortical neurons were left untreated or treated with 0.2mM of palmitic acid (PA) for 24 hours. In this and all subsequent studies, we treated cells with only PA, since both PA and SA had similar effects as demonstrated in chapter 2. Also, in typical high-fat American diets, 35 more than 60% of the saturated fat is PA, while SA contributes only 25% of the dietary energy derived from saturated fats 2‘“. After 24 hrs of direct treatment with PA, the cells were lysed and western blot analysis was performed to determine the cellular levels of BACE]. There was no change in the BACEl levels in primary rat cortical neurons treated directly with PA as compared to controls (Figure 3.2). This lack of FFA effect on neurons may be attributed to the low capacity of primary neurons to take up and metabolize saturated fatty acids '82. This effect is similar to that shown in Chapter 2, where saturated FFAS had no direct effect on neurons; there was no change in the levels of phosphorylated tan in rat cortical neurons treated directly with both PA and SA. BPCEI “'""‘"" M —70Koa Actin ‘- ~—42 KDa Control PA Figure 3.2. Direct treatment of neurons with palmitic acid. Primary rat cortical neurons were treated for 24 hours with 0.2mM of palmitic acid (PA) or with 4% bovine serum albumin (BSA), vehicle for PA (control). Detergent cell lysates from PA-treated and control cells were immunoblotted with BACE] antibody. B—actin is shown as a marker for protein loading. The blots are representative of 3 independent experiments. 3.3.2 Involvement of Saturated FFAS in BACE] Up-regulation and Amyloidogenic Processing of APP Through Astroglial Mediation As mentioned earlier in Chapter 2, primary astroglia possess a significantly higher 182 capacity (>3 times) to take up and utilize saturated fatty acids and the conditioned media from FFA-treated astroglia significantly increased phosphorylation of tau in 36 neurons. Therefore, in this study, we first cultured the rat cortical astroglia with 0.2mM PA for 12 or 24 hrs and transferred the conditioned media to treat the cortical neurons for 24 hours. The conditioned media from PA-treated astroglia induced BACEl upregulation in cortical neurons, as observed by immunofluorescence imaging (Figure 3.3) and immunoblotting (Figure 3.4). Control PA Figure 3.3. BACEl immunostaining. lmmunofluorescence labelling of BACEI in neurons treated for 24 hours with conditioned media from PA-treated (for 24 hours) or untreated astrocytes (control). Images were obtained with confocal fluorescence microscopy (objective lens magnification- 40X). The PA-induced BACEl upregulation was observed to be dependent on the length of time that the astroglia were treated with PA, which might be attributed to the time- dependent increase in PA metabolism by the astroglia '82. BACEl cleaves APP at the major Asp+l site and minor Glu+l I site to generate C99 and C89 fragments, respectively 209. Accordingly, we found increased C99 levels in the PA-astroglia-treated cortical neurons as compared to controls (Figure 3.6). C-tenninal fragments of APP are found to 37 (A) BACEl a” “I“? w 1' Actin "1“" C PA C PA 12 hrs 24 hrs (B) l 80 l 60 140 BACEI l 20 (% Control) 1 00 80 60 40 20 C PA C PA 12 hrs 24 hrs Figure 3.4. Astroglia-mediated, PA-lnduced BACE] upregulation in neurons. Astrocytes were treated for 12 and 24 hours with 0.2mM of PA or 4% BSA (control), followed by transfer of the astrocytes- conditioned media to neurons (24 hours treatment). (A) Western blot analysis of BACE] protein levels in neurons. B-actin is shown as a marker for protein loading. (B) Histograms represent quantitative determinations of intensities of the relative bands. Data represent mean at SD. of three independent experiments. Student’s t-test was used for analyzing differences between different treatment groups. ‘, p<0.05 compared with respective control. be elevated in AD brain and are more toxic to neurons than AB, which is obtained by cleavage of C99 by y-secretase 242. The presenilin (PS) complex, including PS, nicastrin, APH-l and PEN-2, forms a central core of the y-secretase enzyme and the PSI C- 218 terminal tail (PSl-CTF) is critical for y-secretase activity . We found no change in the 38 levels of PSl—CTF suggesting the y-secretase activity is unchanged in the PA-astrocytes- treated neurons as compared to controls (Figure 3.5). It is noteworthy that a slight increase in BACEl levels, without any change in y-secretase activity, has been shown to increase AB production significantly 243. Thus, the increased BACE] levels in the PA- astrocyte-treated cortical neurons resulting in elevated C99 levels, may be followed by increased AB production, despite the lack of change in y-secretase activity. P51 ‘ “ ' --- —98 KDa PSl-CTF F—_‘ —21 KDa Control PA PA+DMU Figure 3.5. Immunoblot analysis of presenilin-l (PSI) levels in neurons treated with astrocytes- conditioned media. Astrocytes were treated for 24 hours with 0.2mM PA or 5% BSA (control), followed by transfer of the astrocytes-conditioned media to neurons (24 hours treatment), with or without 10 mM DMU. Detergent cell lysates from PA-treated and control cells were immunoblotted for PS] and PSl-CTF levels. The immunoblot is representative of 3 independent experiments. 3.3.3 Involvement of Oxidative Stress in FFA-Astroglia-Induced BACEl Up- regulation and Amyloidogenic processing of APP in Primary Neurons As shown in Chapter 2, intracellular levels of reactive oxygen species (ROS) were significantly elevated in the neurons cultured with the conditioned media from FFA- treated astroglia as compared to controls. To investigate the possible involvement of 39 oxidative stress in FFA-induced BACEl up-regulation and amyloidogenic processing of APP, we treated neurons with 1,3-dimethyl urea (DMU), an antioxidant. The co- treatment of neurons with lOmM DMU inhibited PA-astroglia-induced BACE] upregulation and increased C99 production in neurons (Figure 3.6). (A) (B) 203 183 183 ._ ._ ,..-. m BACE] 1: Actin - ~ ~ . 2 C PA PA+DMU 2: .. PA+DMU ppp _I--« b-‘N ‘33 3k 10 . *Ilt ' C99/ 1m APP an E) - 0 C99 ‘0‘” k ‘. 23 0 A ._ __7 V C PA C PA PA+DMU PA+DMU Figure 3.6. Oxidative stress Involved in astroglia-mediated, PA-induced elevations in BACE] and C99 levels in neurons. Astrocytes were treated for 24 hours with 0.2mM PA or 5% BSA (control), followed by transfer of the astrocyteSoconditioned media to neurons (24 hours treatment), with or without lOmM DMU. (A) Western blot analysis of BACE], APP and C99 protein levels in neurons. (B) Histograms represent quantitative determinations of intensities of the relative bands. Data represent mean :l: 8.5. of three independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. ‘, p<0.05 compared with control; '”' p<0.05 compared with PA treatment. This suggests a central role of astroglia-mediated oxidative stress in PA-induced upregulation of BACEl and increased production of C99 in neurons. The role of oxidative stress in causing increased amyloidogenic processing of APP is further supported by various in vitro and in vivo studies 244. Oxidative stress has been shown to 40 increase the expression and activity of BACEl in NT2 neurons, which was accompanied by a proportional elevation of the c-terminal fragments of APP 65’ 66. H202 and UV irradiation have been shown to increase production of AB peptides “'70 , and antioxidants Trolox and dirnethyl sulfoxide blocked stress-induced AB production 70. It is noteworthy that AB itself can cause oxidative stress and increase its own production 69, thus sustaining a vicious cycle 68. 3.4 CONCLUSIONS In conclusion, PA had no direct effect on BACEl levels and the amyloidogenic processing of APP in neurons. On the other hand, the conditioned media from PA-treated astroglia significantly increased BACEl levels in neurons, which was dependent on the length of time that the astroglia were treated with PA. This emphasizes a central role of PA metabolism by astroglia in the observed pathological effects in neurons. Furthermore, elevated BACEl increased amyloidogenic processing of APP, as evident by increased levels of C99 in PA-astroglia-treated neurons as compared to controls. Treatment of neurons with anti-oxidant blocked PA-induced abnormalities in neurons. Thus, the present study illustrates that elevated levels of saturated fatty acids play an important role in the up-regulation of BACE] and consequent amyloidogenic processing of APP through astroglia-mediated oxidative stress. 41 CHAPTER 4. SATURATED FATTY ACID-INDUCED ABNORMAL METABOLIC CHANGES ASSOCIATED WITH ALZHEIMER’S DISEASE 4.1 INTRODUCTION In addition to the two major pathophysiological lesions mentioned earlier (AB plaques and NFTS), AD pathology is also characterized by abnormal metabolic changes. Decreased cerebral glucose metabolism is a distinct characteristic of AD 4’ 5. In AD, brain glucose utilization and ATP formation are decreased significantly (approximately 46% and 19% respectively) as compared to healthy controls 245. In vivo imaging of AD brains using positron emission tomography (PET) with 2-[F-l 8]-fluoro-2-deoxy-D- glucose as a label shows progressive reduction in brain glucose metabolism, which is fiirther correlated with disease severity 246. In accordance with this, glucose hypometabolism has been suggested to be an important marker for early diagnosis of AD 4. The metabolic abnormalities observed in AD are widespread in that even the peripheral cells (fibroblasts) from AD patients show decreased metabolic activities 247’ 248. The activities of various metabolic enzymes, mainly pyruvate dehydrogenase, a-ketoglutarate dehydrogenase, glutamine synthetase, creatine kinase, aconitase and cytochrome oxidase have been shown to be decreased in AD 249'253. Interestingly, decreased cytochrome oxidase activity in post-mortem brain tissue from AD has been shown to be particularly located in NFT-bearing neurons 254’ 255. The decreased activities of these enzymes may be attributed to the increased oxidative stress in AD, as these enzymes are highly vulnerable to oxidative modification 256. Also, patients with mild cognitive impairment (MCI), which is characterized by reduced glucose metabolism, often develop AD 257. Finally, in 42 patients that are genetically predisposed to AD, cerebral metabolic changes occur well before any pathophysiological signs of the disease manifest 258. Glucose is a very important substrate in the efficient physiological functioning of the brain. Glucose, through cellular glycolysis, produces pyruvate that is further oxidized to acetyl-CoA. Acetyl-CoA is utilized by cells to produce cholesterol, acetylcholine and ATP 259. Thus, decreased glucose metabolism in AD may result in decreased production of these important cellular metabolites. Cholesterol is a primary sterol in cellular membranes and is important in the production of various neurosteroids 26°. Acetyl choline is a very important neurotransmitter involved in various cognitive functions, which have been shown to be decreased in AD 26'. In AD, the activity of choline cetyl transferase has been shown to be decreased in the presynaptic cholinergic neurons which might be attributed to decreased availability of acetylcholine 262. Muscarinic M1/M3 acetylcholine receptors have been shown to be involved in regulating APP processing and decreased acetylcholine levels may induce increased amyloidogenic processing of APP 263. In this context, degeneration of cholinergic system in AD has been correlated with disease severity 264. Finally, ATP is a cellular energy currency required for various cellular functions such as synthesis, folding, transport and degradation of proteins, maintenance of ion homeostasis and synaptic transmission among others 265. Experimental evidence suggests that decreased glucose metabolism and energy production may lead to increased amyloidogenesis and hyperphosphorylation of tau 266'270. Taken together, these data suggest that abnormal cerebral metabolism is central to AD pathology and may precede the neuropathological changes associated with the disease '0 . 43 Traditionally, it is considered that glucose in the brain is mainly metabolized by neurons and it is the substrate of choice for most activity-associated neuronal metabolism (Figure 4.1) 27‘. However, recent data suggest that astroglia take-up and metabolize glucose and produce lactate, the latter may be used by neurons as a fuel for metabolic activities and energy production (Figure 4.2) 27"273. In this context, the aim of the current study was to investigate the possible involvement of saturated fatty acids in causing abnormal glucose metabolism in both neurons and astroglia. Furthermore, we also focus on FFA-induced global metabolic changes in astroglia and their possible involvement in observed FFA- astroglia-induced ROS production in neurons, which in turn is involved in tau hyperphosphorylation and amyloidogenic processing of APP as discussed in chapters 2 and 3. NI! «rm- Juana-{n Gl..cn.~.r: ‘ :‘nmr-u- ‘ (L ..l 3 lil l.’ t ‘ Hut-nan a Cl .17 I (in A P 0' ‘ .— 7 “3p Gla ' 5 CI; .1 A170 0 . z) p ' p p In t Eh I U) " 0 > I ‘h' ‘ 9' * ' ” :- A’p ‘ > ' ... N'I .6 A. 'l .( , 8 ~_ . .— pfilnr'nl ‘ _ 3 9‘" —- K‘ n» m Pyramtn Pin-.116 ‘0 1 l mu - 1 12H 1 ru l 3H 5: - M'nln . l RCIR‘J . I M’Jn‘t‘ ‘ , , 'CA J4 '18!”-' .1! "IRA'P TCI . l v v ' (yelp - Iv'fl . A 05: , 3 a my- GP . —— n K ADP - P Figure 4.1. Conventional view of cerebral glucose metabolism. 44 “”7" Av'nryin SL502 ‘ “Linen ‘ 61.)? .1 51 U" I ‘ :1“ch 4 G. UT ' K’- r I 0 K II ' _ G u - 0.. 9 mp Cl .. . .- , I Alv’ ‘ y _. U) X ‘ .. . (u. . m . . , _ »- > 3 my ‘ ‘1’ :I AT: > "0" "a O Achm 9 K’ "‘ ’ ‘ ' R'P o antm', :Ii _ . _ ' 1 .1 N3 m p (' O- p ymvan ‘ Na prawn,» 1:»- t r! L .14"an +— ar'arn ‘— 13731» I VCA \ “2“” H‘A Ev: n . cyrin 5 UP " N" " ’ 09 ' ' - — 9 x- ADP . D Figure 4.2. Cerebral glucose metabolism based on the novel astrocyte-neuron lactate shuttle hypothesis. 4.2 MATERIALS AND METHODS 4.2.1 Isolation and Culture of Primary Rat Cortical Neurons and Astroglia Primary neurons and astroglia were isolated from the cortex of one-day-old Sprague- Dawley rat pups and cultured according to the methods as described in chapter 2. The cells were plated on poly-L-Lysine coated, 6-well plates at the concentration of 2 x 106 cells per well in fresh cortical medium [Dulbecco's Modified Eagle's Medium (DMEM and all other media are from Invitrogen, CA) supplemented with 10% horse serum (Sigma, MO), 25 mM glucose, 10 mM HEPES (Sigma), 2 mM glutamine (BioSource lntemational, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycine] '76. To obtain 45 pure neuronal cell cultures, after 3 days of incubation (37°C, 5% C02) the medium was replaced with the cortical medium supplemented with 5 uM cytosine-B—arabinofiiranoside (Arac, from Calbiochem, CA). After 2 more days, the neuronal culture was switched back to cortical medium without Arac. The neuronal cell culture of more than 95% was obtained by this procedure. The experiments were performed on 6-7 day old neuronal culture. To obtain primary cultures of astroglial cells, the cortical cells from one-day-old Sprague-Dawley rat pups were cultured in DMEM/Ham’s F12 medium (1:1), 10% fetal bovine serum (Biomeda, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycine ‘77. Cells were grown for 8-10 days (37°C, 5% C02) and culture medium was changed every 2 days. The astroglial cell culture of more than 95% was obtained by this procedure. 24 hours prior to treatment with fatty acids, the medium was changed to neuronal cell culture medium. 4.2.2 Western Blot Analysis For western blot analysis, cells were washed three times with ice-cold TBS (25 mM Tris, pH 8.0, 140 mM NaCl, and 5 mM KCl) and lysed for 20 minutes by scraping into scraping into ice-cold radioimmunoprecipitation assay (RIPA) buffer [1% (v/v) Nonidet P40, 0.1% (w/v) SDS, 0.5% (w/v) deoxycholate, 50 mM Tris, pH 7.2, lSOmM NaCl, 1 mM Na3VO4 and lmM PMSF, all chemicals from Sigma] 238. The total cell lysate was obtained by centrifugation at 12,000 rpm for 15 minutes at 4 0C. The total protein concentration was measured by BCA protein assay kit from Pierce (Rockford, IL). Equal amounts of total protein from each condition were run at 200 V on 10% Tris-HCl gels for detection of GLUTl and actin. The separated proteins were transferred to nitrocellulose membranes for 1 hour at 100 V and incubated at 4 0C overnight with the appropriate 46 primary antibodies [1:500 GLUTl, 1:2000 actin]. Blots were washed three times in PBS- Tween (PBS-T) and incubated with appropriate HRP-linked secondary antibodies (Pierce Biotechnology, IL) diluted in PBS-T for 1 hr. After an additional three washes in PBS-T, blots were developed with the Pierce SuperSignal West Femto Maximum Sensitivity Substrate (Pierce Biotechnology) and imaged with the BioRad ChemiDoc. Quantity One software from Bio-Rad was used to quantify the signal intensity of the protein bands. 4.2.3 Biochemical Measurements of Cellular Metabolites For measurement of various cellular metabolites the conditioned media were collected immediately after experimental treatment and centrifuged for 10 minutes at 3000 rpm to remove any cell debris. To calculate the glucose uptake and lactate production in particlular, their concentrations in the media were measured by using enzymatic glucose (Stanbio Laboratories, TX, USA) and lactate (Trinity Biotech, MO, USA) assays. The glucose uptake and lactate production were calculated by using the differences between the metabolite concentrations in the media before and after the treatment. The data were normalized by using total intracellular protein levels. Similarly, the concentrations of FFA (Assay kit from Roche Biochemicals), beta-hydroxybutyrate and acetoacetate (both assays from Stanbio Laboratories) in the extracellular media were measured according to manufacturer’s instructions. High-performance liquid chromatography (HPLC) method (Waters AccQTag amino acid analysis with fluorescence detector) was used to measure concentrations of various amino acids such as Asp, Glu, Gly, Arg, Thr, Ala, Pro, Tyr, Val, Met, Om, Lys, Ile, Leu and Phe. The concentrations of Ser, Asn, Gin and His were measured by a slight modification of the AccQTag method. The extracellular fluxes of these metabolites were measured for metabolic flux analysis by calculating the changes in 47 the levels of the metabolites in the cell culture media after 24 hours of treatment. The linearity of some of these fluxes over this interval was verified. 4.2.4 Metabolic Flux Analysis (MFA) MFA is a powerful mathematical technique that can provide a comprehensive snapshot of the metabolic profile of cells as a function of their environment. The basis of this method is that metabolic pathways have a well-defined stoichiometry relating reactants to products 274. In MFA, a mass-balance on various intracellular metabolites is performed and under the assumption of pseudo-steady state the differential equations representing the changes in the concentrations of the individual metabolites may be written as algebraic summation of the fluxes associated with that metabolite 275. These pseudo- steady state balances for all the intracellular metabolites under consideration can be written in matrix form as: S *v = 0 where, S is the stoichiometric matrix and v is the vector of metabolic fluxes. If m numbers of fluxes are measured, then it is possible to divide the matrix S into two submatn'ces, S,,, and S“, corresponding to the measured and unknown fluxes, respectively. This leads to- S..."‘vm + S..*v.. = 0 v. = (S.)"* (-s..*v..) Thus, with the knowledge of the stoichiometry and measured fluxes v,,,, the vector of the unknown fluxes vu can be calculated. Assumptions The assumptions pertaining to the MFA model employed in the current study are as follows: 48 ii. iii. iv. Pseudo-steady state assumption. The intracellular metabolites are assumed to be under the condition of pseudo-steady state in that there is no significant intracellular accumulation of any metabolite. Experiments were conducted to verify the pseudo- steady state assumption. Linearity of metabolic fluxes over 24 hour period Fluxes of the uptake or release of metabolites were assumed to be linear over the 24 hour period, that is the net change in the extracellular concentration of a given metabolite after 24 hours duration represents the flux of that metabolite. The metabolites are distributed uniformly inside the cell. Based on this assumption, a single MFA model could be applied to perform metabolite balances for the entire cell and separate models were not needed for the individual cellular compartments or organelles. This assumption has been successfully employed previously in various MFA studies 276 275. Urea cycle is not active in brain tissue. Urea cycle, present in liver, is critical in efficiently clearing up ammonia. However, the urea cycle is not present in the brain. The brain depends on the amidation reaction (glutamate to glutamine) catalyzed by glutamine synthetase, to clear ammonia 277. Therefore, the present model consisted of the glutamine synthetase reaction and the urea cycle reactions were not included. Glycerol is converted by the cells to glycerol-3-phosphate, which leads to dicylglycerol (DAG) formation or to glyceraldehyde-3-phosphate, the latter enters glycolysis leading to the production of pyruvate. However, the enzyme involved in the conversion of glycerol-3-phosphate to glyceraldehyde-3-phosphate, glycerol-3- 49 phosphate dehydrogenase, is not expressed in astroglia 278. Therefore, only the DAG formation reaction is included in the present model. Based on these assumptions, a MFA model consisting of 71 cellular metabolic reactions involved in the metabolism of glucose, amino acids and lipids was constructed (Table 4.1 and Figure 4.3). The model consisted of 54 intracellular metabolites as shown in Table 4.2. A total of 23 metabolic fluxes were measured (Table 4.3), yielding an over- determined system of equations, which was solved by least-squares fit using the Moore- Penrose pseudo-inverse calculation. This method has been successfully used in our 275 laboratory to study F F A-induced abnormalities in HepGZ cells Table 4.1. List of the cellular metabolic reactions Flux # Equation Glycolysis 1 G+ATP -> G-6-P 2 G-6-P -) F-6-P 3 F-G-P +ATP -) Glyceraldehyde-B-P + DHAP 4 DHAP -> Glyceraldehyde-B-P 5 Glyceraldehyde-B-P -) 3-PGA 6 3PGA -) PEP + NADH (+ ZATP) 7 PEP -) Pyr 8 Pyruvate + NADH -) (Lactate) 9 Pyruvate -) Acetyl-CoA + NADH +C02 TCA cycle 10 Acetyl-CoA + 0AA -) Citrate 1 1 Citrate -> a-KetoGlutarate + NADPH +C02 12 a-Ketoglutarate -> Succinyi-CoA + NADH + C02 13 Succinyl-CoA -) Fumavate + FADHZ + ATP 14 Fumarate -) 0AA + NADH Pentose-phosphate pathway 15 G-6-P -) 12 NADPH + 6 C02 16 C02 out Ketone Body production 17 2 Acetyl-COA -) Acetoacetyl-CoA 18 Acetoacetyl-CoA —) Acetoacetate 50 19 20 Table 4.1 Continued Acac out Acetoacetate + NADH -) (B-OH butyrate) Oxygen uptake and Oxidative Phosphorylation 21 22 23 02 (In) NADH + 0.5 02 -> 2.5 ATP FADHZ + 0.5 02 -) 2 ATP FFA synthesis and oxidation 25 69 68 26 24 Amino 27 28 29 30 31 32 33 34 35 36 37 38 49 4O 41 42 43 44 45 46 47 48 59 SO 51 52 53 S4 54 8 Acetyl-CoA + 14 NADH -> FA-CoA Glycerol + ATP -) Glycerol-3-P ZFA-CoA + Gcherol-3-P -) DAG FA-CoA + DAG -) TG FA-CoA (In) acid metabolism Ser (In) 3-PGA + Glu --> Ser + a-KG + NADH Gln In Asp (In) Glu (In) Gly (In) Gly 9 2 C02 + NH3 + NADH + THF + ATP NH4 (In) Arg (In) Thr In Ala (In) Glu + Pyr -) Ala + aKG Pro In Tyr (In) Tyr + aKG + 2 02 -) Glu + C02 + Acetoacetate + Fumarate Val (In) Orn IN Lys IN Ile IN Lue In Phe In Gln -) Glu + NH4 0m + a-KG + 0.5 NADPH + 0.5 NADH --> Pro Asp + NH4 -) Asn Thr -) Pyr + C02 + NH4 + 2 NADH + FADHZ Val + aKG -) Glu + C02 + 2NADH + FADHZ + Succ-CoA Lys + 2 aKG + NADPH -) ZGIu + Acetoacetyl-CoA + 2C02 + 4 NADH + FADHZ Ile + aKG -) Glu + Succ-CoA + Acetyl-CoA + NADH + FADHZ Leu + aKG -) Glu + HMG-CoA + NADH + FADHZ 51 Table 4.1 Continued 56 Phe + 02 9 Tyr 59 a-KG + NH4 + NADPH --> Glu 70 Cys + 02 + a-KG --> Glu + Pyr + $04 71 Cystine --> 2 Cys Synthesis of Cholesterol and Cholesteryl ester 60 AcetoacetyI-CoA + Acetyl-CoA 9 HMG-CoA 61 HMG-CoA + 2 NADPH (+ 3ATP) 9 IPP 62 2 IPP 9 GeranyI-PP 63 Geranyl-PP + IPP 9 Farnesyl-PP 64 2 FarnesyI-PP + 0.5 NADPH + 0.5 NADH 9 Squalene 65 Squalene + 02 + NADPH --> Lanosterol 66 Lanosterol + 10.5 NADPH + 4.5 NADH + 10 02 9 Chol + 3 C02 67 Chol + FA-CoA --> Cholesterol Ester Sphingolipid Metabolism 57 Ser + I Palm-CoA + 1 FA-CoA + NADPH 9 Ceramide + C02 + FADHZ 58 Ceramide + Phosphatidleholine 9 Sphingomyelin illumn I I Quanta 15’16 p< PPP) 69 I glymrol I /E|0WM°> 26 2,3,4 Shawls?- phosvhab 68 Diacayl <1praldehydo glprol SphingomyelD -3-ph/oaphab Palmihb 3U 58 / Palm CoA 57 [Puruwb56 /\-CAC c A) - 17 “Comm“; CPU/v 51 70 W” C“ 18.19.20 ’ \ 22:22: I 0AA)“ K 56 henylalanin) C Val > 14 Co"- I] (”X41 5, 55/...) (min!) (”'3‘ 53 / c223 G“'°°'")+\/_/-@m.>.4/ .448 6......) (when!) /12 38 w vmb (I. 54 CM) 13 Figure 4.3. Astroglial metabolic network. Boxes represent extracellular metabolites. while ovals represent intracellular metabolites. The direction of reaction assumed in the model is indicated by arrows. 52 Table 4.2. List of intracellular metabolites Metabolite _a ommeU'l-th—i DhAb-hwwwwwwwwwwNNNNNNNNNN-'-|-'-'did—Dd AwN-‘O‘Dmflmm-bdeOtOmeUl-bwml—Iommem-th-i glucose-6-P fructose—6-P glyceraldehyde-3—P 3-PGA DHAP Phosphoenolpyruvate Pyruvate C02 acetyl-CoA Oxaloacetate Citrate a-ketoglutarate succinyI-CoA Fumarte Acetoacetate acetoacetyl-CoA B-OH 02 GcheroI-3-P Palm-CoA NADH NADPH FADHZ Ser NH4 Glu Gln Val Ile Leu Arg Orn Ala Gly Tyr Thr Lys Phe Pro Asp Asn Cys Ceramide Sphingomyelin 53 Table 4.2 Continued 45 HMG-CoA 46 lsopentenyI-PP 47 GeranyI-PP 48 FarnesyI-PP 49 Squalene 50 Lanosterol 51 Cholesterol 52 Cholesterol Ester 53 DAG 54 Sulfate Table 4.3. List of measured fluxes Flux # Metabolite 1 Glucose 8 Lactate 19 Acetoacetate 20 Beta-hydroxybugrate 21 02 In 24 Palm 27 Ser 29 Gln 30 Asp 31 Glu 32 Gly 34 NH4 35 Arg 36 Thr 37 Ala 39 Pro 40 Tyr 42 Val 43 Orn 44 Lys 45 lie 46 Leu 47 Phe 4.2.5 Measurement of Intracellular ATP in Astroglia To measure the intracellular ATP levels, astroglia were washed with ice-cold PBS and then lysed with 0.7% perchloric acid (PCA). The PCA was neutralized by using 0.7N 54 NaOH. The neutralized samples were used to measure intracellular ATP levels with a luciferase-based chemiluminescent assay (Molecular Probes, CA). 4.2.5 Measurement of Intracellular Ceramide in Astroglia Astroglia were washed with ice-cold PBS and then lipids were extracted by using a chloroform/methanol method as described by Bligh and Dyer 279. The organic phase was dried under N2 and the ceramide was measured after its deacylation to sphingolipid base and derivitization with o-phthaldehyde (OPA) as described earlier 28°. Briefly, the dried lipids from the organic phase were re-suspended in 500 pl of 1N KOH in methanol and incubated at 100°C for 1 hour to deacylate ceramide to free sphingolipid bases. The lipids were then dissolved in 50 ul of methanol and 50 pl of OPA reagent is added to this. The CPA reagent was prepared by mixing 99 ml of boric acid (3% w/v in water, pH 10.5), lml of ethanol containing 50 mg OPA (Sigma) and 50 ul of B-mercaptoethanol (Sigma). The derivatized sample aliquots (20 ul) were quantified by high performance liquid chromatography (HPLC) using Nova Pak C18 column (60 A0, 4 pm, 3.9 mm x 150 mm; from Waters, MA, USA). Fluorescent-labeled lipids were eluted isocratically by using methanol:5mM potassium phosphate (pH 7.0) (90:10, v/v) at a flow rate of 0.6 ml/min and detected with a fluorescence detector (excitation wavelength 340 nm, emission wavelength 455 nm). A standard curve obtained by running known amounts of ceramide (type 111, from bovine brain Sphingomyelin; from Sigma) was used as a comparison to determine the ceramide levels in the experimental samples. 55 4.2.6 Data Analyses Data are shown as means : SD. for indicated number of experiments. Student’s t-test and one-way ANOVA with Tukey’s post hoc method were used to evaluate statistical significances between different treatment groups. Statistical significance was set at p<0.05. 4.3 RESULTS AND DISCUSSION 4.3.1 FFA-Induced Abnormal Glucose Metabolism To study the effects of PA on cellular glucose metabolism, neurons and astroglia were treated with 0.2mM of PA for 24 hours. As shown in Figure 4.4A, there was no change in the glucose uptake and lactate production in neurons treated with PA as compared to the untreated ones. This is in line with the observed, lack of direct effect of PA on neurons in terms of AD-associated pathophysiological changes as discussed earlier (Chapters 2 and 3) and may similarly be attributed to the low affinity of primary neurons towards PA '82. However, as mentioned earlier, astroglia have a higher capacity to take up and metabolize PA '82. Thus, in the case of astroglia, PA may compete with glucose for cellular uptake. Previously, PA has been shown to inhibit glycolysis in primary hepatocytes 28" 282. Furthermore, high fat diet has been shown to increase palrnitate oxidation and decrease fructose oxidation in isolated primary hepatocytes 283. We found that PA treatment significantly decreased basal glucose uptake and lactate release from astroglia (Figure 4.4B). 56 Glucose 35 Lactate Jumoleslmg protein 60 . Glucose umoleslmg protein to h in O O O N O 1 FL“ ,: o . flu. <- —4r_-;E‘ 0;: PA 80 Lactate umoleslmg protein .h 0 C PA PA+AICAR Figure 4.4. PA downregulates glucose uptake and lactate release by astroglia. The cortical neurons and astroglia were treated for 24 hours with 0.2mM of PA or 4% BSA. (A) In neurons, PA treatment did not change glucose uptake and lactate production. (8) PA treatment significantly decreased glucose uptake and lactate production by astroglia. AMPK-activator AICAR did not inhibit PA-induced downregulation in glucose metabolism. Data represent mean 1- SD. of six experiments. Student’s t-test was used for analyzing the differences between the two treatment groups. *, p<0.05 compared with respective control. 57 Here it is imperative to note that although PA did not directly affect glucose metabolism in neurons, PA-astroglia-induced ROS production in neurons (as discussed in chapter 2) may affect glucose uptake and metabolism in neurons; oxidative stress has been shown to affect the activities of glucose transporter and glycolytic enzymes and in turn affect glucose uptake and metabolism in neurons 28”“. To investigate perturbed astroglial metabolism due to PA treatment, we measured cellular ATP production in astroglia. We expected PA-treatment to decrease astroglial ATP production in light of our observation that PA decreases glucose uptake. We, however, found that there was an increase in ATP production in the PA-treated astroglia as compared to the untreated ones (Figure 4.5). 140 it 120 100 ' 80 60 40 20 0 V . C PA Figure 4.5. Measurement of intracellular ATP in astroglia. The cortical astroglia were treated for 24 hours with 0.2mM of PA or 4% BSA. PA treatment increased cellular ATP production in astroglia. Data represent mean i SD. of 3 experiments. Student's t-test was used for analyzing the differences between the two treatment groups. ', p<0.05 compared with respective control. ATP (°/o of control) increased ATP production in PA-treated astroglia may be due to the uptake and increased oxidization of PA by astroglia. In support of this, previously it has been shown that although PA inhibits glucose oxidation, PA-oxidation itself makes up for this and thereby prevents ATP loss, which otherwise would be expected due to the reduced glucose 58 oxidation 287’ 288. Furthermore, it has been shown that although glucose utilization is reduced in AD brain, oxygen consumption and C02 production are unchanged or even increased as compared to healthy controls 289' 29° . Thus, it has been hypothesized that substrates other than glucose (e.g. FF As and endogenous amino acids) may be oxidized in AD brain 245, which may partially compensate for the energy deficit observed in AD brain due to decreased glucose metabolism 29'. 4.3.2 Cellular Mechanism of FFA-Induced Abnormal Glucose Metabolism in Astroglia We hypothesized that the observed, PA-induced abnormal glucose metabolism may be due to the potential effect of PA on the level of astroglial glucose transporter (GLUTl) or due to possible involvement of PA in perturbing the signaling mechanism involved in the cellular glucose metabolism, e.g. AMP-activated protein kinase (AMPK). Along those lines, it has been previously shown that polyunsaturated fatty acids (arachidonic acid) deficiency results in down-regulation of the glucose transporter in astroglia 292. However, the effects of elevated levels of saturated fatty acids on astroglial glucose transporters have not been studied. AMPK is a serine-threonine kinase, which is involved in regulating cellular metabolism 293. It was first isolated from liver and is also expressed in many other tissues including lung, kidney, heart, skeletal muscle, and brain 294. At the sub-cellular level in the brain, AMPK is expressed in both neurons and astroglia; however, its activity is 3X higher in astroglia as compared to neurons 295. The importance of AMPK in AD research is emphasized by the fact that inflammation is central to the AD pathology and AMPK activation has been shown to inhibit the production of 59 296 inflammatory cytokines in astrocytes AMPK activation has also been shown to protect cortical and hippocampal neurons from oxygen-glucose deprivation 297. Therefore, we treated astroglia for 24 hours with a cell-permeable pharmacological activator of All/IPK, 5 ' ' 'J ‘ 4 L " ribonucleoside (AICAR). Co- treatment of PA-treated astroglia with 0.25mM AICAR did not increase glucose uptake and lactate production as compared to astroglia treated with PA (Figure 4.4B). This suggests that AMPK may not be involved in the observed PA-induced abnormal glucose metabolism in astroglia. On the other hand, we found that the level of astroglial glucose transporter (GLUTl) was significantly downregulated in PA-treated astroglia as compared to untreated cells (Figure 4.6). Thus, the observed downregulation in glucose uptake by astroglia in the presence of PA may be attributed to the PA-induced downregulation of GLUT] levels in astroglia. Interestingly, AD brain is characterized by 298 significant reductions in GLUTl levels , and disease severity is associated with progressive decline in GLUTl gene expression 299. 120 l 100 - 80 - GLUTl «I. -—.. 60- Actin w- ‘0 ‘ C PA 20' C PA Figure 4.6. PA downregulates GLUTI level in astroglia. Astroglia were treated for 24 hours with 0.2mM of PA or 4% BSA. The immunoblot analysis shows that PA-treatment significantly decreased the levels of GLUTl as compared to the untreated ones. B-actin is shown as a marker for protein loading. Histogram represents quantitative determinations of intensities of the relative bands normalized with actin. Data represent mean at SD. of three independent experiments. Student’s t-test was used for analyzing the differences between the two treatment groups. ‘, p<0.05 compared wim respective control. 60 4.3.3 FFA-Induced Global Metabolic Changes in Astroglia Abnormal metabolism precedes the cascade of neuropathological changes in AD pathology 1°. Decreased glucose metabolism and energy production have been shown to be involved in increased amyloidogenesis and hyperphosphorylation of tau in neurons 2“ 27°. However, as we showed here, PA treatment did not affect the glucose metabolism in neurons but only in astroglia. Therefore, we hypothesized that metabolic changes, other than the abnormal glucose metabolism induced by PA in astroglia may be involved in causing the observed, PA-astroglia-induced pathophyiological changes in neurons (chapters 2 and 3). The main focus was to investigate various FFA-metabolizing pathways in astroglia that may potentially be involved in causing cellular ROS production, specifically in neurons, as is observed in our studies. Based on extensive literature review, we propose 3 major pathways by which PA may induce cellular ROS production (Figure 4.7). FFA metabolism fi-oxidation Diacylglycerol Sphingolipid (DAlG) metabolism ® Ha/(c ramifie)——~—’/vh’* Ros NAD(P) /7/' oxidase iNOS (NO) ROS Cytokines Astrocyte Neuron Figure 4.7. FFA-metabolizing pathways involved in cellular ROS production. 61 Elevated levels of PA may lead to its increased cycling through B-oxidation pathways in mitochondria and other organelles such as peroxisomes and glyoxysomes. The increased oxidation of PA may lead to enhanced cellular ROS production 300. PA may also increase diacylglyecrol (DAG) production, which in turn may activate NAD(P)H oxidase and increase cellular ROS 300’ 30'. However, as shown earlier in Chapter 2, PA did not induce ROS production in astroglia (Figure 2.6), which may suggest the lack of or minimal activation of these two pathways (ti-oxidation and DAG production) in PA-treated astroglia. Finally, PA may also be metabolized by the cells to synthesize ceramide 302. Ceramides are also potent intracellular activators of NAD(P)H oxidase 303 and thus may increase cellular ROS production. More importantly, ceramide secreted from astrocytes may directly act on neurons and mediate oxidative stress-induced effects in neurons 304. Furthermore, PA-induced increase in ceramide levels may induce secretion of cytokines 305 or other signaling molecules, e.g., NO 306 by astrocytes, which in turn may elevate production of ROS in the neurons 307’ 308. Thus, taken together, these data may suggest the sphingolipid pathway (ceramide) to be predominantly activated in PA-treated astroglia and ceramide to be a possible mediator of the FFA-induced pathological damage in neurons as shown in our present studies. We sought to support this hypothesis by using mathematical modeling (MFA) and additional experiments. 4.3.3.1 Verification of the pseudo-steady state assumption and linearity of fluxes over 24 hours To verify whether the pseudo-steady state assumption for MFA is valid, intracellular and extracellular lactate concentrations were measured after 6, 12 and 24 hour treatments. 62 Lactate was the metabolite of choice due to its highest flux, which offered two advantages 275-(1) due to its highest molar synthesis, it is likely the most concentrated metabolite, simplifying accurate detection, and (2) its highest molar change provides the most stringent test of the pseudo-steady state assumption. As shown in Figures 4.8A and B, both the intracellular and extracellular concentrations of lactate increased with time in response to PA treatment. The extracellular lactate release was approximately linear over the 24 hour period for both the control and PA treatments. Furthermore, as shown in Table 4.4, the changes in the intracellular lactate levels were about a thousand-fold smaller as compared to changes in the extracellular lactate levels. This indicates that the intracellular accumulation of lactate is negligible and the pseudo-steady state hypothesis is valid. Lactate (Intracellular) 0.09 +C 0.08 H + pA 0.07 tein 0.06 E 0.05 \ 3 0.04 0.03 mlcromol 0.02 0.01 - 6 h 12 h 24 h Time Figure 4.8A. Time-dependent measurements of intracellular lactate levels. Astroglia were treated for 6, 12 and 24 hours with 0.2mM of PA or 4% BSA. The cells were trypsinized, washed with TBS and lyzed by using 0.7% perchloric acid. The lactate levels in cell lysate were measured by enzymatic assay. Data represent mean :1: SD. of three independent experiments. 63 Lactate (Extracellular) IO 0 +C +PA m 0 l micromoles/ mg protein H N U A 01 Ch \I O O o O o o o . i . i 1 Hi. 0 6 n 12 h 24 h Time Figure 4.88. Time-dependent measurements of extracellular lactate levels. Astroglia were treated for 6, 12 and 24 hours with 0.2mM of PA or 4% BSA. The conditioned media were collected and the lactate levels in the media were measured by enzymatic assay. Data represent mean :1: SD. of three independent experiments | I24h lntra I24h Extra IRatio (lntralExtra) | IControl | 0.07303I 73.35371l 9.32E-04I IPA | 0.0777eel 61.93194] 1.26503 Table 4.4. Ratio of intracellular to extracellular lactate levels. 4.3.3.2 MFA analysis Table 4.5 shows a complete list of fluxes calculated from MFA analysis for both the control and PA-treated astroglia. Firstly, in both the control and PA-treated cells fatty acid synthesis but not the fatty acid oxidation, was more prominent (flux 25, Table 4.5). In PA-treated astroglia fatty acid synthesis was lower as compared to controls, which 64 might be attributed to the exogenously added PA. In this context, there is evidence that astrocytes do synthesize fatty acids 309 and exogenous addition of fatty acids decrease the endogenous fatty acid synthesis 3'0. Secondly, in PA-treated cells, glycerol uptake was significantly decreased (flux 69, Table 4.5). Previously, PA treatment has been shown to decrease glycerol uptake inHepGZ cells 275. As glycerol is involved in DAG production, PA-induced decrease in glycerol uptake may further contribute to decreased synthesis of DAG in the PA—treated astroglia (flux 68, Table 4.5). Finally, flux through de novo synthesis of ceramide was significantly elevated in PA-treated astroglia as compared to controls (flux 57, Table 4.5). Experimental measurements also showed higher intracellular ceramide levels in PA-treated astroglia as compared to controls and co- treatment of astroglia with 2mM L-cycloserine (L-CS), an inhibitor of de novo synthesis of ceramide, inhibited PA-induced ceramide increase (Figure 4.9), thus further validating the MFA findings. Table 4.5. Metabolic flux values calculated by MFA Flux Control Palmitate # Equation Average Error Average Error 52.8850 5.8490 37.4540 3.8160 1 G+ATP -> G-6-P 52.7431 5.8491 37.2262 3.8160 2 G-G-P -) F-6-P F-G-P +ATP —) Gcheraldehyde-3-P + 52.7431 5.8491 37.2262 3.8160 3 DHAP 52.7431 5.8491 37.2262 3.8160 4 DHAP —) Glyceraldehyde-3-P 105.4862 11.6982 74.4523 7.6320 5 Glyceraldehyde-3-P -) 3-PGA 104.1620 11.7016 72.4859 7.6465 6 3PGA -> PEP + NADH (+ ZATP) 104.1620 11.7016 72.4859 7.6465 7 PEP -) Pyr 78.3500 1.2500 61.9300 3.1000 8 Pyruvate + NADH -) (Lactate) 9 Pyruvate 9 Acetyl-CoA + NADH +C02 24-4819 “-7797 94574 82970 65 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 Table 4.5 Continued. Acetyl-CoA + 0AA 9 Citrate Citrate 9 a-KetoGIutarate + NADPH +C02 a-Ketoglutarate 9 SuccinyI-CoA + NADH + C02 Succinyl-CoA 9 Fumarate + FADHZ + ATP Fumarate 9 0AA + NADH G-6-P 9 12 NADPH + 6 C02 C02 out 2 Acetyl-COA -) AcetoacetyI-CoA AcetoacetyI-CoA 9 Acetoacetate Acac out Acetoacetate + NADH 9 (B-OH butyrate) 02 (In) NADH + 0.5 02 9 2.5 ATP FADHZ + 0.5 02 9 2 ATP FA-CoA (In) 8 Acetyl-CoA + 14 NADH -) FA-CoA FA-CoA + DAG 9 TG Ser (In) 3-PGA + Glu --> Ser + a-KG + NADH Gln In Asp (In) Glu (In) Gly (In) Gly 9 2 C02 + NH3 + NADH + THF + ATP NH4 (In) 66 5.8041 6.1589 0.7998 4.7440 5.4492 0.1419 39.0347 1.2702 1.9907 2.3390 0.0020 24.5230 33.2840 13.5899 0.1510 3.3083 0.8802 0.7100 1.3241 4.7620 0.2140 -0.9460 1.7060 1.5013 3.5020 1.0401 1.035 1.1461 1.0824 1.0467 0.0359 10.9023 0.9366 0.8355 0.8040 0 2.1000 3.7417 1.1080 0.0210 1.5521 0.5339 0.0380 0.2431 0.7520 0.0030 0.1830 0.1180 0.1373 0.1760 5.0068 5.4583 0.1808 3.8455 4.5552 0.2278 25.7209 0.9077 2.1769 2.4320 0.0030 23.2030 32.5936 12.5787 1 .0600 1 .2357 0.2139 0.7260 1 .9665 4.8990 0.2050 -0.5090 2.9720 2.5585 4.0760 0.9997 1 .0044 1 .5399 1.1778 1.0048 0.0047 7.8214 1.2612 0.9631 0.8280 0.0010 2.1000 4.4499 1.1972 0.1450 1.1552 0.4917 0.0790 0.4702 1.8360 0.0010 0.4850 0.9830 0.9404 0.0220 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 S3 54 55 56 57 58 59 Table 4.5 Continued. Arg (In) Thr In Ala (In) Glu + Pyr 9 Ala + aKG Pro In Tyr (In) Tyr+aKG+2029Glu+C02+ Acetoacetate + Fumarate Val (In) Orn IN Lys IN Ile IN Lue In Phe In Gln 9 Glu + NH4 0m + a-KG + 0.5 NADPH + 0.5 NADH --> Pro Asp + NH4 9 Asn Thr 9 Pyr + C02 + NH4 + 2 NADH + FADHZ Val + aKG 9 Glu + C02 + 2NADH + FADHZ + Succ-CoA Lys + 2 aKG + NADPH 9 ZGIu + Acetoacetyl-CoA + ZCOZ + 4 NADH + FADHZ Ile + aKG 9 Glu + Succ-CoA + Acetyl- CoA + NADH + FADHZ Leu + aKG 9 Glu + HMG-CoA + NADH + FADHZ Phe + 02 9 Tyr Ser 4» 1 Palm-CoA + 1 FA-CoA + NADPH 9 Ceramide + C02 + FADl-IZ Ceramide + PhosphatidyIChoIine 9 Sphingomyelin a-KG + NH4 + NADPH --> Glu 67 1.5340 1.7910 -2.5070 2.7117 -0.6020 -0.3900 0.3503 0.1580 -0.0440 1.7030 3.1310 2.1720 0.4400 -4.8166 0.1016 0.2094 1.5863 0.3082 1.2936 3.2812 1.9673 0.5902 0.4094 0.2047 1.3589 0.1240 0.4230 0.2100 0.2138 0.0360 0.0980 0.2274 0.0350 0.0060 0.4310 0.2010 0.1600 0.1230 0.6129 0.0309 0.0402 0.4071 0.1005 0.3607 0.2044 0.1710 0.1461 0.1607 0.0804 0.5890 1 .4980 2.0220 -2.1700 2.5835 -0.4570 -0.2940 0.2581 0.4430 -0.0380 2.7340 2.6940 1.5910 0.4760 -5.2744 -0.0163 0.3092 1 .6085 0.481 1 1.9070 2.7321 1.1775 0.5141 0.8270 0.4135 2.2459 0.2810 0.5570 0.1290 0.1973 0.0530 0.2420 0.4920 0.3480 0.01 1 0 0.0560 0.2630 0.3230 0.0500 1 .4865 0.0557 0.0776 0.5480 0.3861 0.3133 0.3272 0.3420 0.2304 0.31 05 0.1552 1.3018 Table 4.5 Continued. Acetoacetyl-CoA + Acetyl-CoA 9 4.9673 0.1710 -1-1775 0.3420 60 HMG-CoA e1 HMG-c°A + 2 NADPH (+ 3ATP) .9 |PP -o.oooo 0.0000 -0.0000 0.0000 -0.0000 0.0000 ~0.0000 0.0000 62 2 lPP 9 Geranyl-PP -0.0000 0.0000 -0.0000 0.0000 63 Geranyl-PP + IPP 9 FarnesyI-PP 2 Farnesyl-PP + 0.5 NADPH + 0.5 00000 (1000‘) 0-0000 00000 64 NADH 9 Squalene Squalene + 02 + NADPH --> -0.0000 0.0000 -0.0000 0.0000 65 Lanosterol Lanosterol + 10.5 NADPH + 4.5 NADH 00000 00000 00000 00000 66 +10 02 9 Chol+ 3 C02 -0.0000 0.0000 -0.0000 0.0000 67 Chol + FA-CoA -—> Cholesterol Ester ZFA—CoA + GcheroI-3-P -) 0.8802 0.5339 0.2139 0.4917 68 DAG 0.8802 0.5339 0.2139 0.4917 69 Glycerol + ATP 9 Glycerol-3-P -0.2047 0.0804 -O.4135 0.1552 70 Cys + 02 + a-KG --> Glu + Pyr + 504 -0.1024 0.0402 -0.2067 0.0776 71 Cystine --> 2 Cys 200 ~ * 180 1130 - 140 4 # Ceramide 12° ‘ (% Control) 100 . C PA PA+L-CS Figure 4.9. PA-lnduced, de novo syntnests or ceramtne in astroglia. Astroglia were treated tor 24h with 0.2mM PA or 4% BSA (control), after which cellular lipids were extracted for ceramide determination by HPLC. PA significantly increased ceramide synthesis in astroglia, which was completely inhibited by treatment of astroglia with 2mM L-CS, an inhibitor of de novo synthesis of ceramide. Data are taken from 3 different experiments and are expressed as mean :1: SD. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. ‘, p<0.05 compared with control; #, p<0.05 compared with PA treatment. 68 4.4 CONCLUSIONS In conclusion, PA had no direct effect on either glucose uptake and lactate production in neurons. On the other hand, PA significantly decreased both glucose uptake and lactate release in astroglia. The observed downregulation in glucose uptake by astroglia in the presence of PA may be attributed to PA-induced downregulation of astroglial glucose transporter (GLUTI) levels. In addition to these PA-induced abnormalities in astroglial glucose metabolism, we also showed by using MFA that flux through de novo synthesis of ceramide is elevated in PA-treated astroglia as compared to control. This MFA finding was further validated experimentally; PA-treated astroglia had elevated intracellular levels of ceramide as compared to controls, which were decreased by the astroglial treatement with L-CS, which inhibits serine palmitoyltransferase-l (SPT-l), an enzyme that catalyzes the first committed step of de novo synthesis of ceramide. Thus, the present data establish a central role of saturated FFAS in causing abnormalities in astroglial glucose metabolism and further warrant investigating a possible causal role of increased astroglial ceramide levels in FFA-astroglia-induced pathological changes in neurons. 69 CHAPTER 5. INVOLVEMENT OF CERAMIDE IN FFA- ASTROGLIA-INDUCED PATHOPHYSIOLOGICAL ABNORMALITIES ASSOCIATED WITH AD 5.1 INTRODUCTION Ceramides are one of the most important sphingolipids and are composed of sphingosine and fatty acid, which are joined in an amide bond. Ceramides are involved in the synthesis of Sphingomyelin, one of the major components of the cellular lipid bilayer, thus playing a critical role in structural integrity of cell membranes. In addition, ceramides also act as signaling molecules and are involved in many important signaling 31 1. Ceramides are functions such as cell growth, differentiation, cell death etc. synthesized in cells through two pathways- (1) the sphingomyelinase (Smase) pathway and (2) the de novo synthesis pathway (Figure 5.1). Smase is regulated by the cellular redox state; increased oxidative stress activates Smase 3”. Currently, there are five different enzymes characterized as Smases based on their pH dependence, cation dependence and cellular localization 312. Smases break down membrane sphingomyelins into ceramide and fiee fatty acids. On the other hand, the de novo synthesis of ceramide uses serine and palmitoyl-CoA to synthesize ceramide and is initiated by serine palmitoyltransferase (SPT-l), which is the rate-limiting step of ceramide synthesis. Ketosphinganine formed in this first reaction is then converted to sphinganine by ketosphinganine reductase. A double bond is introduced to sphinganine by dihydroceramide synthase leading to the production of dihydroceramide, which is converted to ceramide by dihydroceramide desaturase. 70 serine 8pm , , ketosphinganine . ' ——* ketosphmganlne ; sphinganine PalmitoyI-COA reductase dihydroceramide synthase dihydroceramide dihydroceramide desaturase Smase Sphingomyelin .____' ceramide Figure 5.1. Cellular ceramide production. Ceramide is produced in cells by de novo synthesis from serine and palmitoyl-CoA or by breakdown of membrane sphingomyelins. Abnormal ceramide metabolism has been implicated in many diseases such as diabetes, AIDS and neurodegenerative disorders 3 11_ Ceramides may induce insulin resistance associated with diabetes and inhibition of ceramide synthesis has been shown to ameliorate glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance 313' Ceramide level has been also shown to be elevated in the cerebrospinal fluid of AIDS patients; elevated cerebral ceramides may be involved in neuronal cell death thus leading to the dementia often associated with AIDS 3 '4. In AD brain, ceramide levels were found elevated as compared to those of healthy controls, and the levels were higher in the affected regions (cortex and hippocampus) as compared to the unaffected regions (cerebellum) of the AD brain 3'5' 3'6, thus suggesting a central role of elevated ceramide levels in AD pathology. On the sub-cellular level, immunohistochemical analysis of AD brain showed abnormal expression of ceramides in the astroglia as compared to neurons 3 '7. In addition, ceramides have also been shown to be elevated in the white matter of AD 7l brain 318. The involvement of increased ceramide levels in AD pathogenesis is further emphasized by a recent study that showed that the gene expression of the enzymes involved in the de nova synthesis of ceramides is significantly upregulated in AD brain 3 '9. In addition, increased ceramide synthesis has been implicated in Aft-induced death of neurons 320' 32' and oligodendrocytes 322 in AD. In chapters 2 and 3, we showed that saturated fatty acids induced tau hyperphosphorylation and amyloidogenic processing of APP in neurons through astroglia-mediated oxidative stress. Furthermore, in chapter 4 we showed that astroglia treatment with PA significantly increased de nova synthesis of ceramide. Therefore, the aim of the current study was to investigate the possible involvement of PA-induced, increased astroglial ceramide levels in causing AD- associated, pathophysiological changes observed in neurons. 5.2 MATERIALS AND METHODS 5.2.1 Isolation and Culture of Primary Neurons and Astroglia from Rat Cortex and Cerebellum Primary cortical neurons and astroglia were isolated from the brains of one-day-old Sprague-Dawley rat pups and cultured according to the methods as described in chapter 2. The cells were plated on poly-L-Lysine coated, 6-well plates at the concentration of 2 x 106 cells per well in fresh cortical medium [Dulbecco's Modified Eagle's Medium (DMEM and all other media are from Invitrogen, CA) supplemented with 10% horse serum (Sigma, MO), 25 mM glucose, 10 mM HEPES (Sigma), 2 mM glutamine (BioSource lntemational, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycine] '76. 72 To obtain pure neuronal cell cultures, the medium was replaced with the cortical medium supplemented with 5 11M cytosine-B-arabinofuranoside (Arac, from Calbiochem, CA) after 3 days of incubation (37°C, 5% C02). After 2 more days, the neuronal culture was switched back to cortical medium without Arac. The neuronal cell culture of more than 95% was obtained by this procedure. The experiments were performed on 6-7 day old neuronal culture. Primary cerebellar neurons were isolated from 7-day-old rat pups, according to enzyme digestion and trituration techniques described previously 323. Briefly, dissected cerebellar tissue was placed in a cerebellar buffer solution containing 136.89mM NaCl, 5.36mM KCl, 0.34mM NazHPO4, 0.44mM KH2P04, 5.55mM dextrose, 20.02mM Hepes, and 4.17mM NaHCO3, PH 7.4. Cerebellar tissue was minced, transferred to 0.025% trypsin solution in cerebellar buffer, and incubated in water bath for 15 minutes at 37°C. 0.04% DNase I solution in cerebellar medium (DMEM supplemented with 10% horse serum, 25mM KC], 5 mg/ml insulin, 50 M GABA, lOOIU/ml penicillin, and 0.1 mg/ml streptomycine) was then added to inactivate trypsin. After the supernatant was collected from the trituration steps, cerebellar neurons were separated from the debris into 4% BSA solution in cerebellar buffer supplemented with 0.03% MgSO4. Finally, the purified neurons were plated onto poly-D-Lysine coated six- well culture dishes at a density of 2.0><105 cells/ml in 2ml of fresh cerebellar medium. One day after incubation (37°C, 10% C02/95% air), half the medium was subsequently replaced with fresh cerebellar medium supplemented with 20uM cytosine-[3- arabinofaranoside. Total medium was replaced with fresh cerebellar medium afier three days. Afterwards, half of the fresh cerebellar medium was replaced every third day. The experiments were performed on 10-12 day old neuronal culture. To obtain primary 73 cultures of astroglial cells, the cortical and cerebellar cells from one-day-old and seven- day-old Sprague-Dawley rat pups, respectively were cultured in DMEM/Ham’s F 12 medium (1:1), 10% fetal bovine serum (Biomeda, CA), 100 IU/ml penicillin, and 0.1 mg/ml streptomycine 177. Cells were grown for 8-10 days (37°C, 5% C02) and culture medium was changed every 2 days. The astroglial cell culture of more than 95% was obtained by this procedure. 24 hours prior to treatment with fatty acids, the medium was changed to neuronal cell culture medium. 5.2.2 Immunostaining of Reactive Oxygen Species (ROS) Intracellular reactive oxygen species (ROS) were detected by staining with the oxidant- sensitive dye 5-(6)-chloromethyl-Z',7'-dichlorodihydrofluorescein diacetate (CM- HzDCFDA, from Molecular Probes, OR). HzDCFDA is cleaved of the ester groups by intracellular esterases and converted into membrane impermeable, nonfluorescent derivative H2DCF. Oxidation of I-IzDCF by ROS results in highly fluorescent 2,7- dichlorofluorescein (DCF) '79. The cells were incubated for 30 minutes at 37 0C with 2M CM-HzDCFDA in Hanks’ Balanced Salt Solution without phenol red (Invitrogen). The cells were then washed three times with PBS and analyzed with confocal microscopy (Zeiss LSM 5 Pascal). A 63X oil-immersion objective lens was used for data acquisition. 5.2.3 Western Blot Analysis For Western blotting, the following antibodies were used: BACE] (Chemicon, CA, USA), APP/C99 (QED Bioscience Inc., CA, USA), AT8 (Pierce Biotechnology, IL, USA), PHF-l (from Dr. P. Davies, Albert Einstein, NY, USA), Tau-l (Chemicon), GSK- 3a/B (Chemicon), Phospho GSK-3a/8 (Sigma), MAP Erkl/2 (Cell Signaling), Phospho 74 MAP Erkl/2 (Cell Signaling Technology, MA, USA), cdk5 (Santa Cruz Biotechnology, CA, USA), p35/p25 (Santa Cruz) and actin (Sigma). To extract membrane proteins (BACEI and APP/C99), cells were washed three times with ice-cold TBS (25 mM Tris, pH 8.0, 140 mM NaCl, and 5 mM KCl) and lysed for 20 minutes by scraping into ice-cold radioimmunoprecipitation assay (RIPA) buffer [1% (v/v) Nonidet P40, 0.1% (w/v) SDS, 0.5% (w/v) deoxycholate, 50 mM Tris, pH 7.2, 150 mM NaCl, 1 mM Na3VO4 and 1 mM PMSF, all chemicals from Sigma] 238 . To extract all other proteins, RIPA buffer containing 1% (v/v) Triton, 0.1% (w/v) SDS, 0.5% (w/v) deoxycholate, 20 mM Tris, pH 7.4, 150 mM NaCl, 100 mM NaF, 1 mM Na3VO4, 1 mM EDTA, 1 mM EGTA, and 1 mM PMSF [all chemicals from Sigma] was used '78. The total cell lysate was obtained by centrifugation at 12,000 rpm for 15 minutes at 4 0C. The total protein concentration was measured by BCA protein assay kit from Pierce (Rockford, IL, USA). Equal amounts of total protein from each condition were run at 200 V on SDS-PAGE gels (BioRad, CA, USA). The separated proteins were transferred to nitrocellulose membranes for 1 hour at 100 V and incubated at 4 0C overnight with the appropriate primary antibodies [1:1000 BACE], 1:1000 APP/C99, 1:200 AT8, 1:200 PHF-l, 1:2000 Tau-l, 1:1000 GSK-301/8, 1:1000 Phospho GSK-3a/B, 1:1000 MAP Erk1/2, 1:1000 Phospho MAP Erk1/2, 1:1000 cdk5, 1:1000 P35/p25, 1:500 GLUTl, 1:2000 actin]. Blots were washed three times in PBS-Tween (PBS-T) and incubated with appropriate HRP-linked secondary antibodies (Pierce) diluted in PBS-T for 1 hr at room temperature. After washing three times in PBS-T, blots were developed with the Pierce SuperSignal West Femto Maximum Sensitivity Substrate (Pierce) and imaged with the BioRad ChemiDoc. Quantity One software from Bio-Rad was used to quantify the signal intensity of the protein bands. 75 5.2.4 ELISA measurements of A840 and A842 For AB measurements, the media and neuronal cells were collected after 24 hours of treatment. The media were treated with protease inhibitor cocktail (Sigma, MO, USA) and cleared by brief centrifugation (3000 rpm, 5 minutes, 40C). A840 and A842 in the media were measured by using colorimetric sandwich ELISA according to the manufacturer's instructions (Wako Chemicals, VA, USA). The cells were lysed and the intracellular protein was measured by using the BCA protein assay kit from Pierce (Rockford, IL, USA), which was used to normalize the A040 and A842 values. 5.2.5 Data Analyses Data are shown as means :1: SD. for indicated number of experiments. Student’s t-test and one-way ANOVA with Tukey’s past hac method were used to evaluate statistical significances between different treatment groups. Statistical significance was set at p<0.05. 5.3 RESULTS AND DISCUSSION 5.3.1. Involvement of Astroglial Ceramide in FFA-Astroglia-Induced ROS Production in Neurons In chapter 2, we showed that treatment of neurons with the conditioned media from F FA- treated astroglia increased ROS production in neurons. This suggests a central role of astroglial FFA metabolism in the observed FFA-astroglia-induced ROS production in neurons. Furthermore, our literature-based hypothesis followed by mathematical and 76 experimental studies suggested astroglial ceramide as a possible mediator of ROS production in neurons (as discussed in chapter 4). In this context, here we found that inhibition of de nova synthesis of ceramide in PA-treated astroglia by using 2 mM L-CS, significantly inhibited PA-astroglia—induced ROS production in neurons (Figure 5.2). The treatment of neurons directly with L-CS did not inhibit the PA-astroglia-induced ROS production observed in neurons. This further emphasizes the role of astroglial ceramide in causing ROS production in neurons. Control PA PA+L-CS Figure 5.2. Involvement of astroglial ceramide in PA-astroglia-induced ROS production in neurons. Co-treatment of astroglia with 2mM L-CS inhibited PA-astroglia-induced ROS production in neurons. The neurons were stained with C M-HZDCFDA for intracellular ROS detection and examined with confocal fluorescence microscopy. (Objective lens magnification, 40X). As mentioned earlier, ceramide secreted from astroglia can act directly on neurons and mediate the oxidative stress-induced effects in the neurons 304. In addition, PA-induced 305 increase in ceramide levels can induce the secretion of cytokines (e.g. IL-6) or other signaling molecules, e.g. NO 306 by astrocytes, which in turn may elevate the production 307‘ 308. In this context, we found that PA treatment did not induce of ROS in the neurons IL-6 expression in astroglia, however it increased the level of inducible nitric oxide synthase (iNOS) in astroglia, which was blocked by the co-treatment of astroglia with 2mM L-CS, thus suggesting an involvement of ceramide in astroglial iNOS expression (Figure 5.3). 77 iNOS .............. ——' "“"" lL-6 actin -— “I C PA PA+L-CS Figure 5.3. The expression of iNOS and IL-6 in astroglia. The expression of iNOS, but not IL-6 was increased by PA treatment in astroglia. Co-treatment of astroglia with 2mM L-CS inhibited PA-induced iNOS expression in astroglia. B-actin is shown as a marker for protein loading. 5.3.2 Involvement of astroglial ceramide in FFA-astroglia-induced amyloidogenesis and tau hyperphoshorylation in neurons We treated cortical astroglia with 0.2mM PA for 24 hours and then used the astroglia- conditioned media to treat cortical neurons for 24 hours. After 24 hours of treatment, the media were collected for measurement of secreted A6 levels and the neurons were washed, lysed and the total cellular protein was used for western blot analysis of a number of proteins. As shown in Figure 5.4, treatment of neurons with the conditioned media from PA-treated astroglia significantly increased BACE] levels and consequent amyloidogenic processing of APP leading to the formation of c-terrninal fragments of APP (C99) (Figure 5.4). C99 is processed by y-secretase to form A8. BACEI is a rate- limiting enzyme in the amyloidogenic processing of APP and a slight increase in BACE] levels leads to a dramatic increase in the production of AB40/42 243. Indeed, we found that the PA-astroglia-treated neurons significantly increased secretion of A840 and A842 as compared to controls (Figure 5.4). 78 (A) 180 9c BACEl—> ' .1 5..., Actin 7* -" C PA PA+L-CS BACE1 (‘5 of cancel) C PA PA+L-CS 183 :1: 140 g 120 # 5 g 100 - .3 so- I a .1 8 “I C PA PA+L-CS 2‘“ D 4 PA PA+L-CS (C) A 42 3... AB40 '3 * 350 a 23m 2300 c 9 g 250 # g 250 # g 200 a; 200 39 150 g: 150 O 100 N 100 v wr 21 50 it 50 g 0 C PA PA+L-CS C PA PA+L-CS Figure 5.4. Involvement of astroglial ceramide in PA-astroglla—induced elevations in BACEl and amyloidogenic processing of APP in neurons. In neurons treated with conditioned media from PA-treated astroglia, the levels of (A) BACE], (B) C99 and (C) A840 and A842 were found elevated. These PA- astroglia—induced tau abnormalities were blocked by inhibiting astroglial ceramide synthesis with 2mM L- CS. Data represent mean :t SD. of three independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. ‘, p<0.05 compared with control; it, p<0.05 compared with PA Ireatrnent. 79 In addition, phosphorylation of tau was significantly increased in neurons treated with the conditioned media from PA-treated astroglia as observed by immunoblotting with two different antibodies, PHF-l and AT8 (Figure 5.5). As mentioned earlier, PHF—l and AT8 antibodies recognize tau protein hyperphosphorylated at two different, AD-specific phospho-epitopes. In addition to PHF-l and AT8 antibodies, we also carried out immunoblotting with Tau-1 antibody, which detects all the isoforms of dephosphorylated tau, thus acting as a negative control. As expected, dephosphorylated Tau-1 was significantly decreased in PA-astroglia-treated neurons as compared to control-astroglia- treated neurons (Figure 5.5). 160 * 120 100 AT8 as-“ :3 20 -_-~ 0 PHF'l PA PA+L-CS 140 ‘ * Tau-1 “one. 120 # Actin ‘1 I . 10° 80 C PA PA+L-CS 60 40 20 0 c PA+L-CS Figure 5.5. Involvement of astroglial ceramide in PA-astroglia-induced tau hyperphosphorylation in neurons. In neurons treated with conditioned media from PA-treated astroglia, tau was found pathologically hyperphosphorylated as shown by immunoblotting with PHF-l and AT8 antibodies. Tau-l detects dephosphorylated tau, thus showing decreased levels in PA-astroglia-treated neurons. These PA- astroglia—induced tau abnormalities were blocked by inhibiting astroglial ceramide synthesis with 2mM L- CS. Histograms corresponding to PHF-l and AT8 blots represent quantitative determinations of intensities of the relevant bands normalized with actin. Data represent mean i SD. of three independent experiments. One-way ANOVA with Tukey’s post hoc method was used for analyzing the differences between treatment groups. *, p<0.05 compared with control; #, p<0.05 compared with PA treatment. 80 As PA treatment was found to elevate de nova synthesis of ceramide in astroglia, we wanted to investigate a possible involvement of astroglial ceramide in the observed, PA- astroglia-induced amyloidogenesis and tau hyperphosphorylation in neurons. For this, we treated astroglia with 2mM L-CS together with 0.2mM PA for 24 hours and then transferred the astroglia—conditioned medium to the neurons (24 hours treatment). The inhibition of astroglial ceramide synthesis by L-CS treatment blocked PA-astroglia- induced BACEl upregulation, AB production and hyperphosphorylation of tau (Figures 5.4 and 5.5), strongly suggesting a central role of astroglial ceramide in causing AD- associated pathophysiological changes in neurons. Previously, exogenous addition of 24 - 3 . Furthermore, increased ceramide to neurons has been shown to induce A8 production tau phosphorylation has been reported in cholesterol-deficient neurons, which had a significant increase in ceramide levels 325. Our findings reported here, however, are the first to demonstrate a direct causal role of astroglial PA metabolism and endogenously synthesized ceramide in astroglia, in causing A8 production and tau hyperphosphorylation in neurons, two characteristic signatures of AD pathology. We further studied the possible activation of various AD-related kinases (GSK-3a/8, cdk5 and MAPK Erkl/Z) 75, one or more of which may be responsible for the observed PA-astroglia-induced tau hyperphosphorylation, and potentially activated by ceramide. An increase in the phosphorylation of these enzymes was used as an indicator of their activation. As shown in Figure 5.6A, levels of phosphorylated GSK-3ot/[3 (Tyr279/Tyr216) were significantly increased in PA-astroglia-treated neurons as compared to controls, without any significant change in the levels of total GSK-3a/8. 81 GSK—3a phosphorylated at Tyr279 and GSK3-B phosphorylated at Tyr216 suggest increased activity 326. In addition to activating GSK-3a/l3, PA-treatment also increased the cleavage of cdk5 activator p35 to p25 (Figure 5.6B). p25 accumulates in the brains of AD patients and the conversion of p35 to p25 suggests augmented activity of cdk5 327' 328. Finally, there was no change in the levels of both total and phosphorylated MAPK Erkl/Z in the PA-astroglia-treated neurons as compared to control-astroglia-treated ones (Figure 5.6C), suggesting no change in its activity. The treatment of astroglia with 2mM L-CS inhibited both the PA-astroglia-induced increase in the level of phosphorylated GSK- 301/[3 and the cleavage of p35 to p25 (Figure 5.6A and B). (A) GSK-3a/B- nosrzsa — "3.... fl .. P-GSK-Bp — Us? 011 ’ 6:8 113 - — H u c PA PA+L-CS (B) cdk5- p35 —' p25—e W 0 PA PA+L-CS (C) h'IAP-Er'kl/Z- P-MAP-Erkr-a—II- ~ .5... P-MAP-ErkZ-F ? P INLAP-E kl --—- _—- 'h .. MAP—Eikfd -- . = C PA PA+L-CS Figure 5.6. PA-astroglia-induced activation of AD-speciflc kinases in neurons is mediated by astroglial ceramide. Conditioned media from PA-treated astroglia activated (A) GSK-3 (increased levels of phosphorylated GSK-3) and (B) cdk5 (increased cleavage of p35 to p25) but not (C) MAP- Erkl/Z (no change in the levels of phosphorylated MAP- Erkl/Z). The treatment of astroglia with 2mM L-CS inhibited PA-astroglia-induced activation of both GSK-3 and cdk5. Data are representative of 3 different experiments. 82 To further evaluate the possible involvement of these enzymes in PA-astroglia-induced tau hyperphosphorylation in neurons, we treated neurons with pharmacological inhibitors of these enzymes; lOmM LiCl (for GSK-Ba/B), lOuM Roscovitine (for cdk5) and 3011M PD98059 (for MAPK Erkl/2). Treatment with PD98059 did not inhibit PA-astroglia- induced tau hyperphosphorylation in neurons (Figure 5.7), which is in agreement with the lack of activation of MAP Erk1/2 (Figure 5.6C). Furthermore, although cdk5 was activated in neurons (Figure 5.6B), co-treatment of neurons with a potent cdk5 inhibitor, roscovitine, did not inhibit the observed PA-astroglia-induced hyperphosphorylation of tau (Figure 5.7). This suggests that PA-astroglia-induced hyperphosphorylation of tau in primary rat cortical neurons is independent of the observed activation of the cdk5 . pathway. Our results agree with a previous report that showed that cleavage of p35 to p25 and subsequent activation of cdk5 were not involved in the hyperphosphorylation of tau in primary rat hippocampal neurons 329. On the other hand, GSK-3 inhibitor (LiCl) inhibited the PA-astroglia-induced hyperphosphorylation of tau in neurons (Figure 5.7). Previously, GSK-3 has been shown to be involved in causing both tau hyperphosphorylation and A8 production and thus, is central and essential in the development of AD. GSK-3B is involved in hyperphosphorylation of tau and its other isoforrn, GSK-3a, is involved in AB production by regulating the activity of y-secretase 330' 33'. These studies together with our present data further emphasize the central role of PA-induced, abnormal astroglial ceramide metabolism in causing AD-associated pathophysiological characteristics. 83 AT8 - Hub“ SE PA PA PA PHF-l wfififl" C M . . + LiCl rosco PD98 150 140 Ta“ ~‘_-— 120 100 . so Actm m..— 5° 40 C PA 20 Pf PA PA 0 . + + LICI rosco PD98 C PA Pf P: + .1. v1 me 059 LiCl rosco PD98 vitine 059 Figure 5.7. GSK-3 is involved in PA-astroglia-indueed tau hyperphosphorylation in neurons. Astroglia-conditioned media were transferred to neurons, with or without various kinase inhibitors, viz., lOmM LiC12 (GSK-3 inhibitor), IOmM Roscovitine (cdk-S inhibitor) and 30mM PD98059 (MAPK inhibitor). Immunoblot analysis with PHF-l and AT8 antibodies show. that only LiCl inhibited the observed, PA-induced tau hyperphosphorylation in neurons. Histogram data represent mean i SD. of three independent experiments. One-way ANOVA and I‘ukey’s post hoc method was used for analyzing the differences between treatment groups. ‘, p<0.05 compared with control; #, p<0.05 compared with PA treatment. 5.3.3 A possible explanation for the region-specific and cell type-specific damage observed in AD AD is a peculiar neurodegenerative disease in that the observed pathophysiological and metabolic damages are found to be region-specific and cell type-specific Basal forebrain, cortex and hippocampus are affected while cerebellum is relatively spared. Also, cholinergic neurons are most affected 332’ 333. Here, it is interesting to note that different brain regions show differential FFA metabolic activities; the activity of fatty acyl—CoA synthetase (FACS) is 10X lower in cerebellum as compared to the other affected brain regions 334. FACS is the first enzyme involved in cellular FFA metabolism, which 84 converts fatty acid to fatty acyl-CoA, which is then utilized by the cells in catabolic (e. g. 8-oxidation) and anabolic (e. g. ceramide synthesis) pathways 335. ceramide production in cerebellum is 2X and 4X lower as compared to cortex and hippocampus, respectively 336. These studies together with our present data may suggest that under pathologically elevated levels of saturated FF As, cerebellum may be less likely to be affected by abnormal F FA metabolism due to its lower activity level of F ACS and consequent, lower levels of ceramide, as compared to cortex and hippocampus. This may explain in part the region-specific damage observed in AD brain. To further investigate this hypothesis, we carried out experiments, whereby we treated cortical neurons (CTN) with the conditioned media from cortical astrocytes (CTA) and from cerebellar astrocytes (CBA). In line with the results discussed earlier, the conditioned media from PA-treated CTA increased tau hyperphosphorylation in CTN (Figure 5.8A). The conditioned media from PA-treated CBA, however, did not increase the phosphorylation of tau in CTN (Figure 5.83). Furthermore, we had previously shown that FFA metabolism by astroglia results in increased reactive oxygen species (ROS) production in neurons 337’ 338. The elevated ROS, in turn, were found to be involved in causing BACEl upregulation and tau 337' 338, suggesting a central role of oxidative stress in hyperphosphorylation in neurons the FFA-induced damage. Thus, elevated FFA metabolism associated with higher FACS activity in the affected regions (basal forebrain, cortex and hippocampus) may lead to increased oxidative stress in these regions compared to unaffected ones (cerebellum). In this context, it is interesting to note that in AD brains, oxidative stress markers are higher in the affected regions than in the unaffected ones 339’ 34°. The increased oxidative stress 85 may be particularly damaging to the basal forebrain cholinergic neurons as these neurons have been shown to lack an important anti-oxidative enzyme, seleno-glutathione peroxidase 3'”. This may account, in part, for the cell type-specific damage observed in AD pathology; substantial loss of basal forebrain cholinergic neurons is a distinct feature of AD 342_ Furthermore, these basal forebrain cholinergic neurons, through their long ascending projections, innervate cortical and hippocampal regions 343 and the increased oxidative stress in these regions of AD brain may lead to the degeneration of these projections from the cholinergic neurons. In this context, it is also interesting to note that A8 plaques, hallmarks of AD, have been shown to be specifically located where the projection of the basal forebrain cholinergic neurons degenerate 344' 3“. (A) CTN-CTA (B) CTN-CBA . .mu)”- 1!. ‘ * AT8 a“ AT8 a... _ . Actin “d Ami“ m“ C PA C PA 160 a 140 120 120 100 1 100 so ‘ 80 60 6o 40 40 20 20 o o Figure 5.8. Differential effects of cortical and cerebellar astroglia on cortical neurons. Cortical neurons (CTN) were treated with conditioned media from (A) cortical astroglia (CTA) and (B) cerebellar astroglia (CBA). Western blot analysis of hyperphosphorylated tau was performed using AT8 antibody. Cortical astroglia but not cerebellar astroglia were involved in PA-induced tau hyperphosphorylation in neurons. Data represent mean i SD. of three independent experiments. Student‘s t-test was used for analyzing differences between difierent treatment groups. *. p<0.05 compared with control. 86 5.4 CONCLUSIONS In conclusion, astroglial ceramide was found to be involved in the observed PA-astroglia- induced ROS production in neurons. Astrolglial ceramide was also found involved in the PA-astroglia-induced hyperphosphorylation of tau in neurons. Although astroglial ceramide activated both GSK-3a/B and cdk5 in neurons, only GSK-3a/f3 was found to be involved in PA-astroglia-induced hyperphosphorylation of tau. Furthermore, Astroglial ceramide increased BACE] levels and consequent amyloidogenic processing of APP leading to the production of AB40/42. Thus, the present results establish a central role of astroglial fatty acid metabolism and consequent increase in the de novo synthesis of astroglial ceramide in causing two of the major pathophysiological changes associated with AD, tau hyperphosphorylation and AB production. 87 CHAPTER 6 CONCLUSIONS AND FUTURE DIRECTIONS 6.1 Conclusions The objective of this dissertation was to investigate the potential involvement of saturated fatty acids in causing AD-associated pathophysiological and abnormal metabolic changes. Our studies established a complex functional interaction between neuronal and non-neuronal (astroglial) cells, leading to the AD-specific abnormalities under the conditions of pathologically elevated levels of saturated fatty acids. It was shown that saturated fatty acids had no direct deleterious effects on neurons; however, they caused increased oxidative stress in neurons through astroglial mediation. Fatty acid-induced oxidative stress played a central role in the hyperphosphorylation of tau and amyloidogenic processing of APP, two of the important characteristics of AD pathology. Both metabolic modeling (MFA) and experimental data suggested a key role of astroglial ceramide in causing F FA-induced, AD-associated abnormalities in neurons. Note that this was the first-ever attempt to apply MFA to comprehensively study the primary astroglial metabolism. Our data place “astroglial fatty acid metabolism” at the center of the pathogenic cascade in AD and also suggest “astroglial ceramide” as a potentially important target for therapeutic intervention in AD. Based on our findings, we hypothesize the following sequence of cellular events by which saturated FFAS may play a central role in the pathogenesis of AD (Figure 6.1). The brain experiences chronically elevated levels of saturated FFAS. Saturated FFAS are taken up and metabolized by astroglia, downregulating astroglial GLUTl levels and glucose metabolism. Astroglial FFA metabolism also results in increased levels of 88 ceramides. Ceramides then induce secretion of cytokines (e.g. IL-IB, TNF-a etc.) or other signaling molecules (e.g. NO, due to increased expressed iNOS expression) by astroglia, which may induce ROS production in neurons. Increased oxidative stress in neurons causes BACEI upregulation and GSK-3 activation resulting in increased AB production and hyperphosphorylation of tau, respectively. The sequence of events suggested here is similar to that observed in AD pathology- decreased glucose metabolism is an early event, which together with increased oxidative stress precedes the pathophysiological changes observed in AD (NFTS and AB plaques). Furthermore, the present “FFA-AD” hypothesis also provides a possible explanation for the region-specific and cell type-specific damage observed in AD (Chapter 5). Astrocyte ,‘ APP Secreted Factor(s) ceramide f A Abetn Production A RACF'l A ROS ‘ Tau‘ Phosphorylnlion ‘ i ' ,- No direct effect (GSK—E) ‘ ‘_ , on neurons Neuron Figure 6.1. The “FFA-AD” hypothesis. Proposed cellular mechanism by which astroglial FFA metabolism may play a central role in causing pathophysiological and metabolic changes associated with AD. 89 The low level of ceramide production in cerebellar astroglia as compared to cortical and hippocampal astroglia 336 , may play a key role in the little damage observed in cerebellum of AD brain as opposed to other regions. Due to the low flux of saturated FFAS through sphingolipid pathway in cerebellar astroglia, FFAS may be diverted to other metabolic pathways, e. g. B-oxidation of fatty acids, which may lead to the increased production of ketone bodies. Here it is interesting to note that ketone bodies have been shown to act as anti-oxidants in that they protect neuronal cells form AIS-induced oxidative damage 346. This may further explain why cerebellum is relatively spared in AD. It is also noteworthy that the single-most factor strongly correlated with AD is aging. Therefore, any hypothesis to explain AD pathology must provide an explanation for the correlation between aging and incidence of AD. Increased oxidative stress has been strongly associated with aging, which might be a result of decreased anti-oxidant enzymatic capacity with aging 54' 347. Under these conditions, enhanced production of ROS induced by saturated FFAS may make the brain more vulnerable to increased oxidative damage as compared to age-matched controls with low levels of saturated FFAS. Thus, our present hypothesis may explain the link between age and the higher incidence of AD. Despite these supporting data, our “FFA-AD” hypothesis is unable to explain the AD pathology comprehensively at this juncture. Specifically, although our cell-culture studies showed potential involvement of saturated fatty acids in causing both the 90 amyloidogenesis and the hyperphoshorylation of tau, in animals high fat diet has been shown to cause only increased AB production but not the tau hyperphosphorylation and the NFT formation associated with it 122424 . The reason behind these disparities between in vitro and in vivo findings is not well understood. In addition, some studies suggest the tau protein abnormalities initiate the AD cascade, while others emphasize AB deposits as the causative factors in AD 348. Our present data is unable to shed any light on this ongoing debate. Furthermore, in the present studies we established a central role of FA—induced oxidative stress in causing AD-associated abnormalities. However, oxidative stress has been shown to be central to many other brain diseases, e. g. Parkinson's disease 349, in addition to AD. Therefore, it is not clear how FFA-induced oxidative stress is specific to AD pathology. In other words, it is not clear how FFA-induced oxidative stress would lead to AD- specific changes but would not induce abnormalities associated with other diseases, in which oxidative stress plays a key role. All the high fat diet animal models show only AD-specific changes in their brain. Our current data does not provide any explanation for this important, fundamental question. In addition, AD follows a specific spatio-temporal pattern of neurodegeneration, where neurodegeneration starts at the basal forebrain and with time proceeds to entorhinal cortex, hippocampus, parts of limbic system and associative cortex 350. The basal forebrain cholinergic neurons lack an important anti-oxidative enzyme, seleno- 4 glutathione peroxidase 3 '. This may explain their higher vulnerability to the FFA- 91 induced increased oxidative stress. It is not clear, however, how saturated fatty acids would contribute to the characteristic spatio-temporal neuronal damage observed in AD. F urtherrnore, our present data explain the potential involvement of saturated fatty acids in the increased AB production. However, it is not just the increased production of AB, but also its aggregation that is important in AD pathology. To be specific, neither monomeric nor mature aggregated polymeric forms, but the intermediate oligomeric forms of AB are responsible for the AD-associated neurotoxicity 35'. In fact, a very recent study showed that accelerating AB fibrillization which reduced AB oligomer levels, helped in reducing functional deficits in AD mouse models 352. Our present data do not answer if (and how) saturated fatty acids play a role in the AB oligomerization. Here, it is also important to note that the risk of AD is higher in women as compared to men 353. It is not clear if saturated fatty acids play any role in the gender-specificity associated with AD. The decreased level of estrogen hormone in menopausal women has been suggested to increase the risk for AD in women 354. It is not clear at present, if there is any correlation between saturated fatty acids and estrogen levels, that may lead to increased risk for AD development in women as compared to men, under the condition of elevated levels of saturated FFAS. Finally, our present studies established a central role of astroglia in causing FFA-induced, AD-associated abnormalities. The critical role of astroglia in AD pathology has been suggested previously by many studies. Increased expression of iNOS in astroglia and 92 consequent increase in NO levels has been shown to stimulate AB production and hyperphosphorylation of tau in neurons 355' 356. Furthermore, reactive astrogliosis associated with AD has been suggested to induce glutamate release, which may lead to excitotoxicity and cell death in neurons 357' 358. In addition, astroglial apoptosis has been associated with AD and AB has been shown to induce astroglial cell death 359’ 360. This astroglial cell death may result in the “loss of good function”, to support the neurons under normal conditions. Together these data place astroglia at the centre stage of AD pathology. However, the major limitation of all these studies, including ours, is that the astroglia are used in these studies as a whole population. It would be worthwhile to investigate the possible involvement of Type I and Type II astroglia separately, in causing AD-associated damage. These data may prove invaluable in finding novel clues that may further help in establishing the in-depth disease mechanism. 6.2 Future Directions Our cell-culture based studies presented in this dissertation have provided important information regarding the key role of saturated fatty acids in causing AD-associated abnormalities. As discussed above, many questions remain unanswered and thus, need further scientific investigation. The focus of the future investigation will be specifically on the following studies as discussed below. 6.2.1 In vivo studies Future work should focus on animal studies where cerebral FFA metabolism will be studied in terms of the de novo synthesis of ceramide at the regional (e.g. cortex, 93 hippocampus vs. cerebellum) as well as sub-cellular (astroglia vs. neurons) levels in response to various diets and stimuli in APP transgenic mice, e.g. Tg2576. This line of mice expresses human APP695 with the 670/671 “Swedish” double mutation and show clear age-dependent AB deposition and memory deficits 39. It would be a significant step forward in AD research if saturated FFAS are shown to exert their risk for the development of AD in vivo, through a similar mechanism as observed in the current in vitro studies. Furthermore, the potential importance of ceramide as a therapeutic target for AD should also be further studied in these animals by using pharmacological inhibitors of de novo synthesis of ceramide, e.g. L-CS, D-serine, myriocin (ISP-l), fumonisin Bl, viridiofungin A, sphingofungin B or lipoxarnycin. Decreased AB production and its deposition in the brains of these mice would serve as measures of the potential protective effects of these ceramide inhibitors. 6.2.2 Delivery of ceramide inhibitors through the blood-brain barrier (BBB) With the use of ceramide inhibitors in vivo, one of the greatest challenges, which also presents a great research opportunity, is to find ways for efficient transport of these inhibitors across the BBB. In its neuroprotective role, the BBB prevents the delivery of many important therapeutic agents to the brain; more than 98% of currently available therapeutics cannot pass through the BBB 361. Future studies should investigate the use of novel delivery systems, e.g. nanoparticles, as carriers of ceramide inhibitors to the brain. In addition to their ability to cross the BBB, these vehicles should also be engineered so as to deliver ceramide inhibitors specifically to astroglia, e.g. by attaching astroglia- 94 specific antibody (GFAP) to nanOparticles. In addition to their use in AD, these successful delivery vehicles will also prove useful in treating other brain diseases. 6.2.3 Studying pathways downstream to ceramide One of the major limitations with ceramide as a therapeutic target is its important role as a signaling molecule in many physiological processes. In addition, the currently available ceramide inhibitors mentioned earlier have very high toxicities, weak inhibition activity and also exhibit low specificity 362' 363. Thus, it would also be worthwhile to focus future studies on the pathways downstream of ceramide generation that may be involved in FF A-induced, AD-associated abnormalities, e.g. ceramide-induced secretion of inflammatory cytokines or other signaling molecules such as NO from astroglia. 95 APPENDIX 1. Brain cells from older animals In our studies we used cortical neurons and astroglia from 1-2-day old rat pups. As AD is strongly associated with aging, use of brain cells from older animals would be more appropriate for these AD studies. However, it is difficult to isolate the cortical cells from the brains of older rats and their viability reduced significantly; the cortical cells isolated from 7-day old rat pups started dying after 2 days in culture. Day 1 Day 2 2. Trans-well experiments In our studies, the conditioned media from astroglia were transferred to neurons, so the two cell types did not share the same growth enviromnent. Physiologically, however, neurons and astroglia are in close proximity and share common growth environment where secreted factors fiorn both the neurons and astroglia may affect both these cell types. We investigated the possible effect of potential factors secreted from the neurons that may affect astroglia, which in turn may modulate the observed astroglial effects on 96 the neurons using trns-well tissue culture. The astroglia were plated in the well inserts and neurons in the wells. The cells were then treated with either 0.2mM PA or 4% BSA (control) and levels of BACE] and phosphorylated tau in neurons were studied. We found that PA-treatment significantly increased levels of BACE] and phosphorylated tau in neurons, similar to our non-trans-well experiments. W tau (AT8) “M” '~ BACE1 w W ‘4‘“ arm"! PHI-t. we!" actin n - (3 PA 3. Exogenous addition of ceramide as a positive control Our studies showed a central role of astroglial ceramide in the FFA-astroglia-induced, AD-associated pathophysiological changes observed in neurons. The role of astroglial ceramide was confirmed by HPLC measurement of elevated ceramide and also by using a pharmacological inhibitor of ceramide synthesis in astrolgia. As a positive control, we exogenously added 1051M C-6 ceramide (a synthetic analog of ceramide, Sigma) to astroglia for 24hr, followed by the transfer of the astroglia-conditioned media to neurons (24hr treatment). We found that C-6 ceramide significantly increased levels of BACEl 97 and phosphorylated tau in neurons, thus further emphasizing role of ceramide in causing AD-associated abnormalities. tau (AT8) H'- H BACE1 m, actin - .1 C ceramide 98 LIST OF PUBLICATIONS This thesis is based on our following original publications- 1) 2) 3) 4) 5) Patil, S. and Chan, C., “Palmitic and Stearic fatty acids induce Alzheimer-like hyperphosphorylation of tau in primary rat cortical neurons”, Neuroscience Letters, 384: 288-293 (2005). Patil, S., Lufang, S., Masserang, A. and Chan, C., “Palmitic acid-treated astrocytes induce BACE] upregulation and accumulation of C-terrninal fragment of APP in primary cortical neurons”, ”, Neuroscience Letters, 406: 55-59 (2006). Patil, S., Li, Z. and Chan, 0, “Cellular to Tissue Informatics: Approaches to Optimizing Cellular Function of Engineered Tissue", Advances in Biochemical Engineering / Biotechnology, eds. K. Lee and D. Kaplan, 102: 139-159 (2006). Patil, S., Melrose, J. and Chan, C., “Involvement of astroglial ceramide in palmitic acid-induced Alzheimer-like changes in primary neurons”, (Accepted, European Journal of Neuroscience). Patil, S., Balu, D., Melrose J. and Chan, C., “Brain region specificity of palmitic acid-induced Alzheimer-like changes in primary neurons”, (In Preparation). 99 BIBLIOGRAPHY l. Korczyn, A. D.; Vakhapova, V., The prevention of the dementia epidemic. J Neurol Sci 2007, 257, (l-2), 2-4. 2. Wang, D. C.; Chen, S. S.; Lee, Y. C.; Chen, T. J., Amyloid-beta at sublethal level impairs BDNF-induced arc expression in cortical neurons. Neurosci Lett 2006, 398, (l- 2), 78-82. 3. Mattson, M. P., Pathways towards and away from Alzheimer's disease. Nature 2004, 430, (7000), 631-9. 4. Mosconi, L., Brain glucose metabolism in the early and specific diagnosis of Alzheimer's disease. EurJ Nucl Med M01 Imaging 2005, 32, (4), 486-510. 5. Ogawa, M.; Watabe, H.; Teramoto, N.; Miyake, Y.; Hayashi, T.; Iida, H.; Murata, T.; Magata, Y., Understanding of cerebral energy metabolism by dynamic living brain slice imaging system with [18F]FDG. Neurosci Res 2005, 52, (4), 357-361. 6. Goate, A.; Chartier-Harlin, M. C.; Mullan, M.; Brown, J .; Crawford, F .; Fidani, L.; Giuffra, L.; Haynes, A.; Irving, N.; James, L., Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature 1991, 349, (6311), 704-6. 7. Mullan, M.; Crawford, F .; Axelrnan, K.; Houlden, H.; Lilius, L.; Winblad, B.; Lannfelt, L. A pathogenic mutation for probable Alzheimer's disease in the APP gene at the N-terrninus of beta-amyloid. Nat Genet 1992, (1), 345-7. 8. Sherrington, R.; Rogaev, IE. 1.; Liang, Y.; Rogaeva, E. A.; Levesque, G.; Ikeda, M.; Chi, H.; Lin, C.; Li, G.; et al., Cloning of a gene bearing missense mutations in early- onset familial Alzheimer's disease. Nature 1995, 375, (6534), 754-60. 9. Levy-Lahad, E.; Wasco, W.; Poorkaj, P.; Romano, D. M.; Oshima, 1.; Pettingell, W. H.; Yu, C. E.; Jondro, P. D.; Schmidt, S. D.; Wang, K., Candidate gene for the chromosome 1 familial Alzheimer's disease locus. Science 1995, 269, (5226), 973-7. 10. Hoyer, S., Brain glucose and energy metabolism abnormalities in sporadic Alzheimer disease. Causes and consequences: an update. Exp Gerantal 2000, 35, (9-10), 1363-1372. 100 11. Rogaeva, E. A.; Fafel, K. C.; Song, Y. Q.; Medeiros, H.; Sato, C.; Liang, Y.; Richard, E.; Rogaev, E. 1.; Frommelt, P.; Sadovnick, A. D.; Meschino, W.; Rockwood, K.; Boss, M. A.; Mayeux, R.; St George-Hyslop, P., Screening for PS1 mutations in a referral-based series of AD cases: 2] novel mutations. Neurology 2001, 57, (4), 621-5. 12. Carder, E. H.; Saunders, A. M.; Strittmatter, W. J.; Schmechel, D. E.; Gaskell, P. C.; Small, G. W.; Roses, A. D.; Haines, J. L.; Pericak-Vance, M. A., Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 1993, 261, (5123), 921-3. 13. Haan Mary, N.; Wallace, R., Can dementia be prevented? Brain aging in a population-based context. Annu Rev Public Health 2004, 25, 1-24. 14. Alzheimer, A.; Stelzmann, R. A.; Schnitzlein, H. N.; Murtagh, F. R., An English translation of Alzheimer's 1907 paper, "Uber eine eigenartige Erkankung der Hirnrinde". Clin Anat 1995, 8, (6), 429-31. 15. Wirno, A.; Jonsson, L.; Winblad, B., An estimate of the worldwide prevalence and direct costs of dementia in 2003. Dement Geriatr Cogn Disord 2006, 21, (3), 175-81. 16. Fratiglioni, L.; De Ronchi, D.; Aguero-Torres, H., Worldwide prevalence and incidence of dementia. Drugs Aging 1999, 15, (5), 365-75. 17. Wolf-Klein, G.; Pekmezaris, R.; Chin, L.; Weiner, J., Conceptualizing Alzheimer's disease as a terminal medical illness. Am J Hosp Palliat Care 2007, 24, (1), 77-82. 18. Selkoe, D. J ., Toward a comprehensive theory for Alzheimer's disease. Hypothesis: Alzheimer's disease is caused by the cerebral accumulation and cytotoxicity of amyloid beta-protein. Ann N Y Acad Sci 2000, 924, 17-25. 19. Perry, E. K.; Tomlinson, B. E.; Blessed, G.; Bergmann, K.; Gibson, P. H.; Perry, R. H., Correlation of cholinergic abnormalities with senile plaques and mental test scores in senile dementia. Br Med J 1978, 2, (6150), 1457-9. 20. Cummings, B. J .; Cotrnan, C. W., Image analysis of beta-amyloid load in Alzheimer's disease and relation to dementia severity. Lancet 1995, 346, (8989), 1524-8. 101 21. Citron, M.; Oltersdorf, T.; Haass, C.; McConlogue, L.; Hung, A. Y.; Seubert, P.; Vigo-Pelfrey, C.; Lieberburg, 1.; Selkoe, D. J ., Mutation of the beta-amyloid precursor protein in familial Alzheimer's disease increases beta-protein production. Nature 1992, 360, (6405), 672-4. 22. Cai, X. D.; Golde, T. E.; Younkin, S. G., Release of excess amyloid beta protein from a mutant amyloid beta protein precursor. Science 1993, 259, (5094), 514-6. 23. Haass, C.; Hung, A. Y.; Selkoe, D. J.; Teplow, D. B., Mutations associated with a locus for familial Alzheimer's disease result in alternative processing of amyloid beta- protein precursor. J Biol Chem 1994, 269, (26), 17741-8. 24. Suzuki, N.; Cheung, T. T.; Cai, X. D.; Odaka, A.; Otvos, L., Jr.; Eckman, C.; Golde, T. E.; Younkin, S. G., An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants. Science 1994, 264, (5163), 1336-40. 25. Citron, M.; Vigo-Pelfrey, C.; Teplow, D. B.; Miller, C.; Schenk, D.; Johnston, J.; Winblad, B.; Venizelos, N.; Lannfelt, L.; Selkoe, D. J., Excessive production of amyloid beta-protein by peripheral cells of symptomatic and presymptomatic patients carrying the Swedish familial Alzheimer disease mutation. Proc Natl Acad Sci U S A 1994, 91, (25), 11993-7. 26. Scheuner, D.; Eckman, C.; Jensen, M.; Song, X.; Citron, M.; Suzuki, N.; Bird, T. D.; Hardy, J.; Hutton, M.; Kukull, W.; Larson, E.; Levy-Lahad, E.; Viitanen, M.; Peskind, E.; Poorkaj, P.; Schellenberg, G.; Tanzi, R.; Wasco, W.; Lannfelt, L.; Selkoe, D.; Younkin, S., Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin l and 2 and APP mutations linked to familial Alzheimer's disease. Nat Med 1996, 2, (8), 864-70. 27. Citron, M.; Westaway, D.; Xia, W.; Carlson, G.; Diehl, T.; Levesque, G.; Johnson-Wood, K.; Lee, M.; Seubert, P.; Davis, A.; Kholodenko, D.; Matter, R; Sherrington, R.; Perry, 3.; Yao, H.; Strome, R.; Lieberburg, 1.; Rommens, J.; Kim, S.; Schenk, D.; Fraser, P.; St George Hyslop, P.; Selkoe, D. J ., Mutant presenilins of Alzheimer's disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice. Nat Med 1997, 3, (1), 67-72. 28. Duff, K.; Eckman, C.; Zehr, C.; Yu, X.; Prada, C. M.; Perez-tut, J.; Hutton, M.; Buee, L.; Harigaya, Y.; Yager, D.; Morgan, D.; Gordon, M. N.; Holcomb, L.; Refolo, L.; Zenk, B.; Hardy, J .; Younkin, S., Increased amyloid-beta42(43) in brains of mice expressing mutant presenilin 1. Nature 1996, 383, (6602), 710-3. 102 29. Schmechel, D. E.; Saunders, A. M.; Strittrnatter, W. J.; Crain, B. J.; Hulette, C. M.; Joo, S. H.; Pericak-Vance, M. A.; Goldgaber, D.; Roses, A. D., Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc Natl Acad Sci U S A 1993, 90, (20), 9649-53. 30. Rebeck, G. W.; Reiter, J. S.; Strickland, D. K.; Hyman, B. T., Apolipoprotein E in sporadic Alzheimer's disease: allelic variation and receptor interactions. Neuron 1993, ll, (4), 575-80. 31. Greenberg, S. M.; Rebeck, G. W.; Vonsattel, J. P.; Gomez-Isla, T.; Hyman, B. T., Apolipoprotein E epsilon 4 and cerebral hemorrhage associated with amyloid angiopathy. Ann Neurol 1995, 38, (2), 254-9. 32. Polvikoski, T.; Sulkava, R.; Haltia, M.; Kainulainen, K.; Vuorio, A.; Verkkoniemi, A.; Niinisto, L.; Halonen, P.; Kontula, K., Apolipoprotein E, dementia, and cortical deposition of beta-amyloid protein. N Engl J Med 1995, 333, (19), 1242-7. 33. Lemere, C. A.; Blusztajn, J. K.; Yamaguchi, H.; Wisniewski, T.; Saido, T. C.; Selkoe, D. J ., Sequence of deposition of heterogeneous amyloid beta-peptides and APO E in Down syndrome: implications for initial events in amyloid plaque formation. Neurobiol Dis 1996, 3, (1), 16-32. 34. Pike, C. J.; Burdick, D.; Walencewicz, A. J.; Glabe, C. G.; Cotrnan, C. W., Neurodegeneration induced by beta-amyloid peptides in vitro: the role of peptide assembly state. J Neurosci 1993, 13, (4), 1676-87. 35. Lorenzo, A.; Yankner, B. A., Beta-amyloid neurotoxicity requires fibril formation and is inhibited by congo red. Proc Natl Acad Sci U S A 1994, 91, (25), 12243-7. 36. Meda, L.; Cassatella, M. A.; Szendrei, G. 1.; Otvos, L., Jr.; Baron, P.; Villalba, M.; Ferrari, D.; Rossi, F., Activation of microglial cells by beta-amyloid protein and interferon-gamma. Nature 1995, 374, (6523), 647-50. 37. Games, D.; Adams, D.; Alessandrini, R.; Barbour, R.; Berthelette, P.; Blackwell, C.; Carr, T.; Clemens, J .; Donaldson, T.; Gillespie, F.; et al., Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein. Nature 1995, 373, (6514), 523-7. 38. Masliah, E.; Sisk, A.; Mallory, M.; Mucke, L.; Schenk, D.; Games, D., Comparison of neurodegenerative pathology in transgenic mice overexpressing V717F 103 beta-amyloid precursor protein and Alzheimer's disease. J Neurosci 1996, 16, (18), 5795- 811. 39. Hsiao, K.; Chapman, P.; Nilsen, S.; Eckman, C.; Harigaya, Y.; Younkin, S.; Yang, F .; Cole, G., Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 1996, 274, (5284), 99-102. 40. Blurton-Jones, M.; Laferla, F. M., Pathways by which Abeta facilitates tau pathology. Curr Alzheimer Res 2006, 3, (5), 437-48. 41. Price, J. L., Aging, preclinical Alzheimer disease, and early detection. Alzheimer Dis Assoc Disord 2003, 17 Suppl 2, 860-2. 42. Birmingham, K.; F rantz, S., Set back to Alzheimer vaccine studies. Nat Med 2002, 8, (3), 199-200. 43. Koudinov, A. R.; Koudinova, N. V., Cholesterol homeostasis failure as a unifying cause of synaptic degeneration. J Neurol Sci 2005, 229-230, 233-40. 44. Jick, H.; Zomberg, G. L.; Jick, S. S.; Seshadri, S.; Drachman, D. A., Statins and the risk of dementia. Lancet 2000, 356, (9242), 1627-31. 45. Wolozin, B.; Kellrnan, W.; Ruosseau, P.; Celesia, G. G.; Siegel, G., Decreased prevalence of Alzheimer disease associated with 3-hydroxy-3-methyglutaryl coenzyme A reductase inhibitors. Arch Neurol 2000, 57, (10), 1439-43. 46. Refolo, L. M.; Malester, B.; LaFrancois, J .; Bryant-Thomas, T.; Wang, R.; Tint, G. S.; Sambamurti, K.; Duff, K.; Pappolla, M. A., Hypercholesterolemia accelerates the Alzheimer's amyloid pathology in a transgenic mouse model. Neurobiol Dis 2000, 7, (4), 321-31. 47. Sparks, D. L.; Scheff, S. W.; Hunsaker, J. C., 3rd; Liu, H.; Landers, T.; Gross, D. R., Induction of Alzheimer-like beta-amyloid irnmunoreactivity in the brains of rabbits with dietary cholesterol. Exp Neurol 1994, 126, (1), 88-94. 48. Bodovitz, S.; Klein, W. L., Cholesterol modulates alpha-secretase cleavage of amyloid precursor protein. J Biol Chem 1996, 271, (8), 4436-40. 104 49. Racchi, M.; Baetta, R.; Salvietti, N.; Ianna, P.; Franceschini, G.; Paoletti, R.; Fumagalli, R.; Govoni, S.; Trabucchi, M.; Soma, M., Secretory processing of amyloid precursor protein is inhibited by increase in cellular cholesterol content. Biochem J 1997, 322 (Pt 3), 893-8. 50. Simons, M.; Keller, P.; De Strooper, B.; Beyreuther, K.; Dotti, C. G.; Simons, K., Cholesterol depletion inhibits the generation of beta-amyloid in hippocampal neurons. Proc Natl Acad Sci U S A 1998, 95, (11), 6460-4. 51. Morell, P.; Jurevics, 11., Origin of cholesterol in myelin. Neurochem Res 1996, 21, (4), 463-70. 52. Fan, Q. W.; Yu, W.; Senda, T.; Yanagisawa, K.; Michikawa, M., Cholesterol- dependent modulation of tau phosphorylation in cultured neurons. J Neurochem 2001, 76, (2), 391-400. 53. Sawamura, N.; Gong, J. S.; Garver, W. S.; Heidenreich, R. A.; Ninomiya, H.; Ohno, K.; Yanagisawa, K.; Michikawa, M., Site-specific phosphorylation of tau accompanied by activation of mitogen-activated protein kinase (MAPK) in brains of Niemann-Pick type C mice. J Biol Chem 2001, 276, (13), 10314-9. 54. Harman, D., Aging: a theory based on free radical and radiation chemistry. J Gerontol 1956, 11, (3), 298-300. 55. Smith, M. A.; Harris, P. L.; Sayre, L. M.; Perry, G., Iron accumulation in Alzheimer disease is a source of redox-generated free radicals. Proc Natl Acad Sci U S A 1997, 94, (18), 9866-8. 56. Sayre, L. M.; Perry, G.; Harris, P. L.; Liu, Y.; Schubert, K A.; Smith, M. A., In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer's disease: a central role for bound transition metals. J Neurochem 2000, 74, (1), 270-9. 57. Oteiza, P. 1., A mechanism for the stimulatory effect of aluminum on iron-induced lipid peroxidation. Arch Biochem Biophys 1994, 308, (2), 374-9. 58. Good, P. F.; Perl, D. P.; Bierer, L. M.; Schmeidler, J., Selective accumulation of aluminum and iron in the neurofibrillary tangles of Alzheimer's disease: a laser microprobe (LAMMA) study. Ann Neurol 1992, 31, (3), 286-92. 105 59. Cras, P.; Kawai, M.; Siedlak, S.; Mulvihill, P.; Gambetti, P.; Lowery, D.; Gonzalez-DeWhitt, P.; Greenberg, 3.; Perry, G., Neuronal and microglial involvement in beta-amyloid protein deposition in Alzheimer's disease. Am J Pathol 1990, 137, (2), 241 - 6. 60. Colton, C. A.; Gilbert, D. L., Production of superoxide anions by a CNS macrophage, the microglia. FEBS Lett 1987, 223, (2), 284-8. 61. Good, P. P.; Werner, P.; Hsu, A.; Olanow, C. W.; Perl, D. P., Evidence of neuronal oxidative damage in Alzheimer's disease. Am J Pathol 1996, 149, (1), 21-8. 62. Yan, S. D.; Yan, S. P.; Chen, X.; Fu, J.; Chen, M.; Kuppusamy, P.; Smith, M. A.; Perry, G.; Godman, G. C.; Nawroth, P.; et al., Non-enzymatically glycated tau in Alzheimer's disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid beta-peptide. Nat Med 1995, 1, (7), 693-9. 63. Yan, S. D.; Chen, X.; Schmidt, A. M.; Brett, J.; Godman, G.; Zou, Y. S.; Scott, C. W.; Caputo, C.; Frappier, T.; Smith, M. A.; et al., Glycated tau protein in Alzheimer disease: a mechanism for induction of oxidant stress. Proc Natl Acad Sci U S A 1994, 91, (16), 7787-91. 64. Munch, G.; Kuhla, B.; Luth, H. J .; Arendt, T.; Robinson, S. R., Anti-AGEing defences against Alzheimer's disease. Biochem Soc Trans 2003, 31, (Pt 6), 1397-9. 65. Tamagno, E.; Bardini, P.; Obbili, A.; Vitali, A.; Borghi, R.; Zaccheo, D.; Pronzato, M. A.; Danni, 0.; Smith, M. A.; Perry, G.; Tabaton, M., Oxidative Stress Increases Expression and Activity of BACE in NT2 Neurons. Neurobiol Dis 2002, 10, (3), 279-288. 66. Tamagno, E.; Guglielrnotto, M.; Bardini, P.; Santoro, G.; Davit, A.; Di Simone, D.; Danni, 0.; Tabaton, M., Dehydroepiandrosterone reduces expression and activity of BACE in NT2 neurons exposed to oxidative stress. Neurobiol Dis 2003, 14, (2), 291-301. 67. Frederikse, P. H.; Garland, D.; Zigler, J. S., Jr.; Piatigorsky, J., Oxidative stress increases production of b-amyloid precursor protein and b-amyloid (A.b) in mammalian lenses, and A.b has toxic effects on lens epithelial cells. J Biol Chem 1996, 271, (17), 10169-74. 106 68. Zhang, L.; Zhao, B.; Yew, D. T.; Kusiak, J. W.; Roth, G. 8., Processing of Alzheimer's amyloid precursor protein during H202-induced apoptosis in human neuronal cells. Biochem Biophys Res Commun 1997, 235, (3), 845-848. 69. Olivieri, G.; Baysang, G.; Meier, F.; Muller-Spahn, F.; Stahelin, H. B.; Brockhaus, M.; Brack, C., N-acetyl-L-cysteine protects SHSYSY neuroblastoma cells from oxidative stress and cell cytotoxicity: effects on b-amyloid secretion and tau phosphorylation. J Neurochem 2001, 76, (1), 224-233. 70. Misonou, H.; Morishima-Kawashima, M.; Ihara, Y., Oxidative Stress Induces Intracellular Accumulation of Amyloid b-Protein (A.b) in Human Neuroblastoma Cells. Biochemistry 2000, 39, (23), 6951-6959. 71. Lovell, M. A.; Xiong, S.; Xie, G; Davies, P.; Markesbery, W. R., Induction of hyperphosphorylated tan in primary rat cortical neuron cultures mediated by oxidative stress and glycogen synthase kinase-3. J Alzheimers Dis 2004, 6, (6), 659-671. 72. Nakashima, H.; Ishihara, T.; Yokota, 0.; Terada, S.; Trojanowski, J. Q.; Lee, V. M. Y.; Kuroda, S., Effects of a-tocopherol on an animal model of tauopathies. Free Radic Biol Med 2004, 37, (2), 176-186. 73. Montine, T. J.; Neely, M. D.; Quinn, J. F .; Beal, M. F.; Markesbery, W. R.; Roberts, L. J .; Morrow, J. D., Lipid peroxidation in aging brain and Alzheimer's disease. Free Radic Biol Med 2002, 33, (5), 620-626. 74. Mattson, M. P.; Fu, W.; Waeg, G.; Uchida, K., 4-Hydroxynonenal, a product of lipid peroxidation, inhibits dephosphorylation of the microtubule-associated protein tau. NeuroReport 1997, 8, (9-10), 2275-2281. 75. Gomez-Ramos, A.; Diaz-Nido, J.; Smith Mark, A.; Perry, G.; Avila, J ., Effect of the lipid peroxidation product acrolein on tau phosphorylation in neural cells. J Neurosci Res 2003, 71, (6), 863-70. 76. Butterfield, D. A.; Boyd-Kimball, D., Amyloid beta-peptide(l-42) contributes to the oxidative stress and neurodegeneration found in Alzheimer disease brain. Brain Pathol 2004, 14, (4), 426-32. 77. Hensley, K.; Carney, J. M.; Mattson, M. P.; Aksenova, M.; Harris, M.; Wu, J. F.; Floyd, R. A.; Butterfield, D. A., A model for beta-amyloid aggregation and neurotoxicity 107 based on free radical generation by the peptide: relevance to Alzheimer disease. Proc NatlAcad Sci USA 1994, 91, (8), 3270-4. 78. Sayre, L. M.; Zagorski, M. G.; Surewicz, W. K.; Kraffi, G. A.; Perry, G., Mechanisms of neurotoxicity associated with amyloid beta deposition and the role of free radicals in the pathogenesis of Alzheimer's disease: a critical appraisal. Chem Res Toxicol 1997, 10, (5), 518-26. 79. Schliebs, R.; Arendt, T., The significance of the cholinergic system in the brain during aging and in Alzheimer's disease. J Neural T ransm 2006, 113, (l 1), 1625-44. 80. Bartus, R. T., On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp Neurol 2000, 163, (2), 495-529. 81. Bierer, L. M.; Haroutunian, V.; Gabriel, S.; Knott, P. J.; Carlin, L. S.; Purohit, D. P.; Perl, D. P.; Schmeidler, J.; Kanof, P.; Davis, K. L., Neurochemical correlates of dementia severity in Alzheimer‘s disease: relative importance of the cholinergic deficits. J Neurochem 1995, 64, (2), 749-60. 82. Sofic, E.; Gotz, M.; Frolich, L.; Burger, R.; Heckers, S.; Riederer, P.; Jellinger, K.; Beckmann, H., Reflection of changes in membrane constituents in various regions of Alzheimer brains to differential scanning therrnograrns. J Neural Transm Suppl 1990, 32, 259-67. 83. Zuchner, T.; Perez-Polo, J. R.; Schliebs, R., Beta-secretase BACEl is differentially controlled through muscarinic acetylcholine receptor signaling. J Neurosci Res 2004, 77, (2), 250-7. 84. Rossner, S.; Ueberham, U.; Schliebs, R.; Perez-Polo, J. R.; Bigl, V., The regulation of amyloid precursor protein metabolism by cholinergic mechanisms and neurotrophin receptor signaling. Prog Neurobiol 1998, 56, (5), 541-69. 85. Lin, L.; Georgievska, B.; Mattsson, A.; Isacson, 0., Cognitive changes and modified processing of amyloid precursor protein in the cortical and hippocampal system after cholinergic synapse loss and muscarinic receptor activation. Proc Natl A cad Sci U S A 1999, 96, (21), 12108-13. 86. Efihimiopoulos, S.; Vassilacopoulou, D.; Ripellino, J. A.; Tezapsidis, N.; Robakis, N. K., Cholinergic agonists stimulate secretion of soluble full-length amyloid 108 precursor protein in neuroendocrine cells. Proc Natl Acad Sci U S A 1996, 93, (15), 8046- 50. 87. Sec, J.; Kim, S.; Kim, H.; Park, C. H.; Jeong, S.; Lee, J.; Choi, S. H.; Chang, K.; Rah, J .; Koo, J .; Kim, E.; Suh, Y., Effects of nicotine on APP secretion and Abeta- or CT(105)-induced toxicity. Biol Psychiatry 2001, 49, (3), 240-7. 88. Utsuki, T.; Shoaib, M.; Holloway, H. W.; Ingram, D. K.; Wallace, W. C.; Haroutunian, V.; Sambamurti, K.; Lahiri, D. K.; Greig, N. H., Nicotine lowers the secretion of the Alzheimer's amyloid beta-protein precursor that contains amyloid beta- peptide in rat. J Alzheimers Dis 2002, 4, (5), 405-15. 89. Hellstrom-Lindahl, E.; Moore, H.; Nordberg, A., Increased levels of tau protein in SH-SY5Y cells after treatment with cholinesterase inhibitors and nicotinic agonists. J Neurochem 2000, 74, (2), 777-84. 90. Wang, H. Y.; Li, W.; Benedetti, N. J.; Lee, D. H., Alpha 7 nicotinic acetylcholine receptors mediate beta-amyloid peptide-induced tau protein phosphorylation. J Biol Chem 2003, 278, (34), 31547-53. 91. Seshadri, S.; Beiser, A.; Selhub, J.; Jacques, P. F.; Rosenberg, I. H.; D'Agostino, R. B.; Wilson, P. W.; Wolf, P. A., Plasma homocysteine as a risk factor for dementia and Alzheimer's disease. N Engl J Med 2002, 346, (7), 476-83. 92. Joosten, E.; Lesaffre, E.; Riezler, R.; Ghekiere, V.; Dereymaeker, L.; Pelemans, W.; Dejaeger, E., Is metabolic evidence for vitamin B-12 and folate deficiency more frequent in elderly patients with Alzheimer's disease? J Gerontol A Biol Sci Med Sci 1997, 52, (2), M76-9. 93. Miller, J. W.; Green, R; Mungas, D. M.; Reed, B. R.; Jagust, W. J., Homocysteine, vitamin B6, and vascular disease in AD patients. Neurology 2002, 58, (10), 1471-5. 94. Nilsson, K.; Gustafson, L.; Hultberg, 3., Plasma homocysteine is a sensitive marker for tissue deficiency of both cobalamines and folates in a psychogeriatric population. Dement Geriatr Cogn Disord 1999, 10, (6), 476-82. 95. Quadri, P.; Fragiacomo, C.; Pezzati, R.; Zanda, E.; Tettamanti, M.; Lucca, U., Homocysteine and B vitamins in mild cognitive impairment and dementia. Clin Chem Lab Med 2005, 43, (10), 1096—100. 109 " .“a'r-l 96. Ravaglia, G.; Forti, P.; Maioli, F .; Martelli, M.; Servadei, L.; Brunetti, N.; Porcellini, E.; Licastro, F., Homocysteine and folate as risk factors for dementia and Alzheimer disease. Am J Clin Nutr 2005, 82, (3), 636-43. 97. Teunissen, C. E.; van Boxtel, M. P.; Jolles, J.; de Vente, J.; Vreeling, F.; Verhey, F .; Polrnan, C. H.; Dijkstra, C. D.; Blom, H. J ., Homocysteine in relation to cognitive performance in pathological and non-pathological conditions. Clin Chem Lab Med 2005, 43, (10), 1089-95. 98. Tucker, K. L.; Qiao, N.; Scott, T.; Rosenberg, 1.; Spiro, A., 3rd, High homocysteine and low B vitamins predict cognitive decline in aging men: the Veterans Affairs Normative Aging Study. Am J Clin Nutr 2005, 82, (3), 627-35. 99. Ho, P. 1.; Ortiz, D.; Rogers, E.; Shea, T. 3., Multiple aspects of homocysteine neurotoxicity: glutamate excitotoxicity, kinase hyperactivation and DNA damage. J Neurosci Res 2002, 70, (5), 694-702. 100. Kruman, II; Culrnsee, C.; Chan, S. L.; Kruman, Y.; Guo, Z.; Penix, L.; Mattson, M. P., Homocysteine elicits a DNA damage response in neurons that promotes apoptosis and hypersensitivity to excitotoxicity. J Neurosci 2000, 20, (l 8), 6920-6. 101. Selley, M. L.; Close, D. R.; Stern, S. E., The effect of increased concentrations of homocysteine on the concentration of (E)-4-hydroxy-2-nonenal in the plasma and cerebrospinal fluid of patients with Alzheimer's disease. Neurobiol Aging 2002, 23, (3), 383-8. 102. Fuso, A.; Seminara, L.; Cavallaro, R. A.; D'Anselrni, F.; Scarpa, S., S- adenosylmethionine/homocysteine cycle alterations modify DNA methylation status with consequent deregulation of PS] and BACE and beta-amyloid production. Mol Cell Neurosci 2005, 28, (1), 195-204. 103. Flicker, L.; Martins, R N.; Thomas, J.; Acres, J.; Taddei, K.; Norman, P.; Jamrozik, K.; Almeida, O. P., Homocysteine, Alzheimer genes and proteins, and measures of cognition and depression in older men. J Alzheimers Dis 2004, 6, (3), 329- 36. 104. Mizrahi, E. H.; Jacobsen, D. W.; Debanne, S. M.; Traore, F.; Lerner, A. J.; Friedland, R P.; Petot, G. J ., Plasma total homocysteine levels, dietary vitamin B6 and folate intake in AD and healthy aging. J Nutr Health Aging 2003, 7, (3), 160-5. 110 105. Luchsinger, J. A.; Tang, M. X.; Shea, S.; Miller, J.; Green, R; Mayeux, R, Plasma homocysteine levels and risk of Alzheimer disease. Neurology 2004, 62, (l 1), 1972-6. 106. White, A. R.; Huang, X.; Jobling, M. P.; Barrow, C. J.; Beyreuther, K.; Masters, C. L.; Bush, A. 1.; Cappai, R, Homocysteine potentiates copper- and amyloid beta peptide-mediated toxicity in primary neuronal cultures: possible risk factors in the Alzheimer's-type neurodegenerative pathways. J Neurochem 2001, 76, (5), 1509-20. 107. Cilley, R. E.; Brighton, V. K., The significance of Helicobacter pylori colonization of the stomach. Semin Pediatr Surg 1995, 4, (4), 221-7. 108. Lowy, D. R.; Schiller, J. T., Prophylactic human papillomavirus vaccines. J Clin Invest 2006, 116, (5), 1167-73. 109. Campbell, L. A.; Kuo, C. C., Chlamydia pneumoniae and atherosclerosis. Semin Respir Infect 2003, 18, (1), 48-54. 110. de Boer, O. J.; van der Wal, A. C.; Becker, A. E., Atherosclerosis, inflammation, and infection. J Pathol 2000, 190, (3), 237-43. 111. Deatly, A. M.; Haase, A. T.; Fewster, P. H.; Lewis, E.; Ball, M. J., Human herpes virus infections and Alzheimer's disease. Neuropathol Appl Neurobiol 1990, 16, (3), 213- 23. 112. Dobson, C. B.; Itzhaki, R P., Herpes simplex virus type 1 and Alzheimer's disease. Neurobiol Aging 1999, 20, (4), 457-65. 113. Fraser, N. W.; Lawrence, W. C.; Wroblewska, Z.; Gilden, D. H.; Koprowski, H., Herpes simplex type 1 DNA in human brain tissue. Proc Natl Acad Sci U S A 1981, 78, (10), 6461-5. 114. Libikova, H.; Pogady, J .; Wiedermann, V.; Breier, S., Search for herpetic antibodies in the cerebrospinal fluid in senile dementia and mental retardation. Acta Virol 1975, 19, (6), 493-5. 115. Sequiera, L. W.; Jennings, L. C.; Carrasco, L. H.; Lord, M. A.; Curry, A.; Sutton, R. N., Detection of herpes-simplex viral genome in brain tissue. Lancet 1979, 2, (8143), 609-12. 111 116. Middleton, P. J .; Petric, M.; Kozak, M.; Rewcastle, N. B.; McLachlan, D. R., Herpes-simplex viral genome and senile and presenile dementias of Alzheimer and Pick. Lancet 1980, l, (8176), 1038. 117. Pogo, B. G.; Casals, J.; Elizan, T. S., A study of viral genomes and antigens in brains of patients with Alzheimer's disease. Brain 1987, 110 (Pt 4), 907-15. 118. Renvoize, E. B.; Awad, I. 0.; Hambling, M. H., A sero-epidemiological study of conventional infectious agents in Alzheimer's disease. Age Ageing 1987, 16, (5), 311-4. 119. Taylor, G. R.; Crow, T. J.; Markakis, D. A.; Lofthouse, R.; Neeley, S.; Carter, G. 1., Herpes simplex virus and Alzheimer's disease: a search for virus DNA by spot hybridisation. J Neurol Neurosurg Psychiatry 1984, 47, (10), 1061-5. 120. Little, C. S.; Hammond, C. J .; MacIntyre, A.; Balin, B. J.; Appelt, D. M., Chlamydia pneumoniae induces Alzheimer-like amyloid plaques in brains of BALB/c mice. Neurobiol Aging 2004, 25, (4), 419-29. 121. Grant, W. 3., Dietary links to Alzheimer's disease: 1999 update. J Alzheimers Dis 1999, 1, (4,5), 197-201. 122. Levin-Allerhand, J. A.; Lominska, C. E.; Smith, J. D., Increased amyloid- levels in APPSWE transgenic mice treated chronically with a physiological high-fat high- cholesterol diet. J Nutr Health Aging 2002, 6, (5), 315-9. 123. Shie, F .-S.; LeBouef, R. C.; Jin, L.-W., Early intraneuronal Ab deposition in the hippocampus of APP transgenic mice. NeuroReport 2003, 14, (1), 123-129. 124. Oksman, M.; Iivonen, H.; Hogyes, E.; Amtul, Z.; Penke, B.; Leenders, 1.; Broersen, L.; Luetjohann, D.; Hartrnann, T.; Tanila, H., Impact of different saturated fatty acid, polyunsaturated fatty acid and cholesterol containing diets on beta-amyloid accumulation in APP/P81 transgenic mice. Neurobiol Dis 2006, 23, (3), 563-572. 125. Wang, S.-W.; Wang, M.; Grossman, B. M.; Martin, R J., Effects of dietary fat on food intake and brain uptake and oxidation of fatty acids. Physio! Behavior 1994, 56, (3), 517-22. 126. Dhopeshwarkar, G. A.; Mead, J. F ., Uptake and transport of fatty acids into the brain and role of the blood-brain barrier system. Adv Lipid Res 1973, 11, 109-42. 112 127. Arvanitakis, Z.; Wilson Robert, S.; Bienias Julia, L.; Evans Denis, A.; Bennett David, A., Diabetes mellitus and risk of Alzheimer disease and decline in cognitive function. Arch Neurol 2004, 61, (5), 661-6. 128. Monti, L. D.; Landoni, C.; Setola, E.; Galluccio, E.; Lucotti, P.; Sandoli, E. P.; Origgi, A.; Lucignani, G.; Piatti, P.; Fazio, F ., Myocardial insulin resistance associated with chronic hypertriglyceridemia and increased FFA levels in Type 2 diabetic patients. Am J Physiol 2004, 287, (3, Pt. 2), H1225-H123 l. 129. Smith, Q. R.; Nagura, H., Fatty acid uptake and incorporation in brain: studies with the perfusion model. J Mol Neurosci 2001, 16, (2/3), 167-172. 130. Guo, Z.; Cupples, L. A.; Kurz, A.; Auerbach, S. H.; Volicer, L.; Chui, H.; Green, R. C.; Sadovnick, A. D.; Duara, R.; DeCarli, C.; Johnson, K.; Go, R. C.; Growdon, J. H.; Haines, J. L.; Kukull, W. A.; Farrer, L. A., Head injury and the risk of AD in the MIRAGE study. Neurology 2000, 54, (6), 1316-23. 131. Homayoun, P.; De Turco, E. B. R.; Parkins, N. E.; Lane, D. C.; Soblosky, J.; Carey, M. E.; Bazan, N. G., Delayed phospholipid degradation in rat brain afier traumatic brain injury. J Neurochem 1997, 69, (1), 199-205. 132. Lipton, P., Ischemic cell death in brain neurons. Physiol Rev 1999, 79, (4), 143 l- 568. 133. Goux, W. J .; Rodriguez, S.; Sparkman, D. R., Analysis of the core components of Alzheimer paired helical filaments. A gas chromatography/mass spectrometry characterization of fatty acids, carbohydrates and long-chain bases. FEBS Letters 1995, 366, (1), 81-5. 134. Roher, A. E.; Weiss, N.; Kokjohn, T. A.; Kuo, Y.-M.; Kalback, W.; Anthony, J.; Watson, D.; Luehrs, D. C.; Sue, L.; Walker, D.; Emmerling, M.; Goux, W.; Beach, T., Increased Ab Peptides and Reduced Cholesterol and Myelin Proteins Characterize White Matter Degeneration in Alzheimer's Disease. Biochemistry 2002, 41, (37), 11080-11090. 135. Petot, G. J.; Traore, F.; Debanne, S. M.; Lerner, A. J.; Smyth, K. A.; Friedland, R. P., Interactions of apolipoprotein E genotype and dietary fat intake of healthy older persons during mid-adult life. Metabolism Clin Exp 2003, 52, (3), 279-281. 136. Braak, H.; Braak, E., Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 1991, 82, (4), 239-59. 113 137. Arriagada, P. V.; Growdon, J. H.; Hedley-Whyte, E. T.; Hyman, B. T., Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease. Neurology 1992, 42, (3 Pt 1), 631-9. 138. Bierer, L. M.; Hof, P. R.; Purohit, D. P.; Carlin, L.; Schmeidler, J.; Davis, K. L.; Perl, D. P., Neocortical neuroftbrillary tangles correlate with dementia severity in Alzheimer's disease. Arch Neurol 1995, 52, (1), 81-8. 139. Lee, M. S.; Kwon, Y. T.; Li, M.; Peng, J.; Friedlander, R M.; Tsai, L. H., Neurotoxicity induces cleavage of p35 to p25 by calpain. Nature 2000, 405, (6784), 360- 4. 140. Hwang, S. C.; Jhon, D. Y.; Bae, Y. S.; Kim, J. H.; Rhee, S. 6., Activation of phospholipase C-gamma by the concerted action of tau proteins and arachidonic acid J Biol Chem 1996, 271, (31), 18342-9. 141. Flanagan, L. A.; Cunningham, C. C.; Chen, J.; Prestwich, G. D.; Kosik, K. S.; Janmey, P. A., The structure of divalent cation-induced aggregates of PIP2 and their alteration by gelsolin and tau. Biophys J 1997, 73, (3), 1440-7. 142. Reszka, A. A.; Seger, R.; Diltz, C. D.; Krebs, E. G.; Fischer, E. H., Association of mitogen-activated protein kinase with the microtubule cytoskeleton. Proc Natl Acad Sci USA 1995, 92, (19), 8881-5. 143. Liao, H.; Li, Y.; Brautigan, D. L.; Gundersen, G. 6., Protein phosphatase 1 is targeted to microtubules by the microtubule-associated protein Tau. J Biol Chem 1998, 273, (34), 21901-8. 144. Cunningham, C. C.; Leclerc, N.; Flanagan, L. A.; Lu, M.; Janmey, P. A.; Kosik, K. S., Microtubule-associated protein 2c reorganizes both microtubules and microfilaments into distinct cytological structures in an actin-binding protein-280- deficient melanoma cell line. J Cell Biol 1997, 136, (4), 845-57. 145. Billingsley, M. L.; Kincaid, R. L., Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. Biochem J 1997, 323 (Pt 3), 577-91. 146. Morita-Fujimura, Y.; Kurachi, M.; Tashiro, H.; Komiya, Y.; Tashiro, T., Reduced microtubule-nucleation activity of tau after dephosphorylation. Biochem Biophys Res Commun 1996, 225, (2), 462-8. 114 147. Geschwind, D. H., Tau phosphorylation, tangles, and neurodegeneration: the chicken or the egg? Neuron 2003, 40, (3), 457-60. 148. Lee, H. G.; Perry, G.; Moreira, P. 1.; Garrett, M. R.; Liu, Q.; Zhu, X.; Takeda, A.; Nunomura, A.; Smith, M. A., Tau phosphorylation in Alzheimer's disease: pathogen or protector? Trends Mol Med 2005, 11, (4), 164-9. 149. Grundke-Iqbal, 1.; Iqbal, K.; Tung, Y. C.; Quinlan, M.; Wisniewski, H. M.; Binder, L. 1., Abnormal phosphorylation of the microtubule-associated protein tau (tan) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci U S A 1986, 83, (13), 4913-7. 150. Bancher, C.; Brunner, C.; Lassmann, H.; Budka, H.; Jellinger, K.; Wiche, G.; Seitelberger, F .; Grundke-Iqbal, 1.; Iqbal, K.; Wisniewski, H. M., Accumulation of abnormally phosphorylated tau precedes the formation of neurofibrillary tangles in Alzheimer's disease. Brain Res 1989, 477, (1-2), 90-9. 151. Braak, E.; Braak, H.; Mandelkow, E. M., A sequence of cytoskeleton changes related to the formation of neurofibrillary tangles and neuropil threads. Acta Neuropathol 1994, 87, (6), 554-67. 152. Alonso, A.; Zaidi, T.; Novak, M.; Grundke-Iqbal, I.; Iqbal, K, Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments. Proc Natl Acad Sci U S A 2001, 98, (12), 6923-8. 153. Perez, M.; Cuadros, R.; Smith, M. A.; Perry, G.; Avila, J., Phosphorylated, but not native, tau protein assembles following reaction with the lipid peroxidation product, 4— hydroxy-Z-nonenal. F EBS Lett 2000, 486, (3), 270-4. 154. Morishima-Kawashima, M.; Hasegawa, M.; Takio, K.; Suzuki, M.; Yoshida, H.; Titani, K.; Ihara, Y., Proline-directed and non-proline-directed phosphorylation of PHF- tau. J Biol Chem 1995, 270, (2), 823-9. 155. Gong, C. X.; Grundke-Iqbal, 1.; Damuni, Z.; Iqbal, K., Dephosphorylation of microtubule-associated protein tau by protein phosphatase-l and -2C and its implication in Alzheimer disease. FEBS Lett 1994, 341, (1), 94-8. 156. Gong, C. X.; Singh, T. J.; Grundke-Iqbal, I.; Iqbal, K., Alzheimer's disease abnormally phosphorylated tau is dephosphorylated by protein phosphatase-23 (calcineurin). J Neurochem 1994, 62, (2), 803-6. 115 157. Gong, C. X.; Lidsky, T.; Wegiel, J.; Zuck, L.; Grundke-Iqbal, I.; Iqbal, K, Phosphorylation of microtubule-associated protein tau is regulated by protein phosphatase 2A in mammalian brain. Implications for neurofibrillary degeneration in Alzheimer's disease. J Biol Chem 2000, 275, (8), 5535-44. 158. Gong, C. X.; Singh, T. J.; Grundke-Iqbal, 1.; Iqbal, K., Phosphoprotein phosphatase activities in Alzheimer disease brain. J Neurochem 1993, 61, (3), 921 -7. 159. Gomez-Ramos, A.; Diaz-Nido, J.; Smith, M. A.; Perry, G.; Avila, J., Effect of the lipid peroxidation product acrolein on tau phosphorylation in neural cells. J Neurosci Res 2003, 71, (6), 863-70. 160. Lovell, M. A.; Xiong, S.; Xie, G.; Davies, P.; Markesbery, W. R., Induction of hyperphosphorylated tan in primary rat cortical neuron cultures mediated by oxidative stress and glycogen synthase kinase-3. J Alzheimers Dis 2004, 6, (6), 659-71; discussion 673-81. 161. Nakashima, H.; Ishihara, T.; Yokota, 0.; Terada, S.; Trojanowski, J. Q.; Lee, V. M.; Kuroda, S., Effects of alpha-tocopherol on an animal model of tauopathies. Free Radic Biol Med 2004, 37, (2), 176-86. 162. Zhu, X.; Rottkamp, C. A.; Boux, H.; Takeda, A.; Perry, G.; Smith, M. A., Activation of p38 kinase links tau phosphorylation, oxidative stress, and cell cycle- related events in Alzheimer disease. J Neuropathol Exp Neurol 2000, 59, (10), 880-8. 163. Butterfield, D. A.; Drake, J.; Pocernich, C.; Castegna, A., Evidence of oxidative damage in Alzheimer's disease brain: central role for amyloid beta-peptide. Trends Mol Med 2001, 7, (12), 548-54. 164. Butterfield, D. A.; Lauderback, C. M., Lipid peroxidation and protein oxidation in Alzheimer's disease brain: potential causes and consequences involving amyloid beta- peptide-associated free radical oxidative stress. Free Radic Biol Med 2002, 32, (11), 1050-60. 165. Nunomura, A.; Perry, G.; Aliev, G.; Hirai, K.; Takeda, A.; Balraj, E. K.; Jones, P. K.; Ghanbari, H.; Wataya, T.; Shimohama, S.; Chiba, S.; Atwood, C. S.; Petersen, R. B.; Smith, M. A., Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol 2001, 60, (8), 759-67. 116 166. Lovell, M. A.; Ehmann, W. D.; Butler, S. M.; Markesbery, W. R., Elevated thiobarbituric acid-reactive substances and antioxidant enzyme activity in the brain in Alzheimer's disease. Neurology 1995, 45, (8), 1594-601. 167. Marcus, D. L.; Thomas, C.; Rodriguez, C.; Simberkoff, K.; Tsai, J. S.; Strafaci, J. A.; Freedman, M. L., Increased peroxidation and reduced antioxidant enzyme activity in Alzheimer's disease. Exp Neurol 1998, 150, (1), 40-4. 168. Mark, R. J.; Lovell, M. A.; Markesbery, W. R.; Uchida, K.; Mattson, M. P., A role for 4-hydroxynonenal, an aldehydic product of lipid peroxidation, in disruption of ion homeostasis and neuronal death induced by amyloid beta-peptide. J Neurochem 1997, 68, (1), 255-64. 169. Lovell, M. A.; Xie, G; Markesbery, W. R., Acrolein is increased in Alzheimer's disease brain and is toxic to primary hippocampal cultures. Neurobiol Aging 2001, 22, (2), 187-94. 170. Berlett, B. S.; Stadtrnan, E. R., Protein oxidation in aging, disease, and oxidative stress. J Biol Chem 1997, 272, (33), 20313-6. 171. Smith, C. D.; Carney, J. M.; Starke-Reed, P. E.; Oliver, C. N.; Stadtrnan, E. R.; Floyd, R. A.; Markesbery, W. R., Excess brain protein oxidation and enzyme dysfunction in normal aging and in Alzheimer disease. Proc Natl Acad Sci U S A 1991, 88, (23), 10540-3. 172. Lyras, L.; Cairns, N. J .; Jenner, A.; Jenner, P.; Halliwell, B., An assessment of oxidative damage to proteins, lipids, and DNA in brain from patients with Alzheimer's disease. J Neurochem 1997, 68, (5), 2061-9. 173. Wille, H.; Drewes, G.; Biernat, J.; Mandelkow, E. M.; Mandelkow, E., Alzheimer—like paired helical filaments and antiparallel dimers formed from microtubule- associated protein tau in vitro. J Cell Biol 1992, 118, (3), 573-84. 174. Schweers, 0.; Mandelkow, E. M.; Biemat, J.; Mandelkow, E., Oxidation of cysteine-322 in the repeat domain of microtubule-associated protein tau controls the in vitro assembly of paired helical filaments. Proc Natl Acad Sci U S A 1995, 92, (18), 8463-7. 175. Smith, M. A.; Perry, G.; Richey, P. L.; Sayre, L. M.; Anderson, V. E.; Beal, M. F.; Kowall, N., Oxidative damage in Alzheimer's. Nature 1996, 382, (6587), 120-1. 117 176. Chandler, L. J.; Newsom, H.; Sumners, C.; Crews, F., Chronic ethanol exposure potentiates NMDA excitotoxicity in cerebral cortical neurons. J Neurochem 1993, 60, (4), 1578-81. 177. Pshenichkin, S. P.; Wise, B. C., Okadaic acid increases nerve growth factor secretion, mRN A stability, and gene transcription in primary cultures of cortical astrocytes. J Biol Chem 1995, 270, (11), 5994—9. 178. Williamson, R.; Scales, T.; Clark, B. R.; Gibb, G.; Reynolds, C. H.; Kellie, S.; Bird, L. N.; Varndell, L. M.; Sheppard, P. W.; Everall, 1.; Anderton, B. H., Rapid tyrosine phosphorylation of neuronal proteins including tau and focal adhesion kinase in response to amyloid-b peptide exposure: involvement of src family protein kinases. J Neurosci 2002, 22, (1), 10-20. 179. Sauer, H.; Klimm, B.; Hescheler, J.; Wartenberg, M., Activation of p90RSK and growth stimulation of multicellular tumor spheroids are dependent on reactive oxygen species generated after purinergic receptor stimulation by ATP. FASEB J 2001, 15, (13), 2539-2541, 10 1096/fj 01-0360ije. 180. Kosik, K. S.; Crandall, J. E.; Mufson, E. J.; Neve, R. L., Tau in situ hybridization in normal and Alzheimer brain: localization in the somatodendritic compartment. Ann Neurol 1989, 26, (3), 352-61. 181. Papasozomenos, S. C., Tau protein immunoreactivity in dementia of the Alzheimer type: 11. Electron microscopy and pathogenetic implications. Effects of fixation on the morphology of the Alzheimer's abnormal filaments. Lab Invest 1989, 60, (3), 375-89. 182. Blazquez, C.; Galve-Roperh, 1.; Guzman, M., De novo-synthesized ceramide signals apoptosis in astrocytes via extracellular signal-regulated kinase. FASEB J 2000, 14, (14), 2315-2322. 183. Gorina, R.; Petegnief, V.; Chamorro, A.; Planas, A. M., AG490 prevents cell death after exposure of rat astrocytes to hydrogen peroxide or proinflammatory cytokines: involvement of the Jak2/STAT pathway. J Neurochem 2005, 92, (3), 505-518. 184. Goedert, M.; Jakes, R.; Vanmechelen, E., Monoclonal antibody AT8 recognizes tau protein phosphorylated at both serine 202 and threonine 205. Neurosci Lett 1995, 189, (3), 167-70. 118 185. Otvos, L., Jr.; Feiner, L.; Lang, E.; Szendrei, G. 1.; Goedert, M.; Lee, V. M. Y., Monoclonal antibody PHF -1 recognizes Tau protein phosphorylated at serine residues 396 and 404. J Neurosci Res 1994, 39, (6), 669-73. 186. Gong, C. X.; Singh, T. J.; Grundke-Iqbal, 1.; Iqbal, K., Alzheimer's disease abnormally phosphorylated t is dephosphorylated by protein phosphatase-2B (calcineurin). J Neurochem 1994, 62, (2), 803-6. 187. Davis, D. R.; Anderton, B. H.; Brion, J .-P.; Reynolds, C. H.; Hanger, D. P., Oxidative stress induces dephosphorylation of t in rat brain primary neuronal cultures. J Neurochem 1997, 68, (4), 1590-1597. 188. Glenner, G. G.; Wong, C. W., Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem Biophys Res Commun 1984, 120, (3), 885-90. 189. Price, D. L.; Sisodia, S. S., Mutant genes in familial Alzheimer's disease and transgenic models. Annu Rev Neurosci 1998, 21, 479-505. 190. Hardy, J .; Selkoe, D. J., The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 2002, 297, (5580), 353-6. 191. St George-Hyslop, P. H.; Petit, A., Molecular biology and genetics of Alzheimer's disease. C R Biol 2005, 328, (2), 119-30. 192. Tanzi, R. E.; Bertram, L., Twenty years of the Alzheimer's disease amyloid hypothesis: a genetic perspective. Cell 2005, 120, (4), 545-55. 193. Desai, P. P.; Ikonomovic, M. D.; Abrahamson, E. E.; Hamilton, R. L.; Isanski, B. A.; Hope, C. E.; Klunk, W. E.; DeKosky, S. T.; Kamboh, M. 1., Apolipoprotein D is a component of compact but not diffuse amyloid-beta plaques in Alzheimer's disease temporal cortex. Neurobiol Dis 2005, 20, (2), 574-82. 194. Frey, H. J.; Mattila, K. M.; Korolainen, M. A.; Pirttila, T., Problems associated with biological markers of Alzheimer's disease. Neurochem Res 2005, 30, (12), 1501-10. 195. Selkoe, D. J., Translating cell biology into therapeutic advances in Alzheimer's disease. Nature 1999, 399, (6738 Suppl), A23-31. 119 196. Klein, W. L., Abeta toxicity in Alzheimer's disease: globular oligomers (ADDLs) as new vaccine and drug targets. Neurochem Int 2002, 41, (5), 345-52. 197. Golde, T. E., Alzheimer disease therapy: can the amyloid cascade be halted? J Clin Invest 2003, 111, (1), 11-8. 198. Pereira, C.; Agostinho, P.; Moreira, P. 1.; Cardoso, S. M.; Oliveira, C. R, Alzheimer's disease-associated neurotoxic mechanisms and neuroprotective strategies. Curr Drug Targets CNS Neural Disord 2005, 4, (4), 383-403. 199. De Strooper, B.; Annaert, W., Proteolytic processing and cell biological functions of the amyloid precursor protein. J Cell Sci 2000, 113 (Pt 11), 1857-70. 200. Nunan, J .; Small, D. H., Regulation of APP cleavage by alpha-, beta- and gamma- , secretases. FEBS Lett 2000, 483, (1), 6-10. 201. LeBlanc, A. C.; Chen, H. Y.; Autilio-Gambetti, L.; Gambetti, P., Differential APP gene expression in rat cerebral cortex, meninges, and primary astroglial, microglial and neuronal cultures. FEBS Lett 1991, 292, (1-2), 171-8. 202. Beyreuther, K.; Masters, C. L, Alzheimer's disease. The ins and outs of amyloid- beta. Nature 1997, 389, (6652), 677-8. 203. Allinson, T. M.; Parkin, E. T.; Turner, A. J.; Hooper, N. M., ADAMS family members as amyloid precursor protein alpha-secretases. J Neurosci Res 2003, 74, (3), 342-52. 204. Sinha, S.; Lieberburg, 1., Cellular mechanisms of beta-amyloid production and secretion. Proc Natl A cad Sci U S A 1999, 96, (20), 11049-53. 205 . Kojro, E.; Gimpl, G.; Lammich, S.; Marz, W.; Fahrenholz, F ., Low cholesterol stimulates the nonamyloidogenic pathway by its effect on the alpha -secretase ADAM 10. Proc Natl Acad Sci USA 2001, 98, (10), 5815-20. 206. Ehehalt, R.; Keller, P.; Haass, C.; Thiele, C.; Simons, K., Amyloidogenic processing of the Alzheimer beta-amyloid precursor protein depends on lipid rafts. J Cell Biol 2003, 160, (1), 113-23. 120 I fill-.1 207. Weidemann, A.; Konig, G.; Bunke, D.; Fischer, P.; Salbaum, J. M.; Masters, C. L.; Beyreuther, K., Identification, biogenesis, and localization of precursors of Alzheimer's disease A4 amyloid protein. Cell 1989, 57, (1), 115-26. 208. Hussain, 1.; Powell, D.; Howlett, D. R; Tew, D. G.; Meek, T. D.; Chapman, C.; Gloger, I. S.; Murphy, K. E.; Southan, C. D.; Ryan, D. M.; Smith, T. S.; Simmons, D. L.; Walsh, F. S.; Dingwall, C.; Christie, G., Identification of a novel aspartic protease (Asp 2) as beta-secretase. Mol Cell Neurosci 1999, 14, (6), 419-27. 209. Vassar, R.; Bennett, B. D.; Babu-Khan, S.; Kahn, S.; Mendiaz, E. A.; Denis, P.; Teplow, D. B.; Ross, S.; Amarante, P.; Loeloff, R.; Luo, Y.; Fisher, S.; Fuller, J.; Edenson, S.; Lile, J .; Jarosinski, M. A.; Biere, A. L.; Curran, E.; Burgess, T.; Louis, J. C.; Collins, F .; Treanor, J .; Rogers, G.; Citron, M., Beta-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. Science 1999, 286, (5440), 735-41. 210. Yan, R.; Bienkowski, M. J.; Shuck, M. E.; Miao, H.; Tory, M. C.; Pauley, A. M.; Brashier, J. R.; Stratman, N. C.; Mathews, W. R.; Buhl, A. E.; Carter, D. B.; Tomasselli, A. G.; Parodi, L. A.; Heinrikson, R. L.; Gurney, M. E., Membrane-anchored aspartyl protease with Alzheimer's disease beta-secretase activity. Nature 1999, 402, (6761), 533- 7. 211. Cai, H.; Wang, Y.; McCarthy, D.; Wen, H.; Borchelt, D. R.; Price, D. L.; Wong, P. C., BACEl is the major beta-secretase for generation of Abeta peptides by neurons. Nat Neurosci 2001, 4, (3), 233-4. 212. K00, E. H.; Squazzo, S. L., Evidence that production and release of amyloid beta- protein involves the endocytic pathway. J Biol Chem 1994, 269, (26), 17386-9. 213. Haass, C.; Lemere, C. A.; Capell, A.; Citron, M.; Seubert, P.; Schenk, D.; Lannfelt, L.; Selkoe, D. J ., The Swedish mutation causes early-onset Alzheimer's disease by beta-secretase cleavage within the secretory pathway. Nat Med 1995, l, (12), 1291-6. 214. Walter, J .; Fluhrer, R.; Hartung, B.; Willem, M.; Kaether, C.; Capell, A.; Lammich, S.; Multhaup, G.; Haass, C., Phosphorylation regulates intracellular trafficking of beta-secretase. J Biol Chem 2001, 276, (18), 14634-41. 215. Haass, C.; Hung, A. Y.; Schlossmacher, M. G.; Teplow, D. B.; Selkoe, D. J., beta- Amyloid peptide and a 3-kDa fragment are derived by distinct cellular mechanisms. J Biol Chem 1993, 268, (5), 3021-4. 121 216. Cordy, J. M.; Hussain, 1.; Dingwall, C.; Hooper, N. M.; Turner, A. J., Exclusively targeting beta-secretase to lipid rafts by GPI-anchor addition up-regulates beta-site processing of the amyloid precursor protein. Proc Natl Acad Sci U S A 2003, 100, (20), 11735-40. 217. Abad-Rodriguez, J.; Ledesma, M. D.; Craessaerts, K.; Perga, S.; Medina, M.; Delacourte, A.; Dingwall, C.; De Strooper, B.; Dotti, C. G., Neuronal membrane cholesterol loss enhances amyloid peptide generation. J Cell Biol 2004, 167, (5), 953-60. 218. Shiraishi, H.; Marutani, T.; Wang, H.-Q.; Maeda, Y.; Kurono, Y.; Takashirna, A.; Araki, W.; Nishimura, M.; Yanagisawa, K.; Komano, H., Reconstitution of g-secretase by truncated presenilin (PS) fragments revealed that PS C-terminal transmembrane domain is critical for formation of g-secretase complex. Genes to Cells 2006, 11, (l), 83- 93. 219. Thinakaran, G.; Borchelt, D. R.; Lee, M. K.; Slunt, H. H.; Spitzer, L.; Kim, G.; Ratovitsky, T.; Davenport, F .; Nordstedt, C.; Seeger, M.; Hardy, J.; Levey, A. 1.; Gandy, S. E.; Jenkins, N. A.; Copeland, N. G.; Price, D. L.; Sisodia, S. S., Endoproteolysis of presenilin 1 and accumulation of processed derivatives in vivo. Neuron 1996, 17, (1), 181-90. 220. Ratovitski, T.; Slunt, H. H.; Thinakaran, G.; Price, D. L.; Sisodia, S. S.; Borchelt, D. R, Endoproteolytic processing and stabilization of wild-type and mutant presenilin. J Biol Chem 1997, 272, (39), 24536-41. 221. Xu, H.; Sweeney, D.; Wang, R.; Thinakaran, G.; Lo, A. C.; Sisodia, S. S.; Greengard, P.; Gandy, S., Generation of Alzheimer beta-amyloid protein in the trans- Golgi network in the apparent absence of vesicle formation. Proc Natl Acad Sci U S A 1997, 94, (8), 3748-52. 222. Cook, D. G.; Forrnan, M. S.; Sung, J. C.; Leight, S.; Kolson, D. L.; Iwatsubo, T.; Lee, V. M.; Doms, R W., Alzheimer's A beta(l-42) is generated in the endoplasmic reticulum/intermediate compartment of NT2N cells. Nat Med 1997, 3, (9), 1021-3. 223. Greenfield, J. P.; Tsai, J.; Gouras, G. K.; Hai, B.; Thinakaran, G.; Checler, F.; Sisodia, S. S.; Greengard, P.; Xu, H., Endoplasmic reticulum and trans-Golgi network generate distinct populations of Alzheimer beta-amyloid peptides. Proc Natl Acad Sci U S A 1999, 96, (2), 742-7. 224. Takahashi, R. H.; Milner, T. A.; Li, F.; Nam, E. E.; Edgar, M. A.; Yamaguchi, H.; Beal, M. F.; Xu, H.; Greengard, P.; Gouras, G. K., Intraneuronal Alzheimer abeta42 122 P‘fir'rl—t accumulates in multivesicular bodies and is associated with synaptic pathology. Am J Pathol 2002, 161, (5), 1869-79. 225. Vetrivel, K S.; Cheng, H.; Lin, W.; Sakurai, T.; Li, T.; Nukina, N.; Wong, P. C.; Xu, H.; Thinakaran, G., Association of gamma-secretase with lipid rafts in post-Golgi and endosome membranes. J Biol Chem 2004, 279, (43), 44945-54. 226. Haass, C.; Schlossmacher, M. G.; Hung, A. Y.; Vigo-Pelfrey, C.; Mellon, A.; Ostaszewski, B. L.; Lieberburg, 1.; K00, E. H.; Schenk, D.; Teplow, D. B.; et al., Amyloid beta-peptide is produced by cultured cells during normal metabolism. Nature 1992, 359, (6393), 322-5. 227. Younkin, S. G., Evidence that A beta 42 is the real culprit in Alzheimer's disease. Ann Neurol 1995, 37, (3), 287-8. 228. Burdick, D.; Soreghan, B.; Kwon, M.; Kosmoski, J .; Knauer, M.; Henschen, A.; Yates, J .; Cotman, C.; Glabe, C., Assembly and aggregation properties of synthetic Alzheimer's A4/beta amyloid peptide analogs. J Biol Chem 1992, 267, (1), 546-54. 229. Irie, K.; Murakami, K.; Masuda, Y.; Morimoto, A.; Ohigashi, H.; Ohashi, R; Takegoshi, K.; Nagao, M.; Shimizu, T.; Shirasawa, T., Structure of beta-amyloid fibrils and its relevance to their neurotoxicity: implications for the pathogenesis of Alzheimer's disease. J Biosci Bioeng 2005, 99, (5), 437-47. 230. Chartier-Harlin, M. C.; Crawford, F.; Hamandi, K.; Mullan, M.; Goate, A.; Hardy, J .; Backhovens, H.; Martin, J. J.; Broeckhoven, C. V., Screening for the beta- amyloid precursor protein mutation (APP717: Val---Ile) in extended pedigrees with early onset Alzheimer's disease. Neurosci Lett 1991, 129, (1), 134-5. 231. Murrell, J.; Farlow, M.; Ghetti, B.; Benson, M. D., A mutation in the amyloid precursor protein associated with hereditary Alzheimer's disease. Science 1991, 254, (5028), 97-9. 232. Eckman, C. B.; Mehta, N. D.; Crook, R.; Perez-tur, J.; Prihar, G.; Pfeiffer, E.; Graff-Radford, N.; Hinder, P.; Yager, D.; Zenk, B.; Refolo, L. M.; Prada, C. M.; Younkin, S. G.; Hutton, M.; Hardy, J ., A new pathogenic mutation in the APP gene (I716V) increases the relative proportion of A beta 42(43). Hum Mol Genet 1997, 6, (12), 2087-9. 123 ‘- 233. Sherrington, R.; Rogaev, E. 1.; Liang, Y.; Rogaeva, E. A.; Levesque, G.; Ikeda, M.; Chi, H.; Lin, C.; Li, G.; Holman, K.; et al., Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature 1995, 375, (6534), 754-60. 234. Rogaev, E. 1.; Sherrington, R.; Rogaeva, E. A.; Levesque, G.; Ikeda, M.; Liang, Y.; Chi, H.; Lin, C.; Holman, K.; Tsuda, T.; et al., Familial Alzheimer's disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer's disease type 3 gene. Nature 1995, 376, (6543), 775-8. 235. Sisodia, S. S.; St George-Hyslop, P. H., gamma-Secretase, Notch, Abeta and Alzheimer's disease: where do the presenilins fit in? Nat Rev Neurosci 2002, 3, (4), 281- 90. 236. Iwatsubo, T., The gamma-secretase complex: machinery for intramembrane proteolysis. Curr Opin Neurobiol 2004, 14, (3), 379-83. 237. Roses, A. D., Apolipoprotein E and Alzheimer's disease. The tip of the susceptibility iceberg. Ann N Y Acad Sci 1998, 855, 738-43. 238. Wen, Y.; Onyewuchi, 0.; Yang, S.; Liu, R.; Simpkins James, W., Increased beta- secretase activity and expression in rats following transient cerebral ischemia. Brain Res 2004, 1009, (1-2), 1-8. 239. Rossner, S.; Lange-Dohna, C.; Zeitschel, U.; Perez-Polo, J. R., Alzheimer's disease beta-secretase BACEl is not a neuron-specific enzyme. J Neurochem 2005, 92, (2), 226-34. 240. Zhao, J.; Fu, Y.; Yasvoina, M.; Shao, P.; Hitt, B.; O'Connor, T.; Logan, S.; Maus, E.; Citron, M.; Berry, R.; Binder, L.; Vassar, R, Beta-site amyloid precursor protein cleaving enzyme 1 levels become elevated in neurons around amyloid plaques: implications for Alzheimer's disease pathogenesis. J Neurosci 2007, 27, (14), 3639-49. 241. Gupta, S. V.; Khosla, P., Palmitic and stearic acids similarly affect plasma lipoprotein metabolism in cynomolgus monkeys fed diets with adequate levels of linoleic acid. JNutr 2001, 131, (8), 2115-20. 242. Chang, K.-A.; Suh, Y.-H., Pathophysiological roles of amyloidogenic carboxy- terrninal fragments of the b-amyloid precursor protein in Alzheimer's disease. J Pharmacol Sci 2005, 97, (4), 461-471. 124 243. Li, Y.; Zhou, W.; Tong, Y.; He, G.; Song, W., Control of APP processing and Ab generation level by BACEl enzymatic activity and transcription. FASEB J 2006, 20, (2), 285-292. 244. Yan, S. D.; Yan, S. F.; Chen, X.; Fu, J.; Chen, M.; Kuppusamy, P.; Smith, M. A.; Perry, G.; Godman, G. C.; et al., Non-enzymatically glycated tau in Alzheimer's disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid b-peptide. Nat Med 1995, 1, (7), 693-9. 245. Hoyer, S., Abnormalities of glucose metabolism in Alzheimer's disease. Ann N Y Acad Sci 1991, 640, 53-8. 246. Munch, G.; Schinzel, R; Loske, C.; Wong, A.; Durany, N.; Li, J. J.; Vlassara, H.; Smith, M. A.; Perry, G.; Riederer, P., Alzheimer's disease--synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts. J Neural Transm 1998, 105, (4-5), 439-61. 247. Sorbi, S.; Piacentini, S.; Latorraca, S.; Piersanti, P.; Amaducci, L., Alterations in metabolic properties in fibroblasts in Alzheimer disease. Alzheimer Dis Assoc Disord 1995, 9, (2), 73-7. 248. Sheu, K. F .; Cooper, A. J.; Koike, K.; Koike, M.; Lindsay, J. G.; Blass, J. P., Abnormality of the alpha-ketoglutarate dehydrogenase complex in fibroblasts from familial Alzheimer's disease. Ann Neurol 1994, 35, (3), 312-8. 249. Sorbi, S.; Bird, E. D.; Blass, J. P., Decreased pyruvate dehydrogenase complex activity in Huntington and Alzheimer brain. Ann Neurol 1983, 13, (1), 72-8. 250. Kish, S. J ., Brain energy metabolizing enzymes in Alzheimer's disease: alpha- ketoglutarate dehydrogenase complex and cytochrome oxidase. Ann N Y A cad Sci 1997, 826, 218-28. 251. Gibson, G. E.; Park, L. C.; Sheu, K. F.; Blass, J. P.; Calingasan, N. Y., The alpha- ketoglutarate dehydrogenase complex in neurodegeneration. Neurochem Int 2000, 36, (2), 97-112. 252. Parker, W. D., Jr.; Mahr, N. J.; Filley, C. M.; Parks, J. K.; Hughes, D.; Young, D. A.; Cullum, C. M., Reduced platelet cytochrome c oxidase activity in Alzheimer's disease. Neurology 1994, 44, (6), 1086-90. 125 I" '_I 253. Cardoso, S. M.; Proenca, M. T.; Santos, S.; Santana, 1.; Oliveira, C. R, Cytochrome c oxidase is decreased in Alzheimer's disease platelets. Neurobiol Aging 2004, 25, (1), 105-10. 254. Kish, S. J.; Bergeron, C.; Rajput, A.; Dozic, S.; Mastrogiacomo, F.; Chang, L. J.; Wilson, J. M.; DiStefano, L. M.; Nobrega, J. N., Brain cytochrome oxidase in Alzheimer's disease. J Neurochem 1992, 59, (2), 776-9. 255. Mutisya, E. M.; Bowling, A. C.; Beal, M. F ., Cortical cytochrome oxidase activity is reduced in Alzheimer's disease. J Neurochem 1994, 63, (6), 2179-84. 256. Tretter, L.; Adam-Vizi, V., Inhibition of Krebs cycle enzymes by hydrogen peroxide: A key role of [alpha]-ketoglutarate dehydrogenase in limiting NADH production under oxidative stress. J Neurosci 2000, 20, (24), 8972-9. 257. Drzezga, A.; Grimmer, T.; Riemenschneider, M.; Lautenschlager, N.; Siebner, H.; Alexopoulus, P.; Minoshima, S.; Schwaiger, M.; Kurz, A., Prediction of individual clinical outcome in MCI by means of genetic assessment and 18F-FDG PET. J Nucl Med 2005, 46, (10), 1625-1632. 258. Gibson, G. E., Interactions of oxidative stress with cellular calcium dynamics and glucose metabolism in Alzheimer‘s disease. Free Radic Biol Med 2002, 32, (11), 1061- 1070. 259. Gibson, G. E.; Jope, R; Blass, J. P., Decreased synthesis of acetylcholine accompanying impaired oxidation of pyruvic acid in rat brain minces. Biochem J 1975, 148, (1), 17-23. 260. Rupprecht, R.; Holsboer, F., Neuroactive steroids: mechanisms of action and neuropsychopharmacological perspectives. Trends Neurosci 1999, 22, (9), 410-6. 261. Ehrenstein, G.; Galdzicki, Z.; Lange, G. D., The choline-leakage hypothesis for the loss of acetylcholine in Alzheimer's disease. Biophys J 1997, 73, (3), 1276-80. 262. Sims, N. R.; Bowen, D. M.; Allen, S. J.; Smith, C. C.; Neary, D.; Thomas, D. J.; Davison, A. N., Presynaptic cholinergic dysfunction in patients with dementia. J Neurochem 1983, 40, (2), 503-9. 126 263. Nitsch, R. M.; Slack, B. E.; Wurtman, R. J.; Growdon, J. H., Release of Alzheimer amyloid precursor derivatives stimulated by activation of muscarinic acetylcholine receptors. Science 1992, 258, (5080), 304-7. 264. Baskin, D. S.; Browning, J. L.; Pirozzolo, F. J.; Korporaal, S.; Baskin, J. A.; Appel, S. H., Brain choline acetyltransferase and mental function in Alzheimer disease. Arch Neurol 1999, 56, (9), 1121-3. 265. Hoyer, S., Oxidative metabolism deficiencies in brains of patients with Alzheimer's disease. Acta Neurol Scand Suppl 1996, 165, 18-24. 266. Hoyer, A.; Bardenheuer, H. J .; Martin, E.; Plaschke, K., Amyloid precursor protein (APP) and its derivatives change after cellular energy depletion. An in vitro- study. J Neural Transm 2005, 112, (2), 239-253. 267. Planel, E.; Miyasaka, T.; Launey, T.; Chui, D.-H.; Tanemura, K.; Sato, S.; Murayama, O.; Ishiguro, K.; Tatebayashi, Y.; Takashima, A., Alterations in glucose metabolism induce hypothermia leading to tau hyperphosphorylation through differential inhibition of kinase and phosphatase activities: implications for Alzheimer's disease. J Neurosci 2004, 24, (10), 2401-241 1. 268. Velliquette, R. A.; O'Connor, T.; Vassar, R., Energy inhibition elevates b- secretase levels and activity and is potentially amyloidogenic in APP transgenic mice: Possible early events in Alzheimer's disease pathogenesis. J Neurosci 2005, 25, (47), 10874-10883. 269. Gong, C.-X.; Liu, F.; Grundke-Iqbal and, 1.; Iqbal, K., Impaired brain glucose metabolism leads to Alzheimer neurofibrillary degeneration through a decrease in tau O- GlcNAcylation. J Alzheimers Dis 2006, 9, (1), 1-12. 270. Liu, F.; Iqbal, K.; Grundke-Iqbal, 1.; Hart, G. W.; Gong, C.-X., O-GlcNAcylation regulates phosphorylation of tau: A mechanism involved in Alzheimer's disease. Proc Natl Acad Sci U SA 2004, 101, (29), 10804-10809. 271. Chih, C. P.; Lipton, P.; Roberts, E. L., Do active cerebral neurons really use lactate rather than glucose? Trends Neurosci 2001, 24, (10), 573-578. 272. Pellerin, L.; Magistretti, P. J ., Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci U SA 1994, 91, (22), 10625-9. 127 273. Kasischke, K. A.; Vishwasrao, H. D.; Fisher, P. J.; Zipfel, W. R.; Webb, W. W., Neural Activity Triggers Neuronal Oxidative Metabolism Followed by Astrocytic Glycolysis. Science 2004, 305, (5680), 99-103. 274. Patil, S.; Li, Z.; Chan, C., Cellular to tissue informatics: approaches to optimizing cellular function of engineered tissue. Adv Biochem Eng Biotechnol 2006, 102, 139-59. 275. Srivastava, S.; Chan, C., Application of metabolic flux analysis to identify the .5 mechanisms of free fatty acid toxicity to human hepatoma cell line. Biotechnol Bioeng 2007. 276. Chan, C.; Berthiaume, F.; Lee, K.; Yarmush, M. L., Metabolic flux analysis of cultured hepatocytes exposed to plasma. Biotechnol Bioeng 2003, 81, (1), 33-49. 277. Rose, C.; F elipo, V., Limited capacity for ammonia removal by brain in chronic '1’ liver failure: potential role of nitric oxide. Metab Brain Dis 2005, 20, (4), 275-83. 278. McGinnis, J. F.; de Vellis, J., Cell surface modulation of gene expression in brain cells by down regulation of glucocorticoid receptors. Proc Natl Acad Sci U S A 1981, 78, (2), 1288-92. 279. Bligh, E. G.; Dyer, W. J., A rapid method of total lipide extraction and purification. Can J Biochem Physio] 1959, 37, 911-17. 280. Merrill, A. H., Jr.; Wang, E.; Mullins, R E.; Jamison, W. C. L.; Nimkar, S.; Liotta, D. C., Quantitation of free sphingosine in liver by high-performance liquid chromatography. Anal Biochem 1988, 171, (2), 373-81. 281. Berry, M. N.; Phillips, J. W.; Henly, D. C.; Clark, D. 6., Effects of fatty acid oxidation on glucose utilization by isolated hepatocytes. FEBS Lett 1993, 319, (1-2), 26- 30. 282. Hue, L.; Maisin, L.; Rider, M. H., Palmitate inhibits liver glycolysis. Involvement of fructose 2,6-bisphosphate in the glucose/fatty acid cycle. Biochem J 1988, 251, (2), 541 -5. 283. Ji, H.; Graczyk-Milbrandt, G.; Osbakken, M. D.; Friedman, M. 1., Interactions of dietary fat and 2,5-anhydro-D-mannitol on energy metabolism in isolated rat hepatocytes. Am J Physiol 2002, 282, (3, Pt. 2), R715-R720. 128 284. Lovell, M. A.; Xie, G.; Markesbery, W. R, Acrolein, a product of lipid peroxidation, inhibits glucose and glutamate uptake in primary neuronal cultures. Free Radic Biol Med 2000, 29, (8), 714-720. 285. Mark, R. J.; Pang, Z.; Geddes, J. W.; Uchida, K.; Mattson, M. P., Amyloid b- peptide impairs glucose transport in hippocampal and cortical neurons: involvement of membrane lipid peroxidation. J Neurosci 1997, 17, (3), 1046-1054. 286. Miwa, I.; Adachi, K.; Murase, S.; Hamada, Y.; Sugiura, M., 4-Hydroxy-2-nonenal hardly affects glycolysis. Free Radic Biol Med 1997, 23, (4), 610-615. 287. Smith, J. M.; Solar, S. M.; Paulson, D. J.; Hill, N. M.; Broderick, T. L., Effect of palrnitate on carbohydrate utilization and Na/K-ATPase activity in aortic vascular smooth muscle from diabetic rats. Mol Cell Biochem 1999, 194, (1-2), 125-32. 288. King, L. M.; Sidell, R J.; Wilding, J. R.; Radda, G. K.; Clarke, K., Free fatty acids, but not ketone bodies, protect diabetic rat hearts during low-flow ischemia. Am J Physiol Heart Circ Physiol 2001, 280, (3), H1173-81. 289. F ukuyama, H.; Ogawa, M.; Yamauchi, H.; Yamaguchi, S.; Kimura, J.; Yonekura, Y.; Konishi, J ., Altered cerebral energy metabolism in Alzheimer's disease: a PET study. JNucl Med 1994, 35, (1), 1-6. 290. Hoyer, S., Risk factors for Alzheimer's disease during aging. Impacts of glucose/energy metabolism. J Neural Transm Suppl 1998, 54, 187-94. 291. Pettegrew, J. W.; Klunk, W. E.; Kanal, E.; Panchalingam, K.; McClure, R J., Changes in brain membrane phospholipid and high-energy phosphate metabolism precede dementia. Neurobiol Aging 1995, 16, (6), 973-5. 292. Pifferi, F .; Roux, F .; Langelier, B.; Alessandri, J.-M.; Vancassel, S.; Jouin, M.; Lavialle, M.; Guesnet, P., (n-3) polyunsaturated fatty acid deficiency reduces the expression of both isoforms of the brain glucose transporter GLUTl in rats. J Nutr 2005, 135, (9), 2241-2246. 293. Li, J.; Hu, X.; Selvakurnar, P.; Russell, R. R, 111; Cushman, S. W.; Holman, G. D.; Young, L. H., Role of the nitric oxide pathway in AMPK-mediated glucose uptake and GLUT4 translocation in heart muscle. Am J Physiol 2004, 287, (5, Pt. 1), E834-E841. 129 294. Turnley, A. M.; Stapleton, D.; Mann, R. J.; Witters, L. A.; Kemp, B. E.; Bartlett, P. F., Cellular distribution and developmental expression of AMP-activated protein kinase isoforms in mouse central nervous system. J Neurochem 1999, 72, (4), 1707-16. 295. Blazquez, C.; Woods, A.; de Ceballos, M. L.; Carling, D.; Guzman, M., The AMP-activated protein kinase is involved in the regulation of ketone body production by astrocytes. J Neurochem 1999, 73, (4), 1674-82. 296. Giri, S.; Nath, N.; Smith, R; Viollet, B.; Singh Avtar, K.; Singh, 1., 5- aminoimidazole-4-carboxamide-l-beta-4—ribofuranoside inhibits proinflammatory response in glial cells: a possible role of AMP-activated protein kinase. J Neurosci 2004, 24, (2), 479-87. 297. McCullough Louise, D.; Zeng, Z.; Li, H.; Landree Leslie, E.; McFadden, J .; Ronnett Gabriele, V., Pharmacological inhibition of AMP-activated protein kinase provides neuroprotection in stroke. J Biol Chem 2005, 280, (21), 20493-502. 298. Simpson, I. A.; Chundu, K. R.; Davies-Hill, T.; Honer, W. G.; Davies, P., Decreased concentrations of GLUTl and GLUT3 glucose transporters in the brains of patients with Alzheimer's disease. Ann Neurol 1994, 35, (5), 546-51. 299. Mandelin, A. M., 2000. Alzheimer's disease severity is associated with a progressive decline in glutl gene expression: role of posttranscriptional regulation. 193 pp. From: Diss. Abstr. Int., B 2002, 62(11), 4951. 300. Listenberger, L. L.; Ory, D. S.; Schaffer, J. 13., Palrnitate-induced apoptosis can occur through a ceramide-independent pathway. J Biol Chem 2001, 276, (18), 14890-5. 301. Palicz, A.; Foubert, T. R.; Jesaitis, A. J.; Marodi, L.; McPhail, L. C., Phosphatidic acid and diacylglycerol directly activate NADPH oxidase by interacting with enzyme components. J Biol Chem 2001, 276, (5), 3090-7. 302. Lu, Z. H.; Mu, Y. M.; Wang, B. A.; Li, X. L.; Lu, J. M.; Li, J. Y.; Pan, C. Y.; Yanase, T.; Nawata, H., Saturated free fatty acids, palmitic acid and stearic acid, induce apoptosis by stimulation of ceramide generation in rat testicular Leydig cell. Biochem Biophys Res Commun 2003, 303, (4), 1002-7. 303. Yi, F .; Zhang, A. Y.; Janscha, J. L.; Li, P.-L.; Zou, A.-P., Homocysteine activates NADH/NADPH oxidase through ceramide-stimulated Rac GTPase activity in rat mesangial cells. Kidney Int 2004, 66, (5), 1977-1987. 130 304. Borg, J.; London, J ., Copper/zinc superoxide dismutase overexpression promotes survival of cortical neurons exposed to neurotoxins in vitro. J Neurosci Res 2002, 70, (2), 180-9. 305. Lieb, K.; Fiebich, B. L.; Hull, M.; Berger, M.; Bauer, J., Potent inhibition of interleukin-6 expression in a human astrocytoma cell line by tenidap. Cell Tissue Res 1997, 288, (2), 251-257. 306. Pahan, K.; Sheikh, F. G.; Khan, M.; Namboodiri, A. M. S.; Singh, 1., Sphingomyelinase and ceramide stimulate the expression of inducible nitric-oxide synthase in rat primary astrocytes. J Biol Chem 1998, 273, (5), 2591-2600. 307. Floyd, R. A.; Hensley, K; Jaffery, F .; Maidt, L.; Robinson, K.; Pye, Q.; Stewart, C., Increased oxidative stress brought on by pro-inflammatory cytokines in neurodegenerative processes and the protective role of nitrone-based free radical traps. Life Sci 1999, 65, (18/19), 1893-1899. FAA-IS ”I HAHJL'A. - 308. Wei, T.; Chen, C.; Hou, J.; Xin, W.; Mori, A., Nitric oxide induces oxidative stress and apoptosis in neuronal cells. Biochim Biophys Acta 2000, 1498, (1), 72-79. 309. Zwingmann, C.; Richter-Landsberg, C.; Brand, A.; Leibfritz, D., NMR spectroscopic study on the metabolic fate of [3-(13)C]alanine in astrocytes, neurons, and cocultures: implications for glia-neuron interactions in neurotransmitter metabolism. Glia 2000, 32, (3), 286-303. 310. Sykes, J. E.; Lopes-Cardozo, M., Effect of exogenous fatty acids on lipid synthesis, marker-enzymes, and development of glial cells maintained in serum-free culture. Glia 1990, 3, (6), 495-501. 311. Sharma, K.; Shi, Y., The yins and yangs of ceramide. Cell Res 1999, 9, (1), 1-10. 312. Perry, D. K.; Hannun, Y. A., The role of ceramide in cell signaling. Biochim Biophys Acta 1998, 1436, (1-2), 233-43. 313. Holland, W. L.; Brozinick, J. T.; Wang, L. P.; Hawkins, E. D.; Sargent, K. M.; Liu, Y.; Narra, K.; Hoehn, K. L.; Knotts, T. A.; Siesky, A.; Nelson, D. H.; Karathanasis, S. K; Fontenot, G. K.; Bimbaum, M. J.; Summers, S. A., Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance. Cell Metab 2007, 5, (3), 167-79. 131 314. Haughey, N. J.; Cutler, R. G.; Tamara, A.; McArthur, J. C.; Vargas, D. L.; Pardo, C. A.; Turchan, J .; Nath, A.; Mattson, M. P., Perturbation of sphingolipid metabolism and ceramide production in HIV-dementia. Ann Neur012004, 55, (2), 257-67. 315. Han, X.; Holtzman, D. M.; McKeel, D. W., Jr.; Kelley, J.; Morris, J. C., Substantial sulfatide deficiency and ceramide elevation in very early Alzheimer's disease: potential role in disease pathogenesis. J Neurochem 2002, 82, (4), 809-818. 316. Cutler, R. G.; Kelly, J .; Storie, K.; Pedersen, W. A.; Tammara, A.; Hatanpaa, K.; Troncoso, J. C.; Mattson, M. P., Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer's disease. Proc Natl Acad Sci U S A 2004, 101, (7), 2070-2075. 317. Satoi, H.; Tomimoto, H.; Ohtani, R.; Kitano, T.; Kondo, T.; Watanabe, M.; Oka, N.; Akiguchi, 1.; Furuya, S.; Hirabayashi, Y.; Okazaki, T., Astroglial expression of L ceramide in Alzheimer's disease brains: a role during neuronal apoptosis. Neuroscience 2005, 130, (3), 657-666. 318. Han, X.; D, M. H.; McKeel, D. W., Jr.; Kelley, J.; Morris, J. C., Substantial sulfatide deficiency and ceramide elevation in very early Alzheimer's disease: potential role in disease pathogenesis. J Neurochem 2002, 82, (4), 809-18. 319. Katsel, P.; Li, C.; Haroutunian, V., 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? Neurochem Res 2007, 32, (4-5), 845-56. 320. Malaplate-Arrnand, C.; Florent-Bechard, S.; Youssef, 1.; Koziel, V.; Sponne, 1.; Kriem, B.; Leininger-Muller, B.; Olivier, J. L.; Oster, T.; Pillot, T., Soluble oligomers of amyloid-beta peptide induce neuronal apoptosis by activating a cPLA2-dependent Sphingomyelinase-ceramide pathway. Neurobiol Dis 2006, 23, (1), 178-89. 321. Jana, A.; Pahan, K., Fibrillar amyloid-beta peptides kill human primary neurons via NADPH oxidase-mediated activation of neutral Sphingomyelinase. Implications for Alzheimer's disease. J Biol Chem 2004, 279, (49), 51451-9. 322. Lee, J. T.; Xu, J.; Lee, J. M.; Ku, G.; Han, X.; Yang, D. 1.; Chen, S.; Hsu, C. Y., Amyloid-beta peptide induces oligodendrocyte death by activating the neutral Sphingomyelinase-ceramide pathway. J Cell Biol 2004, 164, (1), 123-31. 132 323. Marty, M. S.; Atchison, W. D., Pathways mediating Ca2+ entry in rat cerebellar granule cells following in vitro exposure to methyl mercury. T oxicol Appl Pharmacol 1997, 147, (2), 319-30. 324. Puglielli, L.; Ellis, B. C.; Saunders, A. J.; Kovacs, D. M., Ceramide stabilizes b- site amyloid precursor protein-cleaving enzyme 1 and promotes amyloid b-peptide biogenesis. J Biol Chem 2003, 278, (22), 19777-19783. 325. Fan, Q. W.; Yu, W.; Gong, J. S.; Zou, K.; Sawamura, N.; Senda, T.; Yanagisawa, 1, K.; Michikawa, M., Cholesterol-dependent modulation of dendrite outgrth and ” microtubule stability in cultured neurons. J Neurochem 2002, 80, (1), 178-90. ‘ 326. Emamian, E. S.; Hall, D.; Bimbaum, M. J.; Karayiorgou, M.; Gogos, J. A., Convergent evidence for impaired AKTl-GSK3b signaling in schizophrenia. Nat Genet 2004, 36, (2), 131-137. L 327. de la Monte, S. M.; Ganju, N.; Feroz, N.; Luong, T.; Banerjee, K; Cannon, J.; Wands, J. R, Oxygen free radical injury is sufficient to cause some Alzheimer-type molecular abnormalities in human CNS neuronal cells. J Alzheimers Dis 2000, 2, (3-4), 261-281. 328. Lee, M.-s.; Kwon, Y. T.; Li, M.; Peng, J.; Friedlander, R. M.; Tsai, L.-H., Neurotoxicity induces cleavage of p35 to p25 by calpain. Nature 2000, 405, (6784), 360- 364. 329. Kerokoski, P.; Suuronen, T.; Salminen, A.; Soininen, H.; Pirttila, T., Cleavage of the cyclin-dependent kinase 5 activator p35 to p25 does not induce tau hyperphosphorylation. Biochem Biophys Res Commun 2002, 298, (5), 693-698. 330. Takashima, A., GSK-3 is essential in the pathogenesis of Alzheimer's disease. J Alzheimers Dis 2006, 9, 309-317. 331. Phiel, C. J.; Wilson, C. A.; Lee, V. M. Y.; Klein, P. S., GSK-3a regulates production of Alzheimer's disease amyloid-b peptides. Nature 2003, 423, (6938), 435- 439. 332. Beffert, U.; Cohn, J. S.; Petit-Turcotte, C.; Tremblay, M.; Aumont, N.; Ramassamy, C.; Davignon, J.; Poirier, J., Apolipoprotein E and b-amyloid levels in the hippocampus and frontal cortex of Alzheimer's disease subjects are disease-related and apolipoprotein E genotype dependent. Brain Res 1999, 843, (1-2), 87-94. 133 F 333. Szutowicz, A.; Bielarczyk, H.; Gul, S.; Ronowska, A.; Pawelczyk, T.; Jankowska- Kulawy, A., Phenotype-dependent susceptibility of cholinergic neuroblastoma cells to neurotoxic inputs. Metabol Brain Dis 2006, 21, (2/3), 149-161. 334. Szutowicz, A.; Lysiak, W., Regional and subcellular distribution of ATP-citrate lyase and other enzymes of acetyl-CoA metabolism in rat brain. J Neurochem 1980, 35, (4), 775-85. 335. Pei, Z.; Oey, N. A.; Zuidervaart, M. M.; Jia, Z.; Li, Y.; Steinberg, S. J.; Smith, K D.; Watkins, P. A., The Acyl-CoA Synthetase \"Bubblegum\" (Lipidosin): Further Characterization and Role in Neuronal Fatty Acid b-Oxidation. J Biol Chem 2003, 278, (47), 47070-47078. 336. Alessenko, A. V.; Bugrova, A. E.; Dudnik, L. 3., Connection of lipid peroxide oxidation with the Sphingomyelin pathway in the development of Alzheimer's disease. Biochem Soc Trans 2004, 32, (Pt 1), 144-6. 337. Patil, S.; Chan, C., Palmitic and stearic fatty acids induce Alzheimer-like hyperphosphorylation of tau in primary rat cortical neurons. Neurosci Lett 2005, 384, (3), 288-293. 338. Patil, S.; Sheng, L.; Masserang, A.; Chan, C., Palmitic acid-treated astrocytes induce BACEl upregulation and accumulation of C-terrninal fragment of APP in primary cortical neurons. Neurosci Lett 2006, 406, (1-2), 55-9. 339. Sultana, R.; Boyd-Kimball, D.; Poon, H. F.; Cai, J.; Pierce, W. M.; Klein, J. 3.; Merchant, M.; Markesbery, W. R.; Butterfield, D. A., Redox proteomics identification of oxidized proteins in Alzheimer's disease hippocampus and cerebellum: an approach to understand pathological and biochemical alterations in AD. Neurobiol Aging 2006, 27, (11), 1564-1576. 340. Sultana, R.; Poon, H. F.; Cai, J.; Pierce, W. M.; Merchant, M.; Klein, J. B.; Markesbery, W. R.; Butterfield, D. A., Identification of nitrated proteins in Alzheimer's disease brain using a redox proteomics approach. Neurobiol Dis 2006, 22, (1), 76-87. 341. Trepanier, G.; Furling, D.; Puymirat, J.; Mirault, M. E., Irnmunocytochemical localization of seleno-glutathione peroxidase in the adult mouse brain. Neuroscience 1996, 75, (1), 231-243. 134 342. Wu, C.-K; Thal, L.; Pizzo, D.; Hansen, L.; Masliah, E.; Geula, C., Apoptotic signals within the basal forebrain cholinergic neurons in Alzheimer's disease. Exp Neurol 2005, 195, (2), 484-496. 343. McKinney, M.; Coyle, J. T.; Hedreen, J. C., Topographic analysis of the innervation of the rat neocortex and hippocampus by the basal forebrain cholinergic system. J Comp Neurol 1983, 217, (1), 103-21. 344. Ransmayr, G.; Cervera, P.; Hirsch, E.; Ruberg, M.; Hersh, L. B.; Duyckaerts, C.; Hauw, J. J.; Delumeau, C.; Agid, Y., Choline acetyltransferase-like immunoreactivity in the hippocampal formation of control subjects and patients with Alzheimer's disease. Neuroscience 1989, 32, (3), 701-14. 345. Arendt, T.; Bigl, V.; Tennstedt, A.; Arendt, A., Neuronal loss in different parts of the nucleus basalis is related to neuritic plaque formation in cortical target areas in I- Alzheimer's disease. Neurosci 1985, 14, (1), 1-14. 346. Kashiwaya, Y.; Takeshima, T.; Mori, N.; Nakashima, K.; Clarke, K.; Veech, R. L., D-beta-hydroxybutyrate protects neurons in models of Alzheimer's and Parkinson's disease. Proc Natl A cad Sci U S A 2000, 97, (10), 5440-4. 347. Wu, M. M.; Chiou, H. Y.; Wang, T. W.; Hsueh, Y. M.; Wang, 1. H.; Chen, C. J.; Lee, T. C., Association of blood arsenic levels with increased reactive oxidants and decreased antioxidant capacity in a human population of northeastern Taiwan. Environ Health Perspect 2001, 109, (10), 1011-7. 348. Mudher, A.; Lovestone, S., Alzheimer's disease-do tauists and baptists finally shake hands? Trends Neurosci 2002, 25, (1), 22-6. 349. Yuan, H.; Zheng, J. C.; Liu, P.; Zhang, S. F.; Xu, J. Y.; Bai, L. M., Pathogenesis of Parkinson's disease: oxidative stress, environmental impact factors and inflammatory processes. Neurosci Bull 2007, 23, (2), 125-30. 350. Pereira, A. C.; Wu, W.; Small, S. A., Imaging-guided microarray: isolating molecular profiles that dissociate Alzheimer's disease from normal aging. Ann N 1’ Acad Sci 2007, 1097, 225-38. 351. Blanchard, B. J.; Hiniker, A. E.; Lu, C. C.; Margolin, Y.; Yu, A. S.; Ingram, V. M., Elimination of Amyloid beta Neurotoxicity. J Alzheimers Dis 2000, 2, (2), 137-149. 135 352. Cheng, I. H.; Scearce-Levie, K.; Legleiter, J .; Palop, J. J.; Gerstein, H.; Bien-Ly, N.; Puolivali, J.; Lesne, S.; Ashe, K. H.; Muchowski, P. J.; Mucke, L., Accelerating Amyloid-beta Fibrillization Reduces Oligomer Levels and Functional Deficits in Alzheimer Disease Mouse Models. J Biol Chem 2007, 282, (33), 23818-28. 353. Azad, N. A.; A1 Bugami, M.; Loy-English, 1., Gender differences in dementia risk factors. Gend Med 2007, 4, (2), 120-9. 354. Bonomo, S. M.; Rigamonti, A. E.; Giunta, M.; Galimberti, D.; Guaita, A.; Gagliano, M. G.; Muller, E. E.; Cella, S. G., Menopausal transition: A possible risk factor for brain pathologic events. Neurobiol Aging 2007. 355. Saez, T. E.; Pehar, M.; Vargas, M.; Barbeito, L.; Maccioni, R. B., Astrocytic nitric oxide triggers tau hyperphosphorylation in hippocampal neurons. In Vivo 2004, 18, (3), 275-80. 356. Ayasolla, K.; Khan, M.; Singh, A. K.; Singh, 1., Inflammatory mediator and beta- amyloid (25-35)-induced ceramide generation and iNOS expression are inhibited by vitamin E. Free Radic Biol Med 2004, 37, (3), 325-38. 357. Vesce, S.; Rossi, D.; Brambilla, L.; Volterra, A., Glutamate release from astrocytes in physiological conditions and in neurodegenerative disorders characterized by neuroinflammation. Int Rev Neurobi012007, 82, 57-71. 358. Tilleux, S.; Herrnans, E., Neuroinflammation and regulation of glial glutamate uptake in neurological disorders. J Neurosci Res 2007, 85, (10), 2059-70. 359. Takuma, K.; Baba, A.; Matsuda, T., Astrocyte apoptosis: implications for neuroprotection. Prog Neurobiol 2004, 72, (2), 111-27. 360. Assis-Nascimento, P.; Jarvis, K. M.; Montague, J. R.; Mudd, L. M., Beta-amyloid toxicity in embryonic rat astrocytes. Neurochem Res 2007, 32, (9), 1476-82. 361. Pardridge, W. M., The blood-brain barrier and neurotherapeutics. NeuroRx 2005, 2, (1), 1-2. 362. Bennett, J. W.; Klich, M., Mycotoxins. Clin Microbiol Rev 2003, 16, (3), 497- 516. 136 363. Sundaram, K. S.; Lev, M., Inhibition of cerebroside synthesis in the brains of mice treated with L-cycloserine. J Lipid Res 1985, 26, (4), 473-7. 137 HI 5 nlltltltltllllllu