the processing of amyloid precursor protein in the central

141
Washington University in St. Louis Washington University in St. Louis Washington University Open Scholarship Washington University Open Scholarship All Theses and Dissertations (ETDs) 12-31-2013 The Processing of Amyloid Precursor Protein in the Central The Processing of Amyloid Precursor Protein in the Central Nervous System of Humans and Rhesus Macaques Nervous System of Humans and Rhesus Macaques Justyna Anna Dobrowolska Washington University in St. Louis Follow this and additional works at: https://openscholarship.wustl.edu/etd Part of the Neuroscience and Neurobiology Commons Recommended Citation Recommended Citation Dobrowolska, Justyna Anna, "The Processing of Amyloid Precursor Protein in the Central Nervous System of Humans and Rhesus Macaques" (2013). All Theses and Dissertations (ETDs). 1202. https://openscholarship.wustl.edu/etd/1202 This Dissertation is brought to you for free and open access by Washington University Open Scholarship. It has been accepted for inclusion in All Theses and Dissertations (ETDs) by an authorized administrator of Washington University Open Scholarship. For more information, please contact [email protected].

Upload: others

Post on 05-May-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Processing of Amyloid Precursor Protein in the Central

Washington University in St. Louis Washington University in St. Louis

Washington University Open Scholarship Washington University Open Scholarship

All Theses and Dissertations (ETDs)

12-31-2013

The Processing of Amyloid Precursor Protein in the Central The Processing of Amyloid Precursor Protein in the Central

Nervous System of Humans and Rhesus Macaques Nervous System of Humans and Rhesus Macaques

Justyna Anna Dobrowolska Washington University in St. Louis

Follow this and additional works at: https://openscholarship.wustl.edu/etd

Part of the Neuroscience and Neurobiology Commons

Recommended Citation Recommended Citation Dobrowolska, Justyna Anna, "The Processing of Amyloid Precursor Protein in the Central Nervous System of Humans and Rhesus Macaques" (2013). All Theses and Dissertations (ETDs). 1202. https://openscholarship.wustl.edu/etd/1202

This Dissertation is brought to you for free and open access by Washington University Open Scholarship. It has been accepted for inclusion in All Theses and Dissertations (ETDs) by an authorized administrator of Washington University Open Scholarship. For more information, please contact [email protected].

Page 2: The Processing of Amyloid Precursor Protein in the Central

WASHINGTON UNIVERSITY IN ST. LOUIS

Division of Biology and Biomedical Sciences

Neurosciences

Dissertation Examination Committee:

Randall J. Bateman, Chair David L. Brody

Guojun Bu John R. Cirrito

Alison M. Goate Michael L. Gross

Jin-Moo Lee

The Processing of Amyloid Precursor Protein in the

Central Nervous System of Humans and Rhesus Macaques

by

Justyna Anna Dobrowolska

A dissertation presented to the Graduate School of Arts and Sciences

of Washington University in partial fulfillment of the

requirements for the degree of Doctor of Philosophy

December 2013

Saint Louis, Missouri

Page 3: The Processing of Amyloid Precursor Protein in the Central

© 2013, Justyna Anna Dobrowolska

Page 4: The Processing of Amyloid Precursor Protein in the Central

ii

Table of Contents List of Figures .............................................................................................................................................. iv

List of Tables ............................................................................................................................................... vi

List of Abbreviations .................................................................................................................................. vii

Acknowledgments ........................................................................................................................................ xi

Abstract of the Dissertation ....................................................................................................................... xiv

CHAPTER 1 INTRODUCTION AND PERSPECTIVE .............................................................................. 1

Alzheimer’s Disease as a Major Public Health Issue .................................................................................... 1

The Amyloid Hypothesis .............................................................................................................................. 2

Amyloid Precursor Protein Expression ......................................................................................................... 4

Amyloid Precursor Protein Localization ....................................................................................................... 4

Proteolytic Processing of Amyloid Precursor Protein .................................................................................. 5

Trafficking of Amyloid Precursor Protein .................................................................................................... 8

Diurnal patterns of Amyloid-β ...................................................................................................................... 9

Elevated β-secretase in AD ......................................................................................................................... 10

In vivo Stable Isotope Labeling and the LC/MS approach ......................................................................... 11

CHAPTER 2 DIURNAL REGULATION OF AMYLOID PRECURSOR PROTEIN IN HUMANS ...... 12

Introduction ................................................................................................................................................. 12

Experimental Methods and Study Design ................................................................................................... 13

Results ......................................................................................................................................................... 21

Discussion ................................................................................................................................................... 39

CHAPTER 3 SOLUBLE AMYLOID PRECURSOR PROTEIN TURNOVER RATES IN HEALTHY, YOUNG HUMANS .................................................................................................................................... 44

Page 5: The Processing of Amyloid Precursor Protein in the Central

iii

Introduction ................................................................................................................................................. 44

Experimental Methods and Study Design ................................................................................................... 45

Results ......................................................................................................................................................... 54

Discussion ................................................................................................................................................... 56

CHAPTER 4 METHOD DEVELOPMENT FOR SAPPΑ AND SAPPΒ KINETIC MEASUREMENTS IN HEALTHY, YOUNG HUMANS .......................................................................................................... 58

Introduction ................................................................................................................................................. 58

Experimental Methods and Study Design ................................................................................................... 59

Results ......................................................................................................................................................... 64

Discussion ................................................................................................................................................... 73

CHAPTER 5 Β-SECRETASE INHIBITION ON THE EFFECT OF CONCENTRATIONS AND KINETIC OF SAPPΑ, SAPPΒ, AND AΒ IN RHESUS MACAQUES ..................................................... 75

Introduction ................................................................................................................................................. 75

Methods and Experimental Design ............................................................................................................. 76

Results ......................................................................................................................................................... 86

Discussion ................................................................................................................................................... 94

CHAPTER 6 GENERAL CONCLUSIONS AND FUTURE DIRECTIONS ............................................ 99

Bibliography ............................................................................................................................................ cxiii

Page 6: The Processing of Amyloid Precursor Protein in the Central

iv

List of Figures

FIGURE 1.1. PROTEOLYTIC PROCESSING OF AMYLOID PRECURSOR PROTEIN. ................................................................ 5 FIGURE 1.2. DIURNAL FLUCTUATIONS OF AΒ OBSERVED IN HUMANS ............................................................................. 9 FIGURE 2.1 EFFECT OF TWO FREEZE/THAW CYCLES ON CSF CONCENTRATIONS OF SAPPΑ ......................................... 16 FIGURE 2.2. SPECIFICITY AND SELECTIVITY OF THE SAPPΒ ELISA. ............................................................................. 22 FIGURE 2.4. OPTIMIZATION OF SAPPΑ ELISA ............................................................................................................. 24 FIGURE 2.5. DETERMINATION OF DILUTION FACTOR FOR CSF. .................................................................................... 25 FIGURE 2.6. DIURNAL PATTERNS IN APP METABOLITES.. ............................................................................................. 27 FIGURE 2.7. CIRCADIAN RHYTHM PARAMETERS OF FOUR APP METABOLITES IN YNC, AMYLOID-, AND AMYLOID+

GROUPS. ............................................................................................................................................................... 30 FIGURE 2.8. COSINOR-FITTED GROUP-AVERAGED TOTAL PROTEIN CONCENTRATIONS DETERMINED USING A MICRO

BCA ASSAY ......................................................................................................................................................... 36 FIGURE 2.9. CORRELATIONS BETWEEN APP METABOLITES.. ....................................................................................... 37 FIGURE 2.10. SEPARATING PARTICIPANT GROUPS BY THE SAPPΒ/SAPPΑ RATIO.. ....................................................... 39 FIGURE 3.1. METABOLISM OF AΒ IN HEALTHY YOUNG HUMANS (N=6) (BATEMAN ET AL., 2006). ............................... 44 FIGURE 3.2. HUMAN LABELED LEUCINE INFUSION PROTOCOL AND SAMPLE COLLECTION ............................................ 48 FIGURE 3.3. ISOLATION OF AMYLOID PRECURSOR PROTEIN FROM CEREBROSPINAL FLUID. ........................................ 50 FIGURE 3.4. LIQUID CHROMATOGRAPHY-MASS SPECTROMETRY ANALYSIS OF SAPP PEPTIDES. . ............................... 51 FIGURE 3.5. PRECURSOR AND PRODUCT ION CHROMATOGRAMS AND PRODUCT ION MASS SPECTRA FOR SAPP PEPTIDE.

............................................................................................................................................................................ 52 FIGURE 3.6. TOTAL SAPP IMMUNOPRECIPITATED FROM HUMAN CSF WAS DIGESTED WITH TRYPSIN, AND TRYPTIC

PEPTIDES CONTAINING 13C6-LEUCINE WERE USED TO MEASURE TOTAL SAPP KINETICS. THE FRACTIONAL

SYNTHESIS RATE (FSR) OVER 36 H WAS CALCULATED FROM THE 13C6-SAPP KINETIC CURVE. ............................ 55

FIGURE 3.7. METABOLISM OF TOTAL SAPP IN HEALTHY YOUNG HUMANS (DOBROWOLSKA ET AL., 2008). ................. 56 FIGURE 4.1. AMYLOID PRECURSOR PROTEIN SEQUENCE WITH POSITIONS OF PROTEOLYTIC CLEAVAGE, ANTIBODY

BINDING SITES, AND PEPTIDES CHOSEN FOR QUANTITATION. ............................................................................... 58 FIGURE 4.2. CHROMATOGRAMS OF LABELED AND UNLABELED SAPPΒ SPECIFIC PRODUCT IONS FROM A MEDIA

STANDARD SAMPLE. ............................................................................................................................................. 64 FIGURE 4.3. MASS SPECTRA OF LABELED AND UNLABELED SAPPΒ-SPECIFIC B- AND Y- PRODUCT IONS FROM A CHO

MEDIA STANDARD SAMPLE .................................................................................................................................. 65 FIGURE 4.4. CHROMATOGRAPHY ELUTION PROFILES OF LABELED AND UNLABELED SAPPΑ TRYPTIC PEPTIDES AS SEEN

BY PEAKS FROM A CHO MEDIA STANDARD. ......................................................................................................... 66 FIGURE 4.5. MASS SPECTRA OF LABELED AND UNLABELED SAPPΑ-SPECIFIC B- AND Y- PRODUCT IONS FROM A CHO

MEDIA STANDARD SAMPLE .................................................................................................................................. 67 FIGURE 4.7. CHROMATOGRAPHY ELUTION PROFILES OF LABELED AND UNLABELED SAPPΑ AND SAPPΒ TRYPTIC

PEPTIDES AS SEEN BY PEAKS FROM A HUMAN CSF SAMPLE. ................................................................................ 68 FIGURE 4.8. MASS SPECTRA OF LABELED (FOREFRONT) AND UNLABELED (BACKGROUND) SAPPΒ-SPECIFIC B- AND Y-

PRODUCT IONS FROM A HUMAN CSF SAMPLE. ..................................................................................................... 69 FIGURE 4.9. MASS SPECTRA OF LABELED (FOREFRONT) AND UNLABELED (BACKGROUND) SAPPΑ-SPECIFIC B- AND Y-

PRODUCT IONS FROM A HUMAN CSF SAMPLE ...................................................................................................... 69 FIGURE 4.10. LINEAR SAPPΒ-SPECIFIC STANDARD CURVE, USING MEDIA FROM CHO CELLS. ..................................... 70 FIGURE 4.11. LINEAR SAPPΑ-SPECIFIC STANDARD CURVE, USING MEDIA FROM CHO CELLS ...................................... 71 FIGURE 4.12. A HUMAN CSF LABELING CURVE FOR SAPPΒ. ....................................................................................... 72 FIGURE 4.13. A HUMAN CSF LABELING CURVE FOR SAPPΑ. ..................................................................................... 73 FIGURE 5.1. RHESUS MACAQUE APP SEQUENCE........................................................................................................... 83

Page 7: The Processing of Amyloid Precursor Protein in the Central

v

FIGURE 5.2. RHESUS MACAQUE SAMPLE PROCESSING STUDY DESIGN. BLOOD AND CSF ARE COLLECTED FROM

RHESUS MACAQUES FOR BIOCHEMICAL ASSESSMENTS.. ....................................................................................... 85 FIGURE 5.2. CNS METABOLISM OF APP METABOLITES IN VEHICLE-TREATED RHESUS MONKEYS ................................ 88 FIGURE 5.4 EFFECTS OF A BACE1 INHIBITOR ON SILK LABELING, ELISA CONCENTRATIONS, AND NEWLY

GENERATED AΒ (A-C), SAPPΒ (D-F), AND SAPPΑ (G-I) IN CNS OF RHESUS MONKEYS.. .................................... 90 FIGURE 5.5. EFFECTS OF BACE1 INHIBITION ON APP METABOLITES’ AUC-1-57. .......................................................... 92 FIGURE 6.1. MODELS OF CHANGES IN APP PROCESSING IN AD...…………………………………………………...103 FIGURE 6.2. AMYLOID PRECURSOR PROTEIN PROCESSING & TRAFFICKING SCHEMATIC.. ......................................... 109

Page 8: The Processing of Amyloid Precursor Protein in the Central

vi

List of Tables

TABLE 1.1. AMYLOID PRECURSOR PROTEIN PROTEOLYTIC PRODUCTS AND SECRETASES REQUIRED FOR CLEAVAGES. .. 7 TABLE 2.1. COMPARISON OF COSINOR PARAMETERS FOR SAPPΑ AMONG 3 PARTICIPANT GROUPS. ............................ 31 TABLE 2.2. COMPARISON OF COSINOR PARAMETERS FOR SAPPΒ AMONG 3 PARTICIPANT GROUPS ............................. 31 TABLE 2.3. COMPARISON OF COSINOR PARAMETERS FOR AΒ40 AMONG 3 PARTICIPANT GROUPS. ................................ 32 TABLE 2.4. COMPARISON OF COSINOR PARAMETERS FOR AΒ42 AMONG 3 PARTICIPANT GROUPS. ................................ 32 TABLE 5.1. IN VITRO PHARMACOLOGICAL PROFILE OF THE BACE INHIBITOR, MBI-5. ................................................ 78 TABLE 5.2. EFFECTS OF BACE1 INHIBITION ON APP METABOLITES’ AUC-1-57 FROM SILK AND ELISA

ASSAYS……....93

Page 9: The Processing of Amyloid Precursor Protein in the Central

vii

List of Abbreviations

Aβ Amyloid-β

ACN Acetonitrile

AD Alzheimer’s disease

ADAD Autosomal Dominant Alzheimer’s Disease

ADAM10 A Disintegrin and metalloproteinase domain-containing protein 10

ADRC Alzheimer’s Disease Research Center

AICD APP Intracellular Domain

AmBic Ammonium Bicarbonate (NH4HCO3)

ANOVA Analysis of Variance

ApoE Apolipoprotein E

APP Amyloid Precursor Protein

AUC Area Under the Curve

BACE1 Beta-site APP Cleaving Enzyme 1

BACE2 Beta-site APP Cleaving Enzyme 2

BCA Bicinchoninic Acid

BSA Bovine Serum Albumin

CID Collision-Induced Dissociation

CDR Clinical Dementia Rating

CHO Chinese Hamster Ovary

CMP Cisterna Magna Ported

CNBr Cyanogen Bromide

CNS Central Nervous System

CSF Cerebrospinal Fluid

CTF83 C-terminal Fragment 83

Page 10: The Processing of Amyloid Precursor Protein in the Central

viii

CTF99 C-terminal Fragment 99

CV Coefficient of Variation

Da Dalton

DMEM Dulbecco's Modified Eagle Medium

DS Down Syndrome

DTT Dithiothreitol

ECF Extracellular Fluid

ELISA Enzyme-Linked Immuno Sorbent Assay

ESI Electrospray Ionization

ER Endoplasmic Reticulum

ETD Electron-Transfer Dissociation

FBS Fetal Bovine Serum

FSR Fractional Synthesis Rate

FCR Fractional Clearance Rate

FRET Fluorescence Resonance Energy Transfer

FTR Fractional Turnover Rate

GC-MS Gas Chromatography-Mass Spectrometry

GCRC General Clinical Research Center

HPLC High Performance Liquid Chromatography

h Hour(s)

IP Immunoprecipitation

ISF Interstitial fluid

kDa Kilodalton

KPI Kunitz-type serine protease inhibitor

LC-MS Liquid Chromatography-Mass Spectrometry

Page 11: The Processing of Amyloid Precursor Protein in the Central

ix

Leu Leucine

LTQ Linear Trap Quadrupole (Mass Spectrometer)

MCBP Mean Cortical Binding Potential

MCI Mild Cognitive Impairment

MFL Mole Fraction Labeled

min minute(s)

mM millimolar

mol Mole

m/z Mass-to-Charge Ratio

NHP Non-human Primate

OD Optical Density

PBS Phosphate Buffered Saline

PBS-T Phosphate Buffered Saline - 0.05% Tween20

PD Pharmacodynamic

PET Positron Emission Tomography

PIB Pittsburgh compound B

PK Pharmacokinetic

PSEN1 Presenilin 1

PSEN2 Presenilin 2

RP Reversed Phase

RT Room Temperature

sAPP(s) Soluble Amyloid Precursor Protein(s)

sAPPα Soluble Amyloid Precursor Protein Alpha

sAPPβ Soluble Amyloid Precursor Protein Beta

SD Standard Deviation

Page 12: The Processing of Amyloid Precursor Protein in the Central

x

sec second(s)

SEM Standard Error of the Mean

SILK Stable Isotope Labeling Kinetics

t½ Half-life

TIC Total Ion Current

TGN Trans-Golgi network

TTR Tracer to Tracee Ratio

YNC Young Normal (Healthy) Control

Page 13: The Processing of Amyloid Precursor Protein in the Central

xi

Acknowledgments

I am very thankful for the opportunities that I have been given and the people I have worked with during

my graduate school career. Without each of them, it would have been difficult, and in some cases,

impossible, to reach this final step.

Firstly, I must thank my mentor, Randy Bateman, who really went above and beyond to make sure I got

to this point. I knew I had wanted to do my doctoral thesis work in his laboratory from the first day of my

rotation. Not only was he very welcoming, but in my first couple years in his laboratory, he would spend

many hours mentoring me every week. Our hourly one-on-one meetings would frequently turn into 2-3

hour meetings during which he’d teach me everything he knew about the workings of a mass spec, among

other topics. I am grateful when I recall how he came in to the lab on several occasions during his

Christmas vacation in order to teach me how to independently run the LTQ. He has been very

accommodating, making it clear that I may contact him even in the evenings or on weekends if I am at all

in need of guidance in my scientific endeavors. Having such a wise mentor who is not only willing to

teach, but obviously enjoys it, is a blessing.

I appreciate all the time my thesis advisory committee has spent meeting with me during biannual

progress reports and one-on-one meetings. Their advice throughout my graduate career has been very

influential to getting where I am now. I’d like to extend a special thanks to my advisory committee chair,

Alison Goate, for spending additional time with me after each progress report presentation to give me

guidance on how to move forward.

Next, there are many past and present members of the Bateman lab that have been wonderful unofficial

mentors and great friends. Kristin Wildsmith, who joined the lab as a postdoc the same week I started my

rotation there, was a great role model and always looked out for me. I could go to her no matter what:

whether it be a scientific question or a personal issue. In addition to Randy: Kristin, Yuriy Pyatkivkyy,

Vitaliy Ovod, and Ling Munsell were all instrumental in teaching me all things mass spec related since I

Page 14: The Processing of Amyloid Precursor Protein in the Central

xii

was a little behind when I started off: I had joined the lab ever only having been a pure biologist and

having minimal chemistry background. I would especially like to express my gratitude to Ling as she

came out of retirement to help me jump-start the project outlined in Chapter 4 of this thesis. I benefited

greatly from her expertise in protein chemistry. Alex Wen and Tom Kasten were great mentors in all

things related to ELISAs and immunoprecipitations. Additionally, Tom has been a wonderful source for

any sort of trouble-shooting questions that I have had along the way. Yafei Huang has been a wonderful

tutor on diurnal regulation of proteins and cosinor analysis. Rose Connors was a great help with anything

related to cell culture or antibody-bead coupling. Anna Santacruz was indispensable when it came to

helping us work on the Alzheimer’s Association grant proposal in 2011. And, of course, thank you to

Wendy Sigurdson for being one of the most supportive people out there! Wendy joined the lab only a

few months after I did, so she has really been there for it all, through thick and thin. As is evidenced by

this long paragraph, Randy had, and continues to have, a great crew.

I would also like to thank some professional colleagues outside of the Bateman lab. For almost three

years, I have had the great opportunity to be involved in a collaboration with some incredible scientists at

Merck Research Laboratories, especially the lead on the Merck end of the collaboration, Mary Savage. I

am thankful to her and her team for many hours spent on teleconference calls mulling over the sometimes

confusing data with us. I would also like to give a huge thank you to Bruce Patterson, who has spent

many hours working out a non-steady state model for APP processing in the BACE1 inhibitor study, and

then many hours tutoring me on the basics of kinetic studies and modeling.

This thesis work was supported by grants from the US National Institutes of Health (NIH) grants K23 AG

03094601, R-01-NS065667, P50 AG05681-22, P01 AG03991-22, and RR000954, and the Ruth K. Broad

Biomedical Research Foundation. An Eli Lilly research grant provided antibodies used in the work

outlined in Chapters 2-4. A Merck research grant provided antibodies and financial support for the work

outlined in Chapter 5. I am grateful to the Clinical Core of the Knight Alzheimer’s Disease Research

Center for characterization of the older participants and to all the participants for their time and effort.

Page 15: The Processing of Amyloid Precursor Protein in the Central

xiii

And now I may move on to my friends: I had two wonderful roommates and great friends during my time

as a graduate student, Stacey O’Brien and Lucy Li. Thanks to them, and my friend, Graziella Mendonsa,

for being motivators and putting up with my moodiness throughout the years when experiments didn’t go

as planned and when projects that I had invested much time in had to be set aside due to things beyond

my control.

Thank you to my family. My mom and step-mom have always been very supportive, as has my dad, who

inspired me to pursue a Ph.D. Even as a grade-schooler, I wanted to be just like him.

And last, but certainly not least, I must thank my best friend and my partner in life, Mohammed Zakaria.

He has been and continues to be a constant source of encouragement throughout my entire graduate

school career. Mohammed is incessantly supportive of me aspiring toward ambitious professional goals.

Thank you for believing in me.

Justyna Anna Dobrowolska

Washington University in St. Louis

December 2013

Page 16: The Processing of Amyloid Precursor Protein in the Central

xiv

Abstract of the Dissertation

The Processing of Amyloid Precursor Protein in the

Central Nervous System of Humans and Rhesus Macaques

by

Justyna Anna Dobrowolska

Doctor of Philosophy in Neurosciences

Washington University in St. Louis, 2013

Professor Randall J. Bateman, Chair

Alzheimer’s disease (AD) is the most common dementia, currently afflicting 30 million people

worldwide, with prevalence steadily increasing. Over 99% of AD cases are classified as

sporadic, in which the major risk factors are age (greater than 65 years) and the ApoE-ε4 allele

(in a gene dose dependent manner). The minority of AD cases (less than 1%) are caused by

autosomal dominant inheritance of a genetic mutation in one of three genes: Amyloid Precursor

Protein (APP), Presenilin-1 (PSEN1), or Presenilin-2 (PSEN2). Mutation carriers will generally

notice cognitive decline starting at a relatively young age, anytime between their 30s-50s, and

eventually succumb to this early-onset AD. Early-onset AD may also be the result of an extra

copy of the APP gene, as manifested in Down Syndrome (DS) patients, who universally develop

AD.

Alzheimer’s disease pathophysiology appears to revolve around APP. This transmembrane

protein may be processed through one of two pathways. If APP is processed down the

amyloidogenic (β-secretase) pathway, soluble APPβ (sAPPβ) will be released first, and with a

subsequent cleavage, β-amyloid (Aβ), will be released into the brain where it could sequester

Page 17: The Processing of Amyloid Precursor Protein in the Central

xv

into amyloid plaques, which are a major hallmark of AD. The other processing pathway is

caused by cleavage of APP by α-secretase. This results in the release of soluble APPα (sAPPα),

which precludes the release of Aβ, thus making this pathway non-amyloidogenic.

Several studies have focused on measuring the products of APP cleavages in an effort to

elucidate the pathophysiology of AD. These studies give a static view of APP because they

measure absolute amounts of protein in single samples. However, proteins are constantly being

produced, cleared, and aggregated which may result in protein concentrations fluctuating over

time. Recently it was reported that Aβ exhibits a diurnal pattern, the definitive cause of which is

still unknown.

The general goal of this thesis is to determine the interplay of the α- and β-secretase APP

processing pathways in both physiological and pathophysiological settings in humans, to

establish the human metabolism rates of total sAPP, sAPPα, and sAPPβ, and to determine

whether drug intervention of one of the two APP pathways, would have an effect the other

pathway. The aims of this thesis are:

1) To determine the extent of interdependence of the α-secretase and the β-secretase APP

processing pathways in humans and whether Aβ diurnal patterns were being driven by APP.

Human cerebrospinal fluid (CSF) was collected hourly for 36 h, and concentrations of sAPPα,

sAPPβ, Aβ40, and Aβ42 were measured by four metabolite-specific ELISAs. Parameters

associated with diurnal patterns were compared among metabolites. Further, correlation

analyses were used to determine if any correlation among metabolites at a single time-point was

evident. Samples from Alzheimer’s patients, age-matched controls, and young healthy controls

were analyzed.

Page 18: The Processing of Amyloid Precursor Protein in the Central

xvi

2) To determine the physiological and pathophysiological metabolism of sAPPα and sAPPβ

in humans. Total sAPP was isolated by immunoprecipitation from CSF of participants

undergoing stable isotope labeling kinetics studies. Isolated sAPP underwent proteolytic

cleavage by trypsin and turnover rates of total sAPP were determined by LC-MS and compared

to turnover rates of Aβ. Isolated total sAPP that underwent proteolytic cleavage by Arginine-C

produced some preliminary results that could lead to eventual determination of sAPPα or sAPPβ

specific turnover rates in humans.

3) To determine the physiological metabolism of sAPPα and sAPPβ in rhesus macaques in

comparison to Aβ metabolism and to determine if drug intervention of the β-secretase pathway

by a BACE1 inhibitor would affect the α-secretase pathway. Soluble APPβ, sAPPα and Aβ were

isolated from rhesus macaque CSF by serial immunoprecipitation. Monkeys were undergoing

stable isotope labeling kinetics studies. Isolated APP metabolites underwent proteolytic cleavage

by LysN, and then the turnover rates of these metabolites were measured by LC-MS to determine

if the BACE1 inhibitor was hitting its target, and if it was having any effect of the α-secretase

pathway.

Page 19: The Processing of Amyloid Precursor Protein in the Central

1

Chapter 1 Introduction and Perspective

Alzheimer’s Disease as a Major Public Health Issue

Alzheimer’s Disease (AD) is a progressive, debilitating neurological disease that manifests itself

with symptoms of memory deficits and problems with thinking and behavior. It is the 6th leading

cause of death in the U.S.A. There is no effective preventative treatment, and no medical

intervention that will slow disease progression, halt it, or otherwise reverse its symptoms.

Therefore, AD is universally fatal. Of the leading causes of death in the U.S.A., only AD cases

have been on the rise. In the first decade of the 2000s, AD prevalence increased by 68%. It is

estimated that this year (2013) there are 5.2 million Americans afflicted with this disease, of

which approximately five million are over the age of 65. The U.S.A. is facing the stark reality of

a further rapid increase in AD diagnoses as the eldest of those born during the Post World War II

baby boom are now nearing 70 years of age. Estimates indicate that by the year 2025, the

prevalence of AD in people 65 years and older will reach 7.1 million. The direct cost of care for

all AD patients in the USA in 2013 is predicted to be $203 billion, and, unless new medical

breakthroughs become available, the costs will increase exponentially to an estimated $1.2

trillion per year by 2050 (Alzheimer’s Association, 2013). All this taken together indicates that

AD is clearly a major public health issue, and efforts to understand better AD pathophysiology,

as well as discover effective drug interventions need to be a priority.

Page 20: The Processing of Amyloid Precursor Protein in the Central

2

The Amyloid Hypothesis

In 1906, at a medical conference, a psychiatrist, Alois Alzheimer, first discussed his findings of a

clinicopathological disorder that later was named in his honor. Dr. Alzheimer’s description was

based on the analysis of a female patient named Auguste D., who manifested early-onset AD,

dying at the age of 56 years (Alzheimer, 1907). He documented her cognitive deficits, along

with describing two pathological hallmarks of this disease by studying Auguste D.’s brain after

death. The two classic brain lesions that Alzheimer’s described are still considered the hallmarks

of AD: the extracellular neuritic plaques composed of Amyloid-β, Aβ (predominantly Aβ42), and

the intracellular neurofibrillary tangles composed of hyperphosphorylated tau proteins.

However, the molecular pathophysiology of AD is still not fully understood, and decades of

biomedical research have helped to formulate the Amyloid Hypothesis (Hardy and Selkoe,

2002). The evidence supporting this hypothesis centers on Aβ and its precursor, Amyloid

Precursor Protein (APP), as key proteins implicated in AD pathophysiology. Additional data

that corroborate this hypothesis include but are not limited to the list herein:

Cognitively normal participants with low Aβ42 CSF concentrations are those that, when followed

longitudinally, tend to develop AD (Skoog et al., 2003; Gustafson et al., 2007; Stomrud et al.,

2007). There are three genes (APP, Presenilin-1 (PSEN1), and Presenilin-2 (PSEN2)) known to

have a number of potential mutations (50+, 150+, and 10+, respectively) that have been shown to

increase Aβ production and/or aggregation. Carriers of these mutations universally develop

autosomal-dominant AD (ADAD) (Bateman et al., 2011). Recent studies indicated that the

biomarker, clinical, and cognitive presentation of ADAD was remarkably similar to the later

onset sporadic AD (Bateman et al., 2012). The first ADAD mutation discovered was a missense

mutation in APP which increased the production of Aβ42, a longer form of Aβ more prone to

Page 21: The Processing of Amyloid Precursor Protein in the Central

3

aggregate (Goate et al., 1991). The discovery of mutations that cause AD revolutionized the

molecular biology studies of AD by providing definitive evidence that alterations in APP, likely

through Aβ processing, was sufficient to cause AD. Subsequent mutations were found in PSEN1

(Sherrington et al., 1995), and PSEN2 (Levy-Lahad et al., 1995). Interestingly, Auguste D., the

first patient described by Dr. Alzheimer, had a PSEN1 mutation as the likely cause of her

dementia (Müller et al., 2013).

Recently, Jonsson et al. (2012) characterized a coding mutation on the APP gene in an Icelandic

population. This mutation appeared to protect the elderly non-AD human participants from

cognitive decline. The mutation, A673T, was found to decrease Aβ in both cell culture assays

and animal models.

Conclusions from the study of Down Syndrome (DS) patients, who universally develop classic

AD pathology by 35 years of age (Malamud 1972; Wisniewski et al., 1985), were consistent with

the Amyloid Hypothesis. Patients with DS are born with an extra copy of chromosome 21

(Hassold et al., 1993), which is the chromosome on which the gene encoding APP is located.

Down Syndrome patients have 50% more total APP than their healthy human counterparts.

Those with DS have been shown to overproduce APP and Aβ and have amyloid plaques present

in the brain as early as 12 years of age. These plaques precede other pathological hallmarks of

AD, such as neurofibrillary tangles (Lemere et al., 1996; Querfurth et al., 1995).

The type 4 allele of the apolipoprotein E gene, ApoE-ε4, is the most common genetic risk factor

for sporadic AD and has been shown to alter Aβ deposition. In a gene dose dependent manner,

ApoE-ε4 increases both the amount of fibrillar amyloid deposition in the brain (Morris et al.,

2010) and the probability of developing late-onset AD (Corder et al., 1993).

Page 22: The Processing of Amyloid Precursor Protein in the Central

4

Amyloid Precursor Protein Expression

Amyloid Precursor Protein is a ubiquitous transmembrane protein that ranges between 110 and

140 kDa when measured by electrophoresis (Selkoe et al, 1982). The differences in protein size

may be attributed to several factors, such as several potential splice variants and post-

translational modifications. APP has three major isoforms that are 695, 751, or 770 amino acid

residues in length. Neuronal cells predominantly express APP695, and this splice variant is found

in non-neuronal tissue to a very low degree (Haass et al., 1991). Both APP751 and APP770 do

occur in neurons but to a lesser extent than APP695. These two longer APP variants are widely

expressed in non-neuronal tissue. APP695 lacks an exon coding for the 56 amino acid sequence

homologous to the so-called Kunitz-type of serine protease inhibitors (KPI). Post-translational

modifications of APP include N- and O-linked sugar additions and sulfation or phosphorylation

(Hung and Selkoe, 1994; Oltersdorf et al, 1990; Walter et al., 1997; Weidemann et al., 1989).

Amyloid Precursor Protein Localization

Amyloid Precursor Protein is localized in the membranes of the cell, the endoplasmic reticulum

(ER), Golgi apparatus, and endosomes (Koo et al., 1996; Yamazaki et al., 1996; Skovronsky et

al., 2000). The N-terminus of APP is situated on the exterior of the cell or the interior of an

organelle membrane. Amyloid Precursor Protein is cotranslationally translocated into the ER,

and later it becomes mature after posttranslational modifications occurring in the secretory

pathway (here APP obtains the N- and O- sugars mentioned in the previous section). During and

after trafficking through the secretory pathway, APP may undergo post-translational processing

through one of two pathways: the α-secretase (non-amyloidogenic) pathway and the β-secretase

Page 23: The Processing of Amyloid Precursor Protein in the Central

5

(amyloidogenic) pathway (Figure 1.1., Table 1.1.). The relationship between these two

processing pathways of APP in the human CNS is not fully understood.

Proteolytic Processing of Amyloid Precursor Protein

Figure 1.1. Proteolytic processing of Amyloid Precursor Protein. Amyloid Precursor Protein may be processed through the non-amyloidogenic (α-secretase) pathway or the amyloidogenic (β-secretase) pathway.

It is through the amyloidogenic pathway that APP can be processed in two steps to result in the

production of Aβ. In the first step, APP is cleaved by β-secretase (the major neuronal β-

secretase is β-site APP-cleaving enzyme 1, or BACE1) in the ectodomain (C-terminal to amino

acid 671). The soluble fragment resulting from this cleavage is the 671 amino acid sAPPβ.

Owing to this cleavage, the remaining APP ectodomain (C-terminal Fragment (CTF) 99) now

Page 24: The Processing of Amyloid Precursor Protein in the Central

6

contains the first amino acid sequence of Aβ as its N-terminus. It is then available to be

subsequently cleaved in the endodomain by γ-secretase. The result of this cleavage is the release

of Aβ into the intracellular compartment’s lumen or into the extracellular fluid (ECF) (Busciglio

et al., 1993) while the APP fragment that remains in the membrane is the APP Intracellular

Domain (AICD). The processing of APP through the β-secretase pathway occurs predominantly

in endosomes, where the acidic environment is favorable for BACE activity. γ-secretase also

becomes very active in the late endosomes. Therefore, sAPPβ and Aβ are released into the ECF

when either early endosomes are recycled to the cell surface or when late endosomes fuse with

the cell membrane. Conversely late endosomes may fuse with lysosomes and the sAPPβ and Aβ

will be degraded.

During the processing through the non-amyloidogenic pathway, APP first undergoes cleavage by

α-secretase, producing a soluble product known as sAPPα and the remaining APP

transmembrane CTF 83. Cleavage at this site precludes the formation and release of Aβ because

this site is located between the 16th and 17th amino acids of Aβ. Therefore, when γ-secretase

subsequently cleaves the APP endodomain, the resulting soluble fragment, p3, is missing the first

16 amino acids that are included in the Aβ sequence. As in the amyloidogenic pathway, AICD is

the APP fragment remaining in the membrane. The p3 fragment has not been reported to self

aggregate as does Aβ. In addition to preventing the release of Aβ, some evidence indicates that

this pathway may be neuroprotective in its own right. The C-terminus of sAPPα has been shown

to protect neurons against glutamate-induced toxicity, glucose deprivation, and Aβ toxicity,

among others (Furukawa et al., 1996; Goodman et al., 1994; Smith-Swintosky et al., 1994).

Furthermore, some studies have demonstrated sAPPα’s connection to improved memory in

Page 25: The Processing of Amyloid Precursor Protein in the Central

7

rodents. Processing of APP through the α-secretase pathway occurs predominantly at the cell

surface (Parvathy et al., 1999) thus sAPPα is mainly released directly into the ECF.

The α- and β- APP processing pathways may be completely independent of one other, in which

case an increase or decrease of APP processing through one pathway would have no effect on the

other pathway. However, it is possible that the two pathways are interrelated to some extent.

For example, if there is an increase in APP processing through the α-pathway, there will be a

proportional decrease in the amount of APP processed through the β-pathway. It is essential to

know how the two pathways relate to one another because this will help inform drug

development efforts as to the related effects of decreasing Aβ on increasing the α-secretase

pathway. In addition, knowledge of the basic physiology of these secretase systems in the

human CNS is important to understand the pathophysiology of AD and may provide insight into

the function of the APP in the CNS.

Table 1.1. Amyloid Precursor Protein proteolytic products and secretases required for cleavages.

Page 26: The Processing of Amyloid Precursor Protein in the Central

8

Trafficking of Amyloid Precursor Protein

The trafficking of APP is not straight forward. The translated APP may be trafficked from the

ER to the cell membrane in the secretory pathway or it may be trafficked to the Golgi and then to

late endosomes or lysosomes. When APP reaches the cell membrane, if it is uncleaved by α- and

γ-secretases (both active at this location), it may be reinternalized into early endosomes where it

may be cleaved sequentially by β- and γ-secretases. There is much evidence that the β-secretase

processing pathway of APP occurs in the endocytic pathway. Endocytosis of APP appears to be

necessary for Aβ production. Its production is decreased, while sAPPα and cell surface APP

increases, when endocytosis is experimentally inhibited by several different methods (Koo and

Squazzo, 1994). As mentioned above, BACE is localized to cellular compartments that have an

acidic pH (Yu et al., 2000; Haass et al., 1995). The γ-secretase complex, which is responsible

for the final cleavage of APP that releases Aβ, has also been reported to be not only localized at

the cell surface, but also throughout intracellular compartments, such as the ER, TGN,

endosomes, and lysosomes (Ray et al., 1999; Pasternak et al., 2003; Cupers et al., 2001). Taken

together, it is not surprising that Aβ secretion is increased in the endocytic pathway. Aβ may be

released into the ECF through recycling endosomes merging with the cell membrane or it may

travel further down the endocytic pathway to the late endosomes where its fate is either release

into the ECF or degradation if the late endosome fuses with a lysosome. Amyloid Precursor

Protein may also remain uncleaved by α- or β-secretase throughout its trafficking. In this case, it

will be degraded in the lysosome as an intact protein, without forming any APP metabolites.

Page 27: The Processing of Amyloid Precursor Protein in the Central

9

Diurnal patterns of Amyloid-β

The first temporal studies of Aβ concentrations in humans were reported in 2007, indicating that

CSF concentrations of Aβ40 and Aβ42 were sinusoidal over 24 h in younger healthy participants

(Bateman et al., 2007), suggesting a potentially circadian pattern (Figure 1.2.).

Figure 1.2. Diurnal fluctuations of Aβ observed in humans (Bateman et al., 2007)

Subsequent studies in humans and animal models (Kang et al., 2009) demonstrated that diurnal

rhythms of Aβ concentrations in the brain were affected by disruptions of both the sleep-wake

cycle and the signaling of orexin, a neuropeptide that regulates several physiological functions,

including wakefulness (Sakurai 2007). Recently our group reported that Aβ exhibits a diurnal

Page 28: The Processing of Amyloid Precursor Protein in the Central

10

pattern in both CSF (Huang et al., 2012a) and blood (Huang et al., 2012b) in healthy human

adults, whereas the levels of total proteins do not exhibit any significant circadian pattern. The

Aβ diurnal patterns measured, as determined by circadian amplitude, were decreased with aging

and amyloidosis. This decrease coincided with observed disturbances in sleep with age (Redline

et al., 2004) and neurodegeneration (Gagnon et al., 2008), and therefore may be ultimately linked

to the sleep-wake cycle and orexin signaling. However, the immediate mechanism for diurnal

regulation of Aβ has not been described, and possible causes for the Aβ diurnal pattern include

diurnal regulation of APP transcription, translation, or transport, or diurnal regulation affecting

the two secretases (β-secretase or γ-secretase) that cleave APP to produce Aβ.

Elevated β-secretase in AD

Elevated BACE1 expression and activity have been reported in post-mortem human AD brains.

In living human participants, BACE1 activity in the CSF positively correlates with fibrillar

amyloid pathology, as measured by carbon-11-labeled Pittsburgh Compound B positron

emission tomography ([11C] PIB PET) (Grimmer et al., 2012). Some results indicate that BACE

activity was slightly higher in CSF of AD participants than in controls (Fukumoto et al., 2002;

Johnston et al., 2005; Holsinger et al., 2006; Holsinger et al., 2004; Verheijen et al., 2006). In

another study, participants with Mild Cognitive Impairment (MCI) had both higher BACE

activity and protein levels (Zhong et al., 2007). Another group (Zetterberg et al., 2008)

demonstrated that sAPPα and sAPPβ were highly positively correlated in MCI and AD

participants. They also showed that, except in the MCI group that progressed to AD, BACE1

activity positively correlated with sAPPα, sAPPβ, and Aβ40. Owing to the elevated β-secretase

Page 29: The Processing of Amyloid Precursor Protein in the Central

11

activity, APP processing may be directly affected in AD to the amyloidogenic pathway, thus

making BACE inhibitors attractive therapeutics for this disease.

In vivo Stable Isotope Labeling and the LC/MS approach

In most cases, absolute amounts or concentrations of proteins can be detected by different

methods available to researchers using standard curves (e.g. Western Blot, ELISA,

immunoassays). Absolute protein amounts and concentrations are the result of the protein’s

metabolism (production and clearance rates). However, protein quantitation does not inform

about the protein’s kinetics or metabolism. To determine why the changes of protein level are

occurring, protein kinetics need to be measured. Is it because there is an overproduction or

underproduction of a protein? Is the protein being cleared too slowly or too quickly? Or is it a

combination of the two factors? In vivo stable isotope labeling has been utilized to address these

questions in other systems. The administration of a stable-isotope labeled amino acid into an in

vitro or in vivo system provides a method to label proteins that are being produced at a specific

time and track the newly produced proteins kinetics including production and clearance. By

measuring the ratio of labeled to unlabeled proteins the assessment of production and clearance

rates of a specified protein can be made. Mass spectrometry can be utilized to quantify isotope

labeled and unlabeled peptides and proteins. By measuring labeled proteins over time during

and after labeling, kinetic curves can be obtained and used for calculation of kinetic parameters

and model fits. The kinetic measurements then inform about the rates of processing, thus adding

the time dimension to analysis of protein physiology.

Page 30: The Processing of Amyloid Precursor Protein in the Central

12

Chapter 2 Diurnal Regulation of Amyloid Precursor Protein in Humans

Introduction

Although the pathophysiology of AD is incompletely understood, the study of brain and

cerebrospinal fluid (CSF) proteins, such as amyloid-β (Aβ) and tau, have provided insight into

AD molecular pathophysiology (Vanderstichele et al. 1998; Galasko et al. 1998; Andreasen et al

1999; Fagan et al. 2007; Holtzman et al. 2011). The study of Aβ production, transport and

clearance is important for insight into normal brain protein handling and also for the

pathophysiology of AD.

The first human studies of Aβ concentrations over time indicated that CSF concentrations were

sinusoidal over 24 h in younger healthy participants (Bateman et al., 2007) and suggested a

possible circadian pattern. Subsequent studies in humans and animal models (Kang et al., 2009)

demonstrated Aβ concentrations in the brain could be regulated by sleep/wake cycles and orexin

administration. Recently, it was reported that Aβ exhibits a diurnal pattern in both CSF (Huang

et al., 2012a) and blood (Huang et al., 2012b) in healthy adults. The diurnal patterns, as

determined by circadian amplitude, decreased with aging and amyloidosis. The immediate

mechanism for diurnal regulation of Aβ has not been described, and possible causes for the Aβ

diurnal pattern include, but are not limited to, diurnal regulation of APP transcription,

translation, or transport, or diurnal regulation affecting the two secretases (β-secretase or γ-

secretase) that cleave APP to produce Aβ. In this study, we evaluated the temporal relationship

of Aβ with other proteolytic products of APP to try to determine the cause of Aβ diurnal patterns

in the CNS of healthy young and elderly humans, as well as those with amyloid pathology.

Page 31: The Processing of Amyloid Precursor Protein in the Central

13

Amyloid Precursor Protein is a single-pass transmembrane protein processed through at least two

pathways in the CNS: the β- (amyloidogenic) pathway and the α- (non-amyloidogenic) pathway

(Zheng and Koo 2011). It is cleaved in the amyloidogenic pathway by β-secretase releasing a

soluble extracellular fragment called soluble APPβ (sAPPβ). The APP endodomain, C-terminal

fragment 99 (CTF99), which remains in the transmembrane, is subsequently cleaved by gamma

(γ)-secretase, resulting in the generation of Aβ and an APP Intra-Cellular Domain. The non-

amyloidogenic processing of APP occurs when α-secretase cuts APP, producing soluble APPα

(sAPPα). The endodomain of APP (CTF83) may then be cut by γ-secretase, resulting in the

release of a fragment, p3. The formation of Aβ is precluded by α-secretase cleavage.

To elucidate further the potential contributions of APP to the Aβ diurnal pattern and the balance

of the α- and β- pathways in APP processing, we measured the CSF APP proteolytic products

sAPPβ, sAPPα, and Aβ40 and Aβ42 over 36 h from cognitively normal young and elderly, plus

AD participants.

Experimental Methods and Study Design

Participants and Sample Collection.

All human studies were approved by the Washington University Human Studies Committee and

the General Clinical Research Center (GCRC) Advisory Committee. Young Normal Control

(YNC) participants were recruited from the general public. Older participants that would later be

assigned to the Amyloid- or Amyloid+ groups were recruited through the Charles F. and Joanne

Knight Alzheimer’s Disease Research Center (Knight ADRC) at Washington University.

Informed consent was obtained from all participants prior to their enrollment in the study.

Participants were in good general physical health. They had no clinical neurological diseases.

Page 32: The Processing of Amyloid Precursor Protein in the Central

14

Exclusion criteria for the study included having an active infection, a bleeding disorder, or

treatment with anticoagulants. These participants were divided into three groups by age and

brain amyloid status: 1) an Amyloid+ group of participants greater than 60 years of age and with

probable amyloid plaques in the brain, as measured by positron emission tomography using

Pittsburgh compound B (PET PiB) or determined by an Aβ42 CSF mean concentration less than

350 pg/mL; 2) an Amyloid- age-matched group that had no detectable amyloid plaques in the

brain as measured by PET PiB or determined by an Aβ42 CSF mean concentration greater than

350 pg/mL; 3) a young normal control (YNC) group (18-50 years of age) who are likely PET

PiB- (Mintun et al. 2006). PiB binds to fibrillar amyloid plaques in the brain (Klunk et al.,

2004). A mean cortical binding potential (MCBP) was calculated for each participant to

determine PiB (Amyloid) “+” or “–” status (Mintun et al. 2006). To measure the MCBP, binding

potentials of PiB were averaged from specific brain regions: prefrontal cortex, precuneus, lateral

temporal cortex, and gyrus rectus. The MCBP scores that were 0.18 or greater were designated

as amyloid plaque positive (Amyloid+), whereas those less than 0.18 were designated as amyloid

plaque negative (Amyloid-) (Mintun et al. 2006). Some participants did not have reported

MCBP values, and, in those cases, a surrogate marker of amyloid deposition was used to assign

the participant group. This surrogate marker was a low CSF Aβ42 concentration which has been

shown to be inversely correlated with PET PiB measurements (Fagan et al., 2006). A CSF Aβ42

concentration was considered low (and Amyloid+) if less than 350 pg/mL from an Aβ42 ELISA

that used 21F12 (anti-Aβ42) as the capture antibody and biotinylated 3D6 antibody (directed

against Aβ1-5) as the detection antibody.

Page 33: The Processing of Amyloid Precursor Protein in the Central

15

Demographics of Study Participants.

A total of 49 participants (both men and women) were assessed in at least one part of this study.

Specific sample size in each group varied depending on the experiment, and sample size for each

group when diurnal patterns were observed is listed in the cosinor analyses section of the

Methods. For the part of this study where APP metabolites were measured in a single CSF time

point (results shown in Figures 2.12.-2.13.) there were 15 participants in the YNC group, 15 in

Amyloid-, and 18 in Amyloid+.

The mean (SD) age for each participant group when all 49 participants were taken into account:

YNC = 37.1 (± 8.7) years; Amyloid- = 69.6 (± 4.5) years; and Amyloid+: 76.3 (±7.5) years.

Clinical Dementia Rating (CDR) at study onset was available for all participants. Of the

Amyloid- participants, 33.3% had a CDR score greater than zero (exhibited cognitive deficits).

Of the Amyloid+ participants, 29.4% had a CDR score equal to zero. All YNC subjects were

free from any cognitive deficits.

Sample Collection and Storage.

Sample collection and handling were done as previously described (Bateman et al., 2007).

Briefly, for all participants, an intrathecal lumbar catheter was placed between the L3 and L4

interspace or the L4 and L5 interspace between 7:30AM-9:00AM. Collection of CSF began

between 8:00AM and 9:30AM. Every hour for 36 h, 6 mL of CSF and 12 mL of plasma were

withdrawn. Aliquots of CSF (1 mL) were immediately frozen at -80 °C in Axygen maximum-

recovery polypropylene tubes.

Sample and Standard Handling.

Aliquots (1mL) from even hours with two freeze-thaw cycles were measured by sAPPα and

sAPPβ ELISA. The effect of two freeze-thaw cycles was determined to change sAPPα (Figure

Page 34: The Processing of Amyloid Precursor Protein in the Central

16

2.1) and sAPPβ concentrations (data not shown) only marginally. Before plating, CSF samples

were diluted in phosphate buffered saline-0.05% Tween20 (PBS-T) 75- to 150-fold for sAPPα,

and 10- to 25-fold for sAPPβ. Recombinant standards from E.coli were used for both sAPPα

(Sigma S9564-25UG) and sAPPβ (S4316-25UG). The concentration of the standards ranged

from 1.6 - 75 ng/mL for sAPPα and 2.7 - 125 ng/mL for sAPPβ. Each CSF and standard sample

was assessed in triplicate.

Figure 2.1 Effect of two Freeze/Thaw cycles on CSF concentrations of sAPPα. Owing to the CSF being accessed by multiple users, some patients’ CSF samples may have possibly undergone one freeze-thaw cycle prior to being used in this study. Thus, to be consistent, all samples would undergo 2 freeze-thaw cycles. To determine whether an extra freeze/thaw cycle would decrease CSF concentrations of sAPPα, the developed ELISA was used to test samples from the same participant, with one having undergone one freeze/thaw cycle, and the other having undergone two cycles. As represented above, an extra freeze/thaw cycle has no significant effect on sAPPα concentration in CSF.

Page 35: The Processing of Amyloid Precursor Protein in the Central

17

Soluble APPα ELISA Protocol.

For the sAPPα ELISAs, 96-well Nunc MaxiSorp flat bottom ELISA plates (eBiosciences, Inc.;

San Diego, CA) were coated with 100 µL per well of 5 µg/mL of 8E5 (a monoclonal antibody

raised to a bacterially expressed fusion protein corresponding to human APP444-592 of the APP770

transcript (Games et al., 1995), courtesy of Eli Lilly). Plates were incubated for 24 h on a shaker

at 4 °C, and then blocked with 3% dry milk in PBS-T for 1 h 20 min at 37 °C. To avoid plate

position effects, samples were randomly assigned to a well on the plate. Secondary (detection)

antibody (50 µL of 1:10,000 6E10-biotin (Kim et al., 1988), a monoclonal antibody reactive to

Aβ1-16, otherwise known as APP672-687 (in the APP770 transcript), and having the epitope at Aβ3-

8, or APP674-679) (Signet Covance; Dedham, MA) was added to each well. Samples and

secondary antibody were incubated on a shaker at 4 °C for 24 h. Plates were washed 5 times

with PBS-T and then poly-HRP20-Strepavidin (Fitzgerald Industries International; Acton, MA)

diluted at 1:15,000 in 1% bovine serum albumin (Sigma-Aldrich; St. Louis, MO) in PBS-T was

added to each well at 100µL/well. Plates were incubated in the dark for 1 h at 37 °C on a shaker.

Plates were then washed 5 times with PBS-T and 5 times with PBS. The plates were then

developed as described for the sAPPβ ELISA below.

Soluble APPβ ELISA Protocol.

For the sAPPβ ELISA, 96-well Nunc MaxiSorp flat bottom ELISA plates (eBiosciences, Inc.;

San Diego, CA) were coated with 100 µL per well of 10 µg/mL of the monoclonal antibody,

8E5. Plates were incubated for 24 h on a shaker at 4 °C, and subsequently blocked with 3% dry

milk in PBS-T for 1 h 20 min at 37 °C. Samples were randomly assigned a plate-well position

and incubated for 24 h on a shaker at 4 °C. They were then washed 5 times with PBS-T. A

biotinylated antibody against the neo-epitope (amino acid sequence: KM) of sAPPβ (courtesy of

Page 36: The Processing of Amyloid Precursor Protein in the Central

18

Eli Lilly) was used as the secondary (detection) antibody at a volume of 50 µL and a

concentration of 0.5 µg/µL, diluted in 37 °C PBS-T. The sAPPβ-biotin detection antibody was

added to each well and incubated at 37 °C for 1.5 h. Plates were washed 10 times with PBS-T

and 100 µL poly-HRP40-Strepavidin (Fitzgerald Industries International; Acton, MA), diluted at

1:20,000 in 1% bovine serum albumin in PBS-T, was added to each well. Plates were incubated

in the dark for 1 h at 25 °C on a shaker and washed 5 times with PBS-T and 5 times with PBS.

For both the sAPPα and sAPPβ ELISAs, 100 μL per well of ELISA TMB Super Slow (Sigma-

Aldrich; St. Louis, MO), pre-warmed to 25 °C, was then added to each well. Optical density

(OD) was measured at 650 nm by using a Biotek Synergy 2 plate reader.

Amyloid-β40 and Aβ42 ELISA Protocols.

Corning 96-well half area clear flat bottom polystyrene high bind ELISA plates (Corning Life

Sciences, Tewksbury, MA) were coated with 1.25 µg/mL HJ7.4 (Aβ37-42) or 2.5 µg/mL HJ2

(Aβ33-40) in PBS plus 20% glycerol (PBS-G), then incubated 1 h at 25 °C followed by overnight

incubation at 4 °C. The next day the plates were blocked with 2% BSA-PBS-T for 1.5 h at 4 °C.

Single freeze-thaw CSF aliquots from both odd and even hours were thawed on ice. They were

diluted in a final buffer consisting of 2 mg/mL BSA-PBS-T, 3M Tris, 10% Azide, 1X protease

inhibitor cocktail. Samples were randomly assigned a well on the plate. Diluted CSF samples

and standards were pipetted at a volume of 50 µL per well onto freshly washed plates. The

samples were loaded in triplicate and incubated overnight at 4 °C. After incubation and washing,

the plates were incubated with HJ5.1-Biotin (Aβ13-28) 0.2 µg/mL in 1% BSA-PBS-TG for 1.5 h

at 25 °C. Plates were then washed three times with 190 µL PBS-T, followed by incubation in

Strep-poly-HRP40 (RDI) 1:12,000 in 1% BSA-PBS-TG for 1.5 h at 25 °C. Plates were

subsequently washed three times with 190 µL PBS-T. They were then incubated with 50

Page 37: The Processing of Amyloid Precursor Protein in the Central

19

µL/well of Slow ELISA TMB (pre-warmed to 25 °C) for 5-30 min. Optical density (OD) was

read at 650 nm by using a Biotek Synergy 2 plate reader.

Quantification of Metabolites’ Concentrations.

Soluble APPα, sAPPβ, Aβ40, and Aβ42 concentration levels were quantified by using the Biotek

Gen5 software (version #1.08.4) based on the non-linear five parametric standard curves

generated from the recombinant sAPPα, sAPPβ, Aβ40, and Aβ42 standards. The OD values of

the CSF samples fell within the linear range of the standard curve and were converted to

concentration levels. The product of the concentration and the dilution factor was calculated to

determine the final CSF concentration of each protein.

Quantification of Total Protein Concentrations.

Total protein levels of each sample were measured by using a micro BCA protein assay kit

(Thermo Fisher Scientific, Inc.; Rockford, IL), as previously reported (Huang et al., 2012a). The

intra-sample coefficient of variation was 2% for duplicates.

Grouped Cosinor Analysis.

Serial sAPPα and sAPPβ concentrations were binned in 2 h increments as measures were made

from every other hour. Serial Aβ40 and Aβ42 concentrations were left unbinned because hourly

concentrations were measured. For each APP metabolite, each participant’s hourly metabolite’s

concentration was normalized to that metabolite’s mean concentration over 36 h. The

normalized value was calculated as a percentage of the participant’s mean (100*value/mean).

Hourly (Aβ40, and Aβ42) and bi-hourly (sAPPα and sAPPβ) concentrations of each metabolite

were averaged among all participants in each participant group to produce normalized mean 36 h

concentrations. Next, the linear rise over time that was observed for all metabolites was

Page 38: The Processing of Amyloid Precursor Protein in the Central

20

subtracted from the mean concentrations, as previously described (Huang et al, 2012a). A single

cosinor analysis was performed for each metabolite, as described previously (Huang et al.,

2012a). Briefly, a cosine transformation was applied to the time variable by using 24 h as the

default circadian cycle, and Graphpad Prism version 5.01 for Windows (GraphPad Software; San

Diego, CA) was used to estimate the parameters of the circadian patterns for each metabolite’s

fluctuations. Mesor (midline of the metabolite oscillation), amplitude (distance between the peak

and mesor), and amplitude-to-mesor ratio were calculated for each participant group. Student t-

tests and ANOVAs were used to determine whether there were differences in these parameters

between groups. Analyses were performed in GraphPad Prism v. 5.01.

Individual Cosinor Analysis.

For each participant, sAPPα, sAPPβ, Aβ40, and Aβ42 levels over 36 h were analyzed by using a

single cosinor analysis as described in the Grouped Cosinor Analysis section of Experimental

Methods and Study Design. Mesor, amplitude, and amplitude-to-mesor ratio were calculated for

each metabolite and each participant. Participant group averages of each parameter were taken.

Student t-tests and ANOVAs were used to determine whether there was a significant difference

in parameters between groups. Analyses were done in GraphPad Prism v. 5.01.

Statistical Analyses.

Analyses were performed using Microsoft Office Excel 2007 and GraphPad Prism v. 5.01.

Correlations of APP metabolites were measured by calculating the correlation coefficient

(Pearson r values are reported). Soluble APPα, sAPPβ, and sAPPβ/α ratio were compared

among groups by using a student’s t-test. 95% confidence intervals were reported.

Page 39: The Processing of Amyloid Precursor Protein in the Central

21

Results

Development of a Specific and Sensitive sAPPβ ELISA.

Although commercial sAPPβ ELISA kits were available (from Covance and Meso Scale

Discovery), we developed our own sAPPβ sandwich ELISA that was selective for sAPPβ and

sufficiently sensitive to detect this protein in human CSF (final protocol may be found in this

chapter’s Experimental Methods and Study Design section). We tested the specificity of this

assay by running a titration curve of the sAPPβ and sAPPα protein standards on the same

ELISA. The results demonstrated that this assay was specific for sAPPβ and that cross-reactivity

with sAPPα was negligible (Figure 2.2.). The optical density (OD) for the sAPPβ standard of

8.5 ng/mL was approximately the same as that for the sAPPα standard of 300 ng/mL. This

indicated that this ELISA was approximately 35-fold more selective for sAPPβ than for sAPPα.

The CSF OD values fell within a linear range of the sAPPβ standard curve and well above the

sAPPα (300 ng/mL) value. Given that in biological samples sAPPα and sAPPβ were nearly

equal in molar concentrations, this minimal cross-reactivity of sAPPα in the sAPPβ ELISA was

negligible. Thus, we concluded that any fluctuations we observed in sAPPβ levels using this

ELISA were attributed solely to sAPPβ and not to sAPPα. Diluted CSF samples fell within the

linear range of the assay.

Page 40: The Processing of Amyloid Precursor Protein in the Central

22

Figure 2.2. Specificity and selectivity of the sAPPβ ELISA.

Optimization of a Specific and Sensitive sAPPα ELISA.

The developed sAPPα ELISA was adapted from (Olsson et al., 2005). The capture antibody was

8E5 (the same as used for the sAPPβ ELISA). The detection antibody was biotinylated

monoclonal 6E10, reactive to APP672-687, also known as Aβ1-16 (Signet Covance; Dedham, MA).

The standards used for this assay were the same as described in this chapter’s Experimental

Methods and Study Design section. The sAPPα ELISA showed no cross-reactivity with the

sAPPβ synthetic standard (Figure 2.3.).

Page 41: The Processing of Amyloid Precursor Protein in the Central

23

Figure 2.3. Specificity and selectivity of the sAPPα ELISA.

The protocol on which we based our assay directed to incubate the samples concurrently with the

2° antibody (6E10) overnight at 4 °C. To determine whether this was the optimal procedure, we

ran two ELISAs (using aliquots of the same samples) under the same conditions, barring the 2°

antibody incubation step. For one plate, the samples and 6E10 were concurrently incubated; for

the other plate, the samples were incubated separately overnight at 4 °C, and the following day,

the wells were incubated with 6E10 for 1.5 h at 37 °C. As demonstrated (Figure 2.4.),

incubation of sample concurrently with 6E10 was slightly better than the separate incubation.

OD values of all the points of the standard curve were slightly higher in the ELISA with

concurrent incubation; therefore, all values were further away from the “noise.” Also, the

ELISA with concurrent incubation was slightly more sensitive as it only had the three lowest

standards at baseline versus the four lowest standards in the other ELISA. Therefore, we decided

to follow the protocol with concurrent sample and 6E10 incubation.

Page 42: The Processing of Amyloid Precursor Protein in the Central

24

Figure 2.4. Optimization of sAPPα ELISA. Concurrent incubation of secondary antibody with sample for sAPPα ELISA provided better separation at the lower end of the standard curve than separate incubations. Concurrent incubation only showed three lowest points of standard curve at baseline, whereas separate incubation showed four points. All points during concurrent incubation had higher OD values than separate incubation, thus in the latter case all points were closer to “noise.”

At the beginning of the optimization process, we had trouble fitting the CSF OD values into the

range of the standard curve. These values verged on the lower end of the standard curve and

indicated that the CSF sAPPα values were significantly lower than reported in the literature

(Zetterberg et al., 2008; Sennvik et al., 2000; Vuletic et al., 2008; Lannfelt et al., 1995). We

hypothesized that this may have been due to a mistake in making the original dilutions of the

standards when they were first received. If the samples were actually more concentrated than we

had noted, then the CSF concentrations would be lower than expected. Purchasing new

standards and taking out the highest standard points, while adding standards at lower

concentrations allowed us to better place CSF OD values in the linear, middle part of the broad

Page 43: The Processing of Amyloid Precursor Protein in the Central

25

dynamic range of the standard curve (Figure 2.5.). With four dilutions of a CSF sample, we

were able to establish dilution-dependent concentrations. Based on the results, we chose 25-fold

as the working dilution for CSF. All the values of the CSF’s original concentration in four

humans were in a range of approximately 850-1600 ng/mL which was similar to reported values

(Zetterberg et al., 2008; Sennvik et al., 2000; Vuletic et al., 2008; Lannfelt et al., 1995).

Figure 2.5. Determination of dilution factor for CSF.

Circadian Patterns of APP Metabolites.

To determine APP processing over time within the same participant, temporal CSF samples from

a particular participant were randomly assigned a well position on four sandwich ELISAs:

Page 44: The Processing of Amyloid Precursor Protein in the Central

26

specific for sAPPα, sAPPβ, Aβ40, or Aβ42. This allowed for analysis of APP metabolite

concentrations in the CSF over time. To compare age and amyloid deposition effects on hourly

dynamics of APP metabolites, the Young Normal Control (YNC) group was compared to the

Amyloid- and Amyloid+ groups.

sAPPα and sAPPβ exhibit circadian patterns.

Cerebrospinal fluid sAPPα and sAPPβ hourly concentrations had significant fits to a 24 h cosinor

pattern in the YNC group. The average amplitude of the diurnal pattern for sAPPα was 3.1% ±

1.3% (SEM) (Figure 2.6.A). For sAPPβ, the average amplitude was 4.4% ± 1.6% (SEM)

(Figure 2.6.D).

Page 45: The Processing of Amyloid Precursor Protein in the Central

27

Figure 2.6. Diurnal patterns in APP metabolites. Cosinor fits applied to mean 36 h APP metabolite concentrations in each group after adjusting the baseline for each participant and subtracting out the group’s linear increase over time. Results from all three participant groups are reported for sAPPα (A-C), sAPPβ (D-F), Aβ40 (G-I), and Aβ42(J-L).

sAPPα and sAPPβ Circadian Amplitudes Lower with Older Age.

When a 24 h cosine curve was fit to the three group-averaged sAPPα hourly concentrations, the

YNC group exhibited an amplitude significantly deviated from zero (3.1%) and significantly

greater than the Amyloid- (0.9%) and Amyloid+ (2%) groups (Figure 2.6.A-C). Similar

Page 46: The Processing of Amyloid Precursor Protein in the Central

28

findings were observed when a cosine curve was fit to the three group-averaged sAPPβ hourly

concentrations (Figure 2.6.D-F). Amplitude of sAPPβ for the YNC group was 4.4%, Amyloid-

was 1.2%, and Amyloid+ was 2%. Amplitude of Aβ40 for the YNC group was 0.9%, Amyloid-

was 3.2% and Amyloid+ was 2.6% (Figure 2.6.G-I). Amplitude of Aβ42 for the YNC group

was 2.9%, Amyloid- was 3.7%, and Amyloid+ was 2.6% (Figure 2.6.J-L).

Individual sAPPα and sAPPβ Amplitude-to-Mesor Values Decrease with Age.

To control for differences in average values of amplitude and mesor among participants, the

amplitude-to-mesor ratios were calculated for each group. In the YNC group, sAPPα amplitude-

to-mesor ratio was, on average, 10.9% (min.: 2.3%, max.: 18.2%). Both the Amyloid- (6.7%;

Min: 1.2%, max.: 14.0%; *p = 0.01) and Amyloid+ (6.0%; min.: 1.5%, max.: 20.1%; *p = 0.01)

groups had significantly lower sAPPα amplitude-to-mesor ratios than YNC. There was no

significant difference between the Amyloid- and Amyloid+ groups (p = 0.6) (Table 2.1., Figure

2.7.B).

Similar trends were observed among groups for the sAPPβ amplitude-to-mesor ratio. In YNC,

the mean sAPPβ amplitude-to-mesor ratio was 14.4% (min.: 3.8%, max.: 21.2%). The Amyloid-

(8.2%; min.: 1.7%, max.: 19.9%; **p < 0.005) and Amyloid+ (9.2%; min.: 1.9%, max.: 23.3%;

*p = 0.02) groups had significantly lower sAPPβ amplitude-to-mesor ratios than YNC.

However, Amyloid- and Amyloid+ groups did not significantly differ from one another (p = 0.6)

(Table 2.2., Figure 2.7.D).

On the contrary, the Aβ40 amplitude-to-mesor ratio was not statistically different among all three

groups. In YNC, the mean Aβ40 amplitude-to-mesor ratio was 8.5% (min.: 2.2%, max.: 18.5%).

The Amyloid- group had a mean Aβ40 amplitude-to-mesor ratio of 9.1% (min.: 2.7%, max.:

Page 47: The Processing of Amyloid Precursor Protein in the Central

29

16%) and the Amyloid+ group had a mean Aβ40 amplitude-to-mesor ratio of 9.1% (min.: 2.8%,

max.: 24.4%). None of these groups’ Aβ40 amplitude-to-mesor ratios were significantly different

from one another (YNC vs. Amyloid-: p =0.7; YNC vs. Amyloid+: p =0.8; Amyloid- vs.

Amyloid+: p =1.0) (Table 2.3., Figure 2.7.F).

When Aβ42 amplitude-to-mesor ratio was measured, similar trends to the Aβ40 amplitude-to-

mesor ratios were observed. In YNC, the mean Aβ42 amplitude-to-mesor ratio was 9.4% (min.:

1.9%, max.: 18.5%). The Amyloid- group had a mean Aβ42 amplitude-to-mesor ratio of 8.04%

(min.: 3.6%, max.: 23.5%) and the Amyloid+ group had a mean Aβ42 amplitude-to-mesor ratio

of 8.0% (min.: 2.2%, max.: 22%). None of these groups’ Aβ42 amplitude-to-mesor ratios were

significantly different from one another (YNC vs. Amyloid-: p = 0.5; YNC vs. Amyloid+: p =

0.5; Amyloid- vs. Amyloid+: p = 1.0) (Table 2.4., Figure 2.7.H).

Page 48: The Processing of Amyloid Precursor Protein in the Central

30

Figure 2.7. Circadian rhythm parameters of four APP metabolites in YNC, Amyloid-, and Amyloid+ groups. A) Group-averaged sAPPα amplitudes were not significantly different between groups. B) The sAPPα amplitude-to-mesor ratio was highest in YNC and significantly lower in Amyloid- (*p = 0.01) and Amyloid+ (*p = 0.01). There was no significant difference between the Amyloid- and Amyloid+ groups (p = 0.6). C) Group-averaged sAPPβ amplitudes were significantly higher in YNC than in Amyloid- (*p = 0.05) and Amyloid+ (*p = 0.02). D) The sAPPβ amplitude-to-mesor ratio was highest in YNC and significantly lower in Amyloid- (**p = 0.003) and Amyloid+ (*p = 0.02). There was no significant difference between the Amyloid- and Amyloid+ groups (p = 0.6). E) Group-

Page 49: The Processing of Amyloid Precursor Protein in the Central

31

averaged Aβ40 amplitude values were not significantly different between any of the participant groups. F) Amplitude-to-Mesor ratios for Aβ40 amplitude were also not significantly different among groups. G) Group-averaged Aβ42 amplitudes were significantly highest in YNC when compared to Amyloid- (*p = 0.04) and Amyloid+ (***p < 0.0001). The Amyloid- group also had a significantly higher Aβ42 amplitude than the Amyloid+ group (***p < 0.001). H) The Aβ42 amplitude-to-mesor ratios did not different significantly among groups.

Table 2.1. Comparison of Cosinor Parameters for sAPPα among 3 participant groups. Abbreviations: YNC: participants classified as young (cognitively) normal healthy controls; Amyloid-: participants with a Mean Cortical Binding Potential (MCBP) less than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration > 350 pg/mL; Amyloid+: participants with MCBP greater than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration less than 350 pg/mL.

Table 2.2. Comparison of Cosinor Parameters for sAPPβ among 3 participant groups. Abbreviations: YNC: participants classified as young (cognitively) normal healthy controls; Amyloid-: participants with a Mean Cortical Binding Potential (MCBP) less than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration > 350 pg/mL; Amyloid+: participants with MCBP greater than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration less than 350 pg/mL.

Table 2.1. Comparison of cosinor parameters for sAPPα among 3 participant groups Participant Group Amplitude, ng/mL

Mean (SD) Mesor, ng/mLMean (SD)

Amplitude-to-Mesor Ratio, %Mean (SD)

YNC (n=13) 75.7 (40.2) 731.0 (312.4) 10.9 (5.4) Amyloid- (n=19) 59.2 (36.3) 1100.0 (694.8) 6.7 (3.8) Amyloid+ (n=17) 51.1 (40.1) 898.1 (300.8) 6.0 (4.7)

 

Table 2.2. Comparison of cosinor parameters for sAPPβ among 3 participant groups Participant Group Amplitude, ng/mL

Mean (SD) Mesor, ng/mLMean (SD)

Amplitude-to-Mesor Ratio, %Mean (SD)

YNC (n=13) 54.6 (21.3) 416.5 (181.7) 14.4 (5.7) Amyloid- (n=19) 32.8 (33.4) 383.2 (208.2) 8.2 (5.3) Amyloid+ (n=17) 31.6 (27.8) 344.3 (221.1) 9.2 (5.7)

 

Page 50: The Processing of Amyloid Precursor Protein in the Central

32

Table 2.3. Comparison of cosinor parameters for Aβ40 among 3 participant groups Participant Group Amplitude, pg/mL

Mean (SD) Mesor, pg/mL Mean (SD)

Amplitude-to-Mesor Ratio, % Mean (SD)

YNC (n=13) 698.8 (518.6) 8966.0 (3375.3) 8.5 (5.5) Amyloid- (n=15) 526.3 (311.0) 6373.0 (2951.2) 9.1 (4.6) Amyloid+ (n=14) 505.5 (365.5) 5872.0 (2977.5) 9.1 (5.7)

Table 2.3. Comparison of Cosinor Parameters for Aβ40 among 3 participant groups. Abbreviations: YNC: participants classified as young (cognitively) normal healthy controls; Amyloid-: participants with a Mean Cortical Binding Potential (MCBP) less than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration > 350 pg/mL; Amyloid+: participants with MCBP greater than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration less than 350 pg/mL.

Table 2.4. Comparison of cosinor parameters for Aβ42 among 3 participant groups Participant Group Amplitude, pg/mL

Mean (SD) Mesor, pg/mL Mean (SD)

Amplitude-to-Mesor Ratio, % Mean (SD)

YNC (n=13) 64.3 (36.3) 830.7 (423.9) 9.4 (5.7) Amyloid- (n=15) 39.5 (22.9) 518.6 (209.5) 8.0 (4.7) Amyloid+ (n=14) 14.5 (10.2) 206.9 (100.0) 8.0 (6.2)

Table 2.4. Comparison of Cosinor Parameters for Aβ42 among 3 participant groups. Abbreviations: YNC: participants classified as young (cognitively) normal healthy controls; Amyloid-: participants with a Mean Cortical Binding Potential (MCBP) less than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration > 350 pg/mL; Amyloid+: participants with MCBP greater than 0.18, or, in the absence of MCBP measurements, had a mean CSF Aβ42 concentration less than 350 pg/mL.

Individual Aβ42 Amplitude Values Decrease with Age and Amyloidosis. sAPPβ Amplitude

decreases with Age.

On average, for YNC the sAPPα amplitude was 75.7 ng/mL (min.: 7.7 ng/mL, max.: 139.1

ng/mL), in Amyloid- it was 59.2 ng/mL (min.: 15.1 ng/mL, max.: 149.7 ng/mL), and in

Amyloid+ it was 51.1 ng/mL (min.: 15.3 ng/mL, max.: 155.8 ng/mL). Although a trend toward a

decrease of sAPPα amplitude with increase in age was observed, the groups were not

Page 51: The Processing of Amyloid Precursor Protein in the Central

33

significantly different by their sAPPα mean amplitudes (YNC vs. Amyloid-: p = 0.2; YNC vs.

Amyloid+: p = 0.1; Amyloid- vs. Amyloid+: p = 0.5) (Table 2.1.; Figure 2.7.A.).

However, there was a significant difference in YNC from the Amyloid- and Amyloid+ groups

with respect to sAPPβ mean amplitudes. The sAPPβ mean amplitude in the YNC group was

54.6 ng/mL (min.: 21.8 ng/mL, max.: 92.2 ng/mL). The Amyloid- group had a mean sAPPβ

amplitude that was 40% lower (32.8 ng/mL; min.: 5.4 ng/mL, max.: 111.1 ng/mL) than YNC (p

= 0.05), whereas the Amyloid+ group had a mean sAPPβ amplitude that was 42% lower (31.57

ng/mL; min.: 2.4 ng/mL, max.: 93.7 ng/mL) than YNC (*p = 0.02). There was no significant

difference in sAPPβ amplitude between the Amyloid- and Amyloid+ groups (p = 0.9) (Table

2.2.; Figure 2.7.C).

For the YNC group, the mean Aβ40 amplitude was 699 pg/mL (min.: 287 pg/mL, max.: 1834

pg/mL). There was a trend for decreased mean Aβ40 amplitude with age: the Amyloid- group

had a mean Aβ40 amplitude of 526 pg/mL (min.: 148 pg/mL, max.: 1138 pg/mL) and the

Amyloid+ group had a mean Aβ40 amplitude of 506 pg/mL (min.: 91 pg/mL, max.: 1381

pg/mL). This trend did not reach statistical significance (YNC vs. Amyloid-: p = 0.3; YNC vs.

Amyloid+: p = 0.3; Amyloid- vs. Amyloid+: p = 0.9) (Table 2.3.; Figure 2.7.E).

In contrast, the mean Aβ42 amplitudes were significantly different among each group. For the

YNC, the mean Aβ42 amplitude was 64.3 pg/mL (min.: 10.6 pg/mL, max.: 130.1 pg/mL). The

Amyloid- group had a mean Aβ42 amplitude that was 39% lower (39.5 pg/mL; min.: 14.4 pg/mL,

max.: 99 pg/mL) than the YNC group (*p < 0.05). The Amyloid+ group had a mean Aβ42

amplitude that was 77% lower (14.5 pg/mL; min.: 3.7 pg/mL, max.: 41 pg/mL) than the YNC

Page 52: The Processing of Amyloid Precursor Protein in the Central

34

group (***p <0.0001) and 63% lower than the Amyloid- group (**p < 0.001) (Table 2.4; Figure

2.7.G).

Individual Aβ42 Mesor Values Decrease with Age and Amyloidosis. Aβ40 Amplitude

Decreases with Age.

In YNC, sAPPα levels had a mean mesor over 36 h of 731 ng/mL (min.: 250 ng/mL, max.: 1254

ng/mL) (Table 2.1.). In Amyloid-, sAPPα levels displayed a mean mesor of 1100 ng/mL (min.:

192 ng/mL, max.: 2805 ng/mL). The Amyloid+ group had a sAPPα mesor level of 898ng/mL

(min.: 386 ng/mL, max.: 1353 ng/mL). None of these groups’ mesors were significantly

different from one another (YNC vs. Amyloid-: p = 0.2; YNC vs. Amyloid+: p = 0.08; Amyloid-

vs. Amyloid+: p = 0.3) (Table 2.1.).

The mean sAPPβ mesor in the YNC group was 417 ng/mL (min.: 229 ng/mL, max.: 928 ng/mL).

This was not significantly different (p = 0.6) from the mean sAPPβ mesor in Amyloid- (383

ng/mL; min.: 101 ng/mL, max.: 832 ng/mL), nor from the mean sAPPβ mesor level in Amyloid+

(344 ng/mL; min.: 118 ng/mL, max.: 900 ng/mL; p = 0.4). The mean sAPPβ mesors in the

Amyloid- and Amyloid+ groups were also not significantly different from one another (p = 0.6)

(Table 2.2.).

The YNC group had a mean Aβ40 mesor of 8966 pg/mL (min.: 2430 pg/mL, max.: 13433

pg/mL). The Amyloid- group had a 29% lower mean Aβ40 mesor (6373 pg/mL; min.: 1332

pg/mL, max.: 11089 pg/mL; *p < 0.05) than the YNC group. The Amyloid+ group exhibited a

35% lower Aβ40 mesor (5872 pg/mL; min.: 1505 pg/mL, max.: 10768 pg/mL; *p = 0.02) than

the YNC group. There was no statistically significant difference in mean Aβ40 mesor values

between the Amyloid- and Amyloid+ groups (p = 0.7) (Table 2.3.).

Page 53: The Processing of Amyloid Precursor Protein in the Central

35

The mean Aβ42 mesors were significantly different among each group. On average, the YNC

group’s Aβ42 mesor was 831 pg/mL (min.: 256 pg/mL, max.: 1683 pg/mL). The Amyloid-

group had a 38% lower mean Aβ42 mesor (519 pg/mL; min.: 195 pg/mL, max.: 885 pg/mL; p =

0.02) than the YNC group. The Amyloid+ group had a 75% lower mean Aβ42 mesor (207

pg/mL; min.: 48.9 pg/mL, max.: 471 pg/mL; p <0.0001) than the YNC group and a 60% lower

mean Aβ42 mesor than the Amyloid- group (p <0.0001) (Table 2.4.).

No Diurnal Pattern Exhibited in Total Protein Levels.

As a negative control for diurnal rhythms, we assayed total CSF protein over 36 h using a micro

BCA assay. We measured that, on average, total protein concentrations were significantly lower

in YNC as compared with the older participants (YNC = 792.8 μg/mL, Amyloid- = 895.1 μg/mL,

and Amyloid+ = 871.4 μg/mL, ***p < 0.0001). A cosinor fit was applied to the mean of each

group’s total protein level, and showed no significant circadian pattern in any of the groups (p >

0.05) (Figure 2.8.). Therefore, the observed CSF sAPPα, sAPPβ, Aβ40 and Aβ42 diurnal

patterns presented here appear to be independent of potential systemic diurnal changes in CSF

total protein.

Page 54: The Processing of Amyloid Precursor Protein in the Central

36

Figure 2.8. Cosinor-fitted group-averaged total protein concentrations determined using a micro BCA assay. No significant diurnal patterns were apparent (p > 0.05 for each group).

Soluble APP and Aβ Positively Correlated, except in Amyloidosis.

To determine the relationship of α- and β-secretases on APP processing, correlations were

calculated for the average values of sAPPα, sAPPβ, Aβ40, and Aβ42 in three groups of

individuals: YNC, Amyloid-, and Amyloid+. Soluble APPα and sAPPβ were positively

correlated in all groups (YNC: r = 0.95, ***p <0.0001; Amyloid-: r = 0.93, ***p <0.0001;

Amyloid+: r = 0.86, **p < 0.002) (Figure 2.9.A). Soluble APPβ was positively correlated to

Aβ40 in YNC (r = 0.84, *p < 0.02), and Amyloid- groups (r = 0.68, **p < 0.005), but not in the

Amyloid+ group (r = 0.25, p = 0.5) (Figure 2.9.B). Soluble APPα trended towards a positive

correlation with Aβ40 in YNC (r = 0.69, p = 0.09) and was positively correlated in the Amyloid-

group (r = 0.84, **p < 0.003), but not in the Amyloid+ group (r = 0.2, p =0.6) (Figure 2.9.D).

There was a trend for sAPPβ to be positively correlated to Aβ42 in YNC (r = 0.57, p =0.2), and

Amyloid- groups (r = 0.5, p = 0.1); but not in the Amyloid+ group (r = -0.08, p =0.8) (Figure

Page 55: The Processing of Amyloid Precursor Protein in the Central

37

2.9.C). Similarly, sAPPα also trended to positive correlation with Aβ42 in YNC (r = 0.39, p

=0.4) and Amyloid- groups (r = 0.64, p < 0.05); but not in the Amyloid+ group (r =-0.01, p =

0.97) (Figure 2.9.E).

Figure 2.9. Correlations between APP metabolites. Amyloid Precursor Protein metabolites in the first CSF collection for each participant were correlated to determine relationships between the APP processing pathways. Each participant’s cerebrospinal

Page 56: The Processing of Amyloid Precursor Protein in the Central

38

fluid sample was drawn between 7:30AM-9:00AM. The four APP metabolites’ concentrations were measured using four separate metabolite-specific ELISAs, and, for each participant, plotted against one another. The correlation coefficient was then calculated for each group. A) sAPPα and sAPPβ concentrations for each participant were plotted against one another and showed a positive correlation in all groups (YNC: r = 0.95, ***p <0.0001; Amyloid-: r = 0.93, ***p < 0.0001; Amyloid+: r = 0.86, **p < 0.002). B) sAPPβ and Aβ40 concentrations for each participant were plotted against one another : YNC: r = 0.84, *p < 0.05; Amyloid-: r = 0.68, **p < 0.005; Amyloid+: r = 0.25, p = 0.5). C) sAPPβ and Aβ42 concentrations for each participant were plotted against one another and a positive correlation was detected in YNC (r = 0.57, p = 0.2) and Amyloid- groups (r = 0.5, p = 0.14), but no correlation was detected in the Amyloid+ group (r = -0.08, p = 0.8). D) sAPPα and Aβ40 concentrations for each participant were plotted against one another and compared among the groups: YNC: r = 0.69, p = 0.09; Amyloid-: r = 0.84, **p < 0.003; Amyloid+: r = 0.2, p = 0.6). E) sAPPα and Aβ42 concentrations for each participant were plotted against one another. The correlation results are as follows: YNC (r = 0.39, p = 0.4); Amyloid- (r = 0.64, p < 0.05); and Amyloid+ (r = -0.01, p = 1.0).

sAPPβ/sAPPα Ratio is Elevated in Amyloidosis.

In order to determine the effects of age and amyloidosis on the APP processing pathways, APP

metabolites were compared among 3 groups: YNC, Amyloid-, and Amyloid+. The sAPPβ to

sAPPα ratio was 0.26 (≈1:3 ratio, 0.01, n = 15) in YNC, and 0.26 (≈1:3 ratio, 0.02, n = 16) in

Amyloid-. However, it increased to 0.32 (≈1:2 ratio, 0.05, n = 10) for Amyloid+. The

sAPPβ/sAPPα ratio was significantly higher in Amyloid+ participants than in Amyloid- (*p

=0.02) and YNC (**p =0.002) (Figure 2.10.A). However, independent sAPPα and sAPPβ

values were not significantly different between groups, suggesting that the sAPPβ/sAPPα ratio

corrected for other variances which were not associated with amyloidosis (Figure 2.10.B-C).

Page 57: The Processing of Amyloid Precursor Protein in the Central

39

Figure 2.10. Separating participant groups by the sAPPβ/sAPPα ratio. We compared sAPPβ and sAPPα concentrations, as well as the sAPPβ/sAPPα ratio, among groups using in the first CSF collection. Each participant’s first CSF sample was drawn between 7:30AM-9:00AM. sAPPβ and sAPPα concentrations were measured using two separate metabolite-specific ELISAs. Student’s t-test was used and graphs show 95% Confidence Interval error bars. A) sAPPβ/sAPPα ratio was higher with amyloid deposition (Amyloid+) as compared to healthy, older controls (Amyloid-) (*p = 0.02) or young healthy controls (YNC) (**p = 0.002). No significant difference was detected between the ratio of the YNC and Amyloid- groups (p = 0.64). B) sAPPα concentrations were not significantly higher in Amyloid+ than in YNC (p = 1.0) or Amyloid- (p = 0.5). No significant difference was detected between the sAPPα concentration of the YNC and Amyloid- groups (p = 0.4). C) sAPPβ concentrations were not significantly higher in Amyloid+ than in YNC (p = 0.09) nor Amyloid- (p = 0.6). No significant difference was detected between sAPPβ concentrations from the YNC and Amyloid- groups (p = 0.3).

Discussion

We evaluated whether APP exhibited diurnal fluctuations similar to that of Aβ, which would

help inform why Aβ demonstrated a diurnal pattern. We also determined normal α- and β-

processing of APP in the human CNS, and assessed whether AD pathology is associated with

alterations in APP processing.

The regulation of α- and β-secretase over time, including potential dynamic changes of sAPPα

and sAPPβ within an individual, were not previously evaluated, although Aβ diurnal activity was

Page 58: The Processing of Amyloid Precursor Protein in the Central

40

described in healthy, young human participants (Bateman et al., 2007). We recently

demonstrated that both in CSF (Huang et al., 2012a), and in plasma (Huang et al., 2012b), the

physiological Aβ diurnal fluctuation described in young participants diminishes significantly

with increasing age, but is not further decreased in amyloidosis. Further, previous studies in

mice indicated that sleep regulation may play a critical role in the risk and development of AD

(Kang et al., 2009), but more recent findings indicated that it may be Aβ aggregation that

disrupted both the sleep-wake cycle and Aβ diurnal fluctuation (Roh et al., 2012). For example,

longitudinal studies found a strong relationship between sleep circadian patterns as well as sleep

disordered breathing and risk of MCI and AD (Yaffe et al., 2011; Tranah et al., 2011).

Therefore, we sought to determine the relationship between α- and β- processing pathways in

individuals over time, and also determine if APP regulation contributes to Aβ circadian patterns.

In the YNC group, we found that sAPPα, sAPPβ, Aβ40, and Aβ42 concentrations were dynamic

over 36 h, with diurnal patterns. The lowest concentrations were in the morning (approximately

9AM), and the concentrations peaked in the evening, 12 h later. This suggests that in YNC

dynamic changes in these protein levels were due to dynamic changes in APP availability,

whether by its production (transcription or translation) or transport to the site of processing (i.e.

axonal transport). Amyloid-β also demonstrated a diurnal pattern with a peak and trough

approximately 3 h after sAPPα and sAPPβ. This suggests that APP diurnal availability likely

plays a role in Aβ diurnal patterns.

Diurnal patterns of sAPPα and sAPPβ were diminished in the Amyloid- group. Aβ40, and Aβ42

did not show any significant diurnal pattern in the Amyloid- group similar to prior work from our

laboratory (Huang et al., 2012a). Taken together, these findings indicated that with age, there

Page 59: The Processing of Amyloid Precursor Protein in the Central

41

was a loss of APP dynamics or availability, which resulted in the noted loss of not only sAPP,

but also Aβ, diurnal patterns.

A diurnal pattern of sAPPα and sAPPβ was also not seen to a similar extent in the Amyloid+

group as in the Amyloid- group. However, the diurnal patterns in Aβ40 and Aβ42 were even

more significantly diminished in the Amyloid+ group than in the Amyloid- group. This further

marked decrease in Aβ40 and Aβ42 diurnal patterns in the presence of amyloidosis did not

correspond to any decrease in sAPP diurnal patterns. This disconnect may be an effect of

downstream APP cleavage events and not due to APP dynamics or availability, which seems to

be the case in general aging. Potentially the extent of γ-secretase cleavage, which is controlled

by availability of the γ-secretase components or the γ-secretase level of activity, may play a role

in diminishing the diurnal patterns of the two Aβ species we measured.

Further, sAPPα and sAPPβ were positively correlated in all groups. Positive correlation of the α-

and β-secretase products suggested that for the non-competitive model of APP pathways, the

total APP availability drove changes in sAPPα and sAPPβ. Soluble APPα and sAPPβ were

positively correlated with both Aβ species in YNC and elderly controls. However, the

correlation between the sAPP species and Aβ42 was lost with amyloidosis. Previously reported

results taken in the human CNS showed a positive sAPPα to sAPPβ correlation between

individuals also suggesting non-competitive α- and β-pathways (Lewczuk et al., 2010; Gabelle et

al., 2010; Alexopolous et al., 2011). However, in vitro studies of inhibitors or activators, or

genetically decreasing BACE1 (a β-secretase protein) or ADAM10 (an α-secretase protein)

(Caporaso et al., 1992; Buxbaum et al., 1993; Savage et al., 1998; Skovronsky et al., 2000; Kim

et al., 2009; May et al., 2011) supported the hypothesis that α- and β-secretase pathways compete

Page 60: The Processing of Amyloid Precursor Protein in the Central

42

for the same APP pool owing to inverse correlations during inhibition of secretases (i.e. when

processing through one pathway decreases, the processing of the alternative pathway increases).

These studies suggested that there may be an inverse relationship between the α- and β-

pathways in inhibitor studies, whereas our study showed that during physiologic APP processing

in the human CNS, α- and β- processing were positively correlated.

We found that the molar ratio of sAPPα to sAPPβ was approximately 3:1 with a shift from α- to

β-processing in the setting of amyloid deposition to 2:1. The ratio difference between these

groups was not age-related because there was no significant difference between YNC and

Amyloid- groups. Prior reports estimated α to β ratios of 10:1 (Buxbaum et al., 1993,

Skovronsky et al., 2000); however, these estimates, made for an in vitro situation, likely had

lower β-secretase activity than is truly present in the CNS, because β-secretase is mostly found in

the brain (Vassar et al., 1999, Yan et al., 1999; Lin et al., 2000). We further showed that on

average sAPPβ/sAPPα was significantly higher in Amyloid+ participants than in Amyloid-

participants and YNC; therefore the ratio may be a useful indicator of Aβ plaque deposition.

These results are consistent with recent findings of increased CSF sAPPβ in the presence of

decreased Aβ42 and increased tau (Gabelle et al., 2010, Lewczuk et al., 2010). However, some

reports indicate increased sAPPα (Alexopoulos et al., 2011) whereas others show no difference

(Gabelle et al., 2010, Lewczuk et al., 2010), similar to our findings. To summarize, amyloidosis,

not age, was associated with a constitutive change in the β- to α- APP processing between

individuals.

In conclusion, we show that diurnal dynamics of APP metabolites diminished with age, and, only

for Aβ, were further attenuated with amyloidosis. These results may confound other studies’ data

of sAPPα, sAPPβ, Aβ40, and Aβ42 levels in AD versus non-AD participants where CSF was

Page 61: The Processing of Amyloid Precursor Protein in the Central

43

collected and metabolites were measured at a single time point. This may explain the wide range

of concentrations of metabolites presented from various groups. We also indicate that taking a

ratio of sAPPβ/sAPPα may correct for these inconsistencies. Further, we demonstrated that there

was a positive correlation among soluble APP metabolites, which diminished with amyloidosis.

This dissociation was probably due to CSF Aβ42 levels in AD no longer being representative of

APP processing owing to the sequestering of Aβ, particularly Aβ42, in plaques.

Attributes of this study included sampling from the human CNS in three different groups where

total protein showed stability over time. Therefore, CSF APP dynamics seem to be independent

of CSF total protein levels. However, we did not directly measure α- and β-secretase activities

or production rates of APP metabolites. Our study does not answer the question of what causes

APP to rise and fall in a diurnal pattern and does not allow us to distinguish among potential

reasons, such as transcription, translation, or transport. A potential caveat to the study is that

about 30% of our Amyloid- subject group had a CDR > 0, indicating that they may have

dementia other than Alzheimer’s disease. If a CDR+ subject has no significant amyloid plaque

deposition and/or the subject’s Aβ42 level below 350 pg/mL (is considered Amyloid-), we

presume that the potential non-AD dementia that they may have is not associated with APP

processing since free Aβ, as well as Aβ in plaques, isn’t affected. That being said, removing

Amyloid- subjects who were CDR+ from our analyses did not alter the significance of the

observed trends, or lack thereof. Future studies into APP processing pathways, including

production rates of APP and α- and β-secretases should be able to inform about causes of APP

dynamics.

Page 62: The Processing of Amyloid Precursor Protein in the Central

44

Chapter 3 Soluble Amyloid Precursor Protein Turnover Rates in Healthy, Young Humans

Introduction

In 2006, our group reported the initial Aβ turnover rates in young, healthy humans. The

Fractional Synthesis Rate (FSR) and the Fractional Clearance Rate (FCR) were calculated in six

participants, and were determined to be 7.6%/h and 8.3%/h, respectively (Figure 3.1.) (Bateman

et al., 2006).

Figure 3.1. Metabolism of Aβ in healthy young humans (n=6) (Bateman et al., 2006).

As Aβ is a metabolite of the larger Amyloid Precursor Protein, we decided to investigate the

metabolism (production and clearance rates) of total soluble APP. Using 8E5 antibody bound to

CNBr-activated SepharoseTM 4B beads, we isolated total sAPP from CSF (collected over a 36 h

time course, at beginning of which the participant was infused with 13C6-leucine) and H4 media

that was labeled at 1.25%, 2.5%, 5%, 10%, 20%, or unlabeled (0%). This media contained

similar amounts of sAPP as found in human CSF and was, therefore, used as a standard to

Page 63: The Processing of Amyloid Precursor Protein in the Central

45

generate a standard curve. Soluble APP isolated from CSF/media was then proteolytically

cleaved with trypsin, and the digested sample was injected onto an HPLC column. We measured

the production and clearance rates based on the tryptic unlabeled and labeled peptides:

EQNYSDDVL(L*)ANMISEPR (the labeled peptide was 6 Da heavier because the label on the

leucine was 13C6). Both the labeled and unlabeled tryptic peptides eluted from the HPLC

column and were ionized at the same time. The precursor ions were separated based on mass-to-

charge ratio (m/z). The two ions of interest were further fragmented into MS2 (“product”) ions

that were also separated by m/z. The labeled product ions were either 3 or 6 Da heavier than the

unlabeled product ions depending on the charge of the ion. A computer program, SILTmass,

(written by V. Ovod and D. Elbert at Washington University in St. Louis) was used to determine

the ratio of the labeled to unlabeled product ions of interest. The fractional synthesis rate (FSR)

was calculated based on the slope to precursor ratio. The precursor ratio of 9.86% was

calculated based on the total available labeled and unlabeled leucine.

Experimental Methods and Study Design

Participants and Sample Collection.

Participants (Young Normal Controls) were recruited from the general public after approval by

the Washington University Human Studies Committee and the General Clinical Research Center

(GCRC) Advisory Committee. Three Caucasian male participants under the age of 60 years

were assessed in this study (age at time of study: 23.4 yrs., 25.5 yrs., and 58.9 yrs.). The ApoE

genotype was available only for the third participant; he had two copies of the ApoE-ε3 allele.

After informed consent was obtained, participants were screened to determine that they were in

good general health and without any neurologic diseases. Participants fasted for 11 h prior to

Page 64: The Processing of Amyloid Precursor Protein in the Central

46

admittance into the study, at 7:00 A.M. During the course of the study, the GCRC kitchen

provided participants with free access to water and three daily meals at set times: 9:00 A.M.,

1:00 P.M., and 6:00 P.M. To avoid potential confounding effects on the labeled leucine infusion

study, the meals provided to participants were low in leucine. Meals consisted of approximately

60% carbohydrates, 20% fat, and 20% protein. Participants had an intravenous (IV) catheter

placed in one of their contra-lateral antecubital veins to collect 12 mL of blood hourly for 36 h,

except between 11:00 P.M. and 7:00 A.M. when collection occurred bihourly. A subarachnoid

catheter was inserted into the lumbar region, L3-L4 interspace, using a Touhy needle, so as to

allow for serial CSF sampling without the need for multiple lumbar punctures. During the

course of the 36 h study, 6 mL CSF was sampled hourly. Both blood and CSF samples were

apportioned into 1 mL aliquots in Axygen maximum-recovery polypropylene tubes. The

samples were immediately frozen and stored at -80 °C. Once the participants were catheterized,

they were discouraged from leaving their bed, except for restroom use.

13C6-Leucine infusion protocol.

Prior to study onset, an intravenous catheter, through which stable isotope labeled leucine

solution would be administered, was placed in each participant’s antecubital vein. One day prior

to the study, the 13C6-labeled leucine (Cambridge Isotope Laboratories, Inc.; Andover, MA) was

prepared by dissolution in medical grade normal saline solution and filtration through a 0.22μm

filter. Administration of 13C6-labeled leucine took place with an initial bolus of 2 mg/kg over 10

min to reach a steady state of labeled leucine. This was immediately followed by a continuous

intravenous infusion with a medical IV pump at a rate of 1.8, 2.0, or 2.5 mg/kg/h for 9 h.

Page 65: The Processing of Amyloid Precursor Protein in the Central

47

Cell Culture and Media Standards.

Immortalized human H4 neuroglioma cells stably transfected with human APP751 (H4-APPwt)

(courtesy of T. E. Golde Laboratory, UF-Gainesville) were grown in Dulbecco's Modified Eagle

Medium (DMEM) with 10% Fetal Bovine Serum (FBS) and passaged twice. Leucine-free

DMEM, containing no serum, was obtained from the Washington University Tissue Culture

Support Center and supplemented with a mixture of 13C6-labeled leucine solution such that the

total final leucine concentration was the same as in standard DMEM (105 mg/L). The final

media solutions had: 0%, 1.25%, 2.5%, 5%, 10%, or 20% 13C6-leucine. Labeled DMEM was

supplemented with 1:50 B-27 (a serum-free supplement) and 1% each of Penicillin-Streptomycin

and Zeocin (antibiotics). During the third passage, cells were pooled and split evenly. The H4-

APPwt cells were reconstituted in one of the six prepared media solutions and cultured for 24 h to

allow for sufficient incorporation of 13C6-leucine into proteins, such as APP. Following the

incubation with labeled DMEM, the media solutions were collected and analyzed by ELISA to

determine that they contained similar concentrations of sAPP as the human CSF samples. The

labeled media was then apportioned into Axygen tubes in 1 mL aliquots and immediately frozen

and stored at -80 °C.

Page 66: The Processing of Amyloid Precursor Protein in the Central

48

Figure 3.2. Human labeled leucine infusion protocol and sample collection. A human participant enrolled in the study was catheterized prior to the study onset. Continuous intravenous 13C6-labeled leucine infusion occured at a rate of 2mg/kg/h for 9 h, following an initial bolus infusion of 2 mg/kg over 10 min. Through the lumbar catheter, CSF (6 mL) was collected hourly. Simultaneously, blood (12 mL) was collected hourly or bihourly. Cerebrospinal fluid and blood sampling occured over the course of 36 h (Bateman et al., 2006).

Immunoprecipitation and digestion of total sAPP.

We sought to determine total sAPP production and clearance rates in human CSF. To this end,

we first isolated total sAPP from the CSF and media standard samples by immunoprecipitation

Page 67: The Processing of Amyloid Precursor Protein in the Central

49

of the samples with beads bound to 8E5, a mid-domain APP monoclonal antibody (directed

against APP444-592, courtesy of Eli Lilly). To prepare the samples for IP we thawed them on ice:

1 mL of CSF per each time-point (for a total of 37 CSF samples) and a H4-APPwt 13C6-leucine

labeled media standard curve (1 mL media for each of the six points on the standard curve). We

added 100 μL of 5M Guanidine Tris (pH 8.0) and 12.5 μL protease inhibitor cocktail (40 μg/mL

aprotinin and 20 μg/mL leupeptin), and 30 μL of a 50% 8E5-bead slurry (prior to study onset,

CNBr-activated SepharoseTM 4B beads (GE Healthcare) were covalently bound to 8E5 antibody

(1mg/mL) in 0.02% Sodium Azide in PBS, and stored at 4 °C). We incubated the samples with

rotation overnight (approximately 18 h) at 4 °C. The next day the supernatant was removed and

the beads were rinsed three times with 25 mM ammonium bicarbonate (AmBic) and aspirated

until dry. The protein bound to the antibody-bead complex was then eluted with 50 µL formic

acid (FA) for two minutes. The eluant, containing the protein isolated from the sample, was

removed using a crimped gel loading tip, and placed into an Axygen maximum-recovery

polypropylene tube. Subsequently, the samples were evaporated in a rotary evaporator at 37 °C

until fully dry (approximately 30 min) and reconstituted in 25 µL of 10% acetonitrile (ACN) in

AmBiC. The isolated sAPP was proteolytically cleaved by 20 µL Sequencing Grade Modified

Trypsin (Promega; Madison, WI) during an overnight (approximately 18 h) incubation at 37 ºC.

Trypsin predominantly cleaves proteins at the carboxylic side of the amino acids Lysine (K) and

Arginine (R) except when either is bound to a C-terminal Proline (P). The doubly charged

([M+2H]+2) sAPP tryptic peptide used for quantitation in this study had the sequence:

EQNYSDDVL(L*)ANMISEPR (unlabeled: m/z = 990.95; labeled: m/z = 993.95).

Page 68: The Processing of Amyloid Precursor Protein in the Central

50

Figure 3.3. Isolation of Amyloid Precursor Protein from cerebrospinal fluid. Amyloid Precursor Protein was immunoprecipitated from CSF or media standards overnight at 4 °C using 8E5 antibody-bound beads. After washing beads, trypsin was used to proteolytically digest APP bound to the beads. Trypsin cleaves at the carboxylic side of Lysine (K) and Arginine (R) residues, except when either is bound to a C-terminal Proline (P).

Peptide quantitation by liquid chromatography-mass spectrometry (LC-MS).

The pre-determined tryptic peptides were quantified on a Thermo-Finnigan LTQ, a linear trap

quadrupole mass spectrometer, equipped with a New Objective nanoflow ESI source. The

peptides were separated by RP HPLC using an Eksigent 2D-LC nanoflow pump operating in 1D

mode at a flow rate of 200 nL/min. The digested sample (5 μL) of tryptic peptides was injected

onto a New Objective picofrit column packed to 12 cm with 5 μm Magic C18aq packing material

(Michrom). Mobile Phase A contained 0.1% formic acid (FA) in water and Mobile Phase B

contained 0.1% FA in acetonitrile (ACN). Both the labeled and unlabeled tryptic peptides eluted

from the HPLC column and were ionized simultaneously. The precursor ion was separated

based on mass-to-charge ratio (m/z). The doubly charged ([M+2H]+2) sAPP tryptic peptide used

for quantitation in this study had the sequence: EQNYSDDVL(L*)ANMISEPR (unlabeled: m/z

= 990.95; labeled: m/z = 993.95). These were further fragmented into b- and y- product ions that

Page 69: The Processing of Amyloid Precursor Protein in the Central

51

were also separated by m/z. The labeled product ions were 3 or 6 m/z heavier than the unlabeled

product ions depending on whether the ion was doubly or singly charged, respectively.

Figure 3.4. Liquid Chromatography-Mass Spectrometry analysis of sAPP peptides. The labeled and unlabeled sAPP tryptic peptides (EQNYSDDVL(L*)ANMISEPR) that were isolated from CSF/media standards were injected onto a nano-liquid chromatography (nanoLC) column, from which they simultaneously eluted during the LC gradient. Eluted peptides became charged in the electrospray and were then admitted to the LTQ mass spectrometer. A pre-determined MS (“Precursor”) ion corresponding to that peptide was then selected, based on the mass-to-charge ratio (m/z). The precursor ions were further fragmented to produce “Product” ions that were also separated based on m/z.

Page 70: The Processing of Amyloid Precursor Protein in the Central

52

Figure 3.5. Precursor and product ion chromatograms and product ion mass spectra for sAPP peptide. The upper panel shows representative chromatograms of the sAPP peptides monitored (EQNYSDDVL(L*)ANMISEPR), which eluted at approximately 19.5 min into the LC gradient. The lower panel presents the mass spectra of the unlabeled and labeled product ions, with the observed, unique b- and y- product ion profile formed during fragmentation.

Free Leucine Quantitation by gas-chromatography-mass spectrometry (GC-MS).

Cerebrospinal fluid 13C6-leucine enrichment was determined using gas capillary gas

chromatography mass spectrometry (GC-MS; Agilent 6890N gas chromatograph and Agilent

5973N mass selective detector) in negative chemical ionization mode as described previously

(Yarasheski et al., 1992, Bateman et al., 2007). As described in Wolfe et al. (2005), 13C6-leucine

enrichment was quantified as a tracer to tracee ratio (TTR).

Page 71: The Processing of Amyloid Precursor Protein in the Central

53

Quantitation of labeled ratio.

The percentage of labeled sAPP was determined by taking the ratio all of b- and y- product ion

intensities from the unlabeled and labeled peptide (EQNYSDDVL(L*)ANMISEPR) of APP444-

592. The ratio was computed using a custom Microsoft Excel spreadsheet with macros. The

tandem MS quantification of labeled to unlabeled peptides that was used here is detailed in

Bateman et al. (2007a).

Quantitation of fractional synthesis rate and monoexponential slope fractional clearance

rate.

A fractional synthesis rate (FSR) and monoexponential slope fractional clearance rate (FCR) for

sAPP was quantified as previously reported (Wolfe et al., 2005). Briefly, FSR was calculated as

the slope of the labeled metabolite during the production phase (h: 5-14), divided by the CSF

13C6-leucine enrichment which served as the precursor pool for APP synthesis. The standard

equation from Wolfe et al. (2005) was used:

2 1

(Et2 - Et1)sAPP (t2 - t1) is defined as the slope of the line during the labeling period, and

Precursor E is the ratio of labeled free leucine to unlabeled free leucine. The slope of the linear

regression was calculated from 5 to 14 h and divided by the average of CSF 13C6-labeled leucine

level during the infusion period.

Monoexponential slope FCR was computed by fitting the slope of the natural logarithm of the

clearance phase of the sAPP labeling curve (h: 24 - 36). The standard equation monoexponential

FCR equation was used (Wolfe et al., 2005):

Page 72: The Processing of Amyloid Precursor Protein in the Central

54

ln

/∆ 24 36

Statistical Analyses.

Mean and standard deviation were calculated using Microsoft Office Excel 2007.

Results

Soluble APP Turnover Rate is Faster than Aβ Turnover Rate.

We calculated the sAPP FSR for three individuals and determined that the mean was 3.68%/h ±

0.19 (SD) (Figure 3.7). This compared to previous work done in our lab where Aβ FSR was

determined to be 7.6%/h in young healthy controls. Fractional clearance rate (FCR) of sAPP

was calculated and expressed as the natural log of labeled to unlabeled sAPP. The FCR was only

calculated in one participant because the two other participants’ labeled sAPP did not clear in the

36 h time course. The sAPP FCR for this individual was 4.74%/h (Figure 3.7). This compared

to the FCR of Aβ in young healthy controls which was 8.3%/h. A sample sAPP

labeled/unlabeled leucine curve (over 36 h) as measured in human CSF is provided (Figure 3.6),

as well as the FCR curve from the same participant.

Page 73: The Processing of Amyloid Precursor Protein in the Central

55

Figure 3.6. Total sAPP immunoprecipitated from human CSF was digested with trypsin, and tryptic peptides containing 13C6-leucine were used to measure total sAPP kinetics. The fractional synthesis rate (FSR) over 36 h was calculated from the 13C6-sAPP kinetic curve. A) A representative metabolism curve of the sAPP peptide EQNYSDDVL(L*)ANMISEPR from one participant used in quantitation of the kinetics of total sAPP after immunoprecipitation with 8E5. Total sAPP FSR (production) in this participant was 3.9%/h. Total sAPP FCR for the same participant was 4.74%/h. B) H4 neuroglioma cells were labeled with 13C6-leucine at 0%, 1.25%, 2.5%, 5%, 10%, and 20%. The cell media was collected and a mid-domain APP antibody was used to immunoprecipitate total sAPP. Isolated sAPP underwent proteolytic cleavage by trypsin. Quantitation of 13C6-leucine labeled and unlabeled sAPP peptides for each media standard provided that media standard’s observed percentage label of leucine. The observed value was plotted against expected value for all 6 media standards, thus generating the standard curve which demonstrates high accuracy and linearity (r2 = 0.998).

Page 74: The Processing of Amyloid Precursor Protein in the Central

56

Figure 3.7. Metabolism of total sAPP in healthy young humans (Dobrowolska et al., 2008).

Discussion

Although lacking a large sample size, in this experiment we were able to show some preliminary

data that indicated a faster turnover rate for Aβ as compared to total sAPP. Soluble APP FSR

was 3.68%/h whereas Aβ FSR had been previously determined to be 7.6%/h. The FCR for sAPP

was was 4.74%/h, whereas Aβ FCR had been previously determined to be 8.3%/h. We were

initially surprised that the turnover (both FSR and FCR) for sAPP was considerably slower than

Aβ. This was striking considering that Aβ is a later cleavage product of APP than either sAPPα

or sAPPβ, which were effectively both being measured in our sAPP calculations. We presumed

that the individual turnover rates of sAPPα and sAPPβ were markedly different, and perhaps our

slower turnover rate for sAPP was being driven by a slower turnover of APP processed through

the α-pathway, while the APP processing down the β-pathway was faster, and the turnover rate

of sAPPβ would be similar to the previously reported Aβ turnover rate. It is also possible that

our observations were solely an artifact of small sample size (n=3 for FSR and n=1 for FCR).

Page 75: The Processing of Amyloid Precursor Protein in the Central

57

There may be vast inter-subject variability for these FSRs and FCRs in humans, even in healthy

adults. Therefore, to test whether the phenomenon we observed (sAPP turnover was slower than

Aβ turnover) was not an artifact, we decided to develop a SILK method to measure sAPPα and

sAPPβ simultaneously from the same CSF sample and compare the turnover rates of the two

individual sAPPs to the turnover rate of Aβ. Such an analysis would encompass a large sample

size of YNC and AD human subjects. This method development is outlined in the following

chapter (4).

Page 76: The Processing of Amyloid Precursor Protein in the Central

58

Chapter 4 Method Development for sAPPα and sAPPβ Kinetic Measurements in Healthy, Young Humans Portions of this chapter appeared in Patent Pending Pub. No.: WO/2010/056815.

Introduction

As total sAPP production and clearance rates do not provide insight into the processing of sAPP

through the amyloidogenic versus the non-amyloidogenic pathway, we developed a novel LC-

MS assay that allowed us to quantitate sAPPα and sAPPβ kinetics in the same sample at the

same time. Samples were acquired and handled as described in Chapter 4 up until the proteolytic

digestion step. Instead of using the protease trypsin, we used Arginine-C (Arg-C)

(Worthington), which cleaves at the C-terminus of Arginine (R). Based on the positions of β-

secretase, α-secretase, and Arg-C cleavage, two candidate peptides containing a leucine and

unique to either sAPPβ or sAPPα are produced (Figure 4.1.).

Figure 4.1. Amyloid Precursor Protein Sequence with positions of proteolytic cleavage, antibody binding sites, and peptides chosen for quantitation.

Page 77: The Processing of Amyloid Precursor Protein in the Central

59

We monitored and calculated the ratio of labeled to unlabeled b- and y- product ions produced as

described in Chapter 3. However, this time the b- and y- product ions measured were derived

from different peptides. These peptides for sAPPβ are: 1) unlabeled:

PGSGLTNIKTEEISEVKM, [M+H]+3 of m/z = 645; and 2) labeled:

PGSGL*TNIKTEEISEVKM, [M+H]+3 of m/z = 647. The two peptides for sAPPα were almost

identical to those for sAPPβ, with the exception of having five additional amino acids at the C-

terminal end of the peptide: 1) unlabeled: PGSGLTNIKTEEISEVKMDAEFR, [M+4H]+4 of m/z

= 638.98; 2) labeled: PGSGL*TNIKTEEISEVKMDAEFR, [M+4H]+4 of m/z = 640.48. Elution

of the unlabeled and labeled sAPPβ peptides from the HPLC column occurred only 90 sec prior

to the elution of the unlabeled and labeled sAPPα peptides (Figure 4.6). This was a sufficient

separation in time to detect two separate quantifiable precursor ions and allow MS2 profiling of

distinct b- and y- ions for unlabeled versus labeled peptides (Figure 4.7).

Experimental Methods and Study Design

Participants and Sample Collection.

Participants (Young Normal Controls) were recruited from the general public after approval by

the Washington University Human Studies Committee and the General Clinical Research Center

(GCRC) Advisory Committee. A total of one participant was assessed in this study. After

informed consent was obtained, participants were screened to determine that they were in good

general health and without any neurologic diseases. Participants fasted for 11 h prior to

admittance into the study, at 7:00 A.M. During the course of the study, the GCRC kitchen

provided participants with free access to water and three daily meals at set times: 9:00 A.M.,

Page 78: The Processing of Amyloid Precursor Protein in the Central

60

1:00 P.M., and 6:00 P.M. To avoid potential confounding effects to the labeled leucine infusion

study, the meals provided to participants were low in leucine. Meals consisted of approximately

60% carbohydrates, 20% fat, and 20% protein. Participants had an intravenous (IV) catheter

placed in one of their contra-lateral antecubital veins to collect 12 mL of blood hourly for 36 h,

except between 11:00 P.M. and 7:00 A.M. when collection occurred bihourly. A subarachnoid

catheter was inserted into the lumbar region, L3-L4 interspace, using a Touhy needle, so as to

allow for serial CSF sampling without the need for multiple lumbar punctures. During the

course of the 48 h study, 6 mL CSF was sampled hourly. Both blood and CSF samples were

apportioned into 1 mL aliquots in Axygen maximum-recovery polypropylene tubes. The

samples were immediately frozen and stored at -80 °C. Once the participants were catheterized,

they were discouraged from leaving their bed, except for restroom use.

13C6-Leucine infusion protocol.

Prior to study onset, an intravenous catheter, through which stable isotope labeled leucine

solution would be administered, was placed in each participant’s antecubital vein. One day prior

to the study, the 13C6-labeled leucine (Cambridge Isotope Laboratories, Inc. (Andover, MA))

was prepared by dissolution in medical grade normal saline solution and filtration through a 0.22

μm filter. Administration of 13C6-labeled leucine took place by an initial bolus of 2 mg/kg over

10 min to reach a steady state of labeled leucine. This was immediately followed by a

continuous intravenous infusion with a medical IV pump at a rate of 1.8, 2.0, or 2.5 mg/kg/h for

9 h.

Page 79: The Processing of Amyloid Precursor Protein in the Central

61

Cell Culture and Media Standards.

Chinese Hamster Ovary (CHO) cells were grown in Ham’s F12 media for two passages.

Leucine-free Ham’s F12 media was obtained and supplemented with a mixture of labeled leucine

solution in the concentration of leucine in regular Ham’s F12 media. Media solutions prepared

ahead of time: 0%, 1.25%, 2.5%, 5%, 10%, and 20% labeled. At the third passaging of the cells,

all the cells were pooled and split evenly. Cells were reconstituted in one of the six prepared

media solutions and grown for 22 h. Media was collected, aliquoted, and stored at -80 ºC.

Immunoprecipitation of total sAPP.

Cerebrospinal fluid, 1 mL every 2 h (total of 18 samples), and 1 mL of CHO media each for 0%,

1.25%, 2.5%, 5%, 10%, and 20% 13C6-leucine label were thawed on ice. To each of the samples

we added 100 μL 5M Guanidine Tris (pH 8.0) and 10μl of protease inhibitor cocktail (40 μg/mL

aprotinin and 20 μg/mL leupeptin). After rinsing steps, 60μl of 8E5-bound bead slurry

(Sepharose beads coupled to 8E5 antibody (1 mg/mL) in 0.02% sodium azide in Phosphate

Buffered) was added to each sample. To immunoprecipitate all species of sAPP, we rotated the

sample-antibody bead mixture overnight (18 h) at 4 °C. At the end of this period, the

supernatant was collected and the beads were rinsed three times with 25 mM ammonium

bicarbonate (AmBic).

Digestion of sAPP into sAPPα and sAPPβ specific peptides.

To digest the sAPP into peptides that will be specific for sAPPα or sAPPβ, we reconstituted the

beads with 25 μL 10% acetonitrile (ACN) in 25 mM AmBic. We added 160 ng of Arginine-C,

Calcium acetate (final molarity : 2 mM), and Dithiothreitol (DTT) (final molarity : 2.5 mM), to

this slurry. Digestion took place on a shaker for 18 h at 37 °C. When digestion concluded, we

added formic acid (FA) to the bead slurry in order to stop digestion (final: 5% FA). We

Page 80: The Processing of Amyloid Precursor Protein in the Central

62

incubated for another 30 min at 37 °C on a shaker. The supernatant from bead slurry was pulled

off with a crimped gel loading tip and transferred into new Eppendorf tube. The peptide mix was

desalted with 1% FA using NuTip Carbon Tips (Glygen Corp., Columbia, MD). Samples were

eluted in 80% ACN/1% FA and evaporated in a rotary evaporator until no solution was left.

Samples were reconstituted in 10% ACN in NH4HCO3.

Peptide quantitation by liquid chromatography-mass spectrometry (LC-MS).

Samples (5 μL) were injected onto a C18aq Aquasil Zorbax (3 μm) column. The two peptides we

monitored were: PGSGL(L*)TNIKTEEVMDAEFR (sAPPα) and PGSGL(L*)TNIKTEEVKM

(sAPPβ) and they eluted from the column approximately two minutes apart.

Free Leucine Quantitation by gas-chromatography-mass spectrometry (GC-MS).

Cerebrospinal fluid 13C6-leucine enrichment was determined using gas capillary gas

chromatography mass spectrometry (GC-MS; Agilent 6890N gas chromatograph and Agilent

5973N mass selective detector) in negative chemical ionization mode as described previously

(Yarasheski et al., 1992, Bateman et al., 2007). As described in Wolfe et al. (2005), 13C6-leucine

enrichment was quantified as a tracer to tracee ratio (TTR).

Quantitation of labeled ratio.

The percentage of labeled sAPPα was determined by taking the ratio all of b- and y- product ion

intensities from the unlabeled and labeled peptide (PGSGL(L*)TNIKTEEVMDAEFR) of

APP444-592. The percentage of labeled sAPPβ was determined by taking the ratio all of b- and y-

product ion intensities from the unlabeled and labeled peptide (PGSGL(L*)TNIKTEEVKM) of

APP444-592. The ratio was computed using a custom Microsoft Excel spreadsheet with macros.

Page 81: The Processing of Amyloid Precursor Protein in the Central

63

The tandem MS quantification of labeled to unlabeled peptides that was used here is detailed in

Bateman et al. (2007a).

Quantitation of fractional synthesis rate and monoexponential slope fractional clearance

rate.

A fractional synthesis rate (FSR) and monoexponential slope fractional clearance rate (FCR) for

sAPP would have been quantified as previously reported (Wolfe et al., 2005) if the CSF labeling

curves were reliably quantifiable. Briefly, FSR would be calculated as the slope of the labeled

metabolite during the production phase (h: 5-14), divided by the CSF 13C6-leucine enrichment

which served as the precursor pool for APP synthesis. The standard equation from Wolfe et al.

(2005) was:

,

2 1

(Et2 - Et1)sAPPα,β ÷ (t2 - t1) is defined as the slope of the line during the labeling period, and

Precursor E is the ratio of labeled free leucine to unlabeled free leucine. The slope of the linear

regression would have been calculated from 5 to 14 h and divided by the average of CSF 13C6-

labeled leucine level during the infusion period.

Monoexponential slope FCR was computed by fitting the slope of the natural logarithm of the

clearance phase of the sAPP labeling curve (h: 24-36). The standard equation monoexponential

FCR equation was used (Wolfe et al., 2005):

ln ,

,/∆ 24 36

Page 82: The Processing of Amyloid Precursor Protein in the Central

64

Results

Representative chromatography elution profiles of labeled and unlabeled sAPPβ specific product

ions from a media standard sample are below (Figure 4.2.). Unlabeled and labeled peptides

elute from the HPLC column simultaneously, represented as a sharp, uncontaminated peaks that

are ideal for quantitation.

Figure 4.2. Chromatograms of labeled and unlabeled sAPPβ specific product ions from a media standard sample. sAPPβ unlabeled and labeled peptides eluted from the HPLC column simultaneously, and were ionized and fragmented in the mass spectrometer, producing characteristic product ions.

Page 83: The Processing of Amyloid Precursor Protein in the Central

65

Representative mass spectra of labeled and unlabeled sAPPβ specific product ions from a CHO

media standard sample are below (Figure 4.3.). Unlabeled and labeled product ions are formed

from peptides that elute from the HPLC column simultaneously, as represented by sharp, single

peaks, which are ideal for quantitation.

Figure 4.3. Mass spectra of labeled and unlabeled sAPPβ-specific b- and y- product ions from a CHO media standard sample. The retention time of the triply charged unlabeled (PGSGLTNIKTEEVKM of m/z = 645.41) and labeled (PGSGL*TNIKTEEVKM of m/z = 647.41) sAPPβ peptide was approximately 23.3 min.

Representative chromatography elution profiles of labeled and unlabeled sAPPα peptides as

manifested by specific product-ion peaks from a media standard sample are below (Figure 4.4.).

Unlabeled and labeled peptides elute from the HPLC column simultaneously, and they are

fragmented in the mass spectrometer to produce characteristic product ions. The trailing peak

indicates some contamination. Signal-to-noise ratio strength would need to be higher for

quantitation.

Page 84: The Processing of Amyloid Precursor Protein in the Central

66

Figure 4.4. Chromatography elution profiles of labeled and unlabeled sAPPα tryptic peptides as seen by peaks from a CHO media standard. sAPPα unlabeled and labeled peptides eluted from the HPLC column simultaneously, and were ionized and fragmented in the mass spectrometer to characteristic product ions.

Page 85: The Processing of Amyloid Precursor Protein in the Central

67

Representative mass spectra of labeled and unlabeled sAPPα specific product ions from a CHO

media standard sample are below (Figure 4.5.). Unlabeled and labeled product ions are formed

from peptides that elute from the HPLC column simultaneously, as represented by sharp, single

peaks, which are ideal for quantitation.

Figure 4.5. Mass spectra of labeled and unlabeled sAPPα-specific b- and y- product ions from a CHO media standard sample. The retention time of the quadruply charged unlabeled (PGSGLTNIKTEEVKMDAEFR of m/z = 638.98) and labeled (PGSGL*TNIKTEEVKMDAEFR of m/z = 640.48) sAPPα peptide was approximately 25.1 min.

Page 86: The Processing of Amyloid Precursor Protein in the Central

68

Figure 4.7. Chromatography elution profiles of labeled and unlabeled sAPPα and sAPPβ tryptic peptides as seen by peaks from a human CSF sample. sAPPα unlabeled and labeled peptides elute from the HPLC column simultaneously (forefront), and approximately 90 sec later than the elution of sAPPβ unlabeled and labeled peptides (background). This demonstrates that the two sAPP isoforms eluted from the HPLC column with a temporal separation long enough for the accurate detection of both isoforms in the same exact sample, following only one antibody-based immunoprecipitation protocol and one LC-MS run.

Page 87: The Processing of Amyloid Precursor Protein in the Central

69

Figure 4.8. Mass spectra of labeled (forefront) and unlabeled (background) sAPPβ-specific b- and y- product ions from a human CSF sample. The retention time of the triply charged unlabeled (PGSGLTNIKTEEVKM of m/z = 645.41) and labeled (PGSGL*TNIKTEEVKM of m/z = 647.41) sAPPβ peptide was approximately 29.6 min.

Figure 4.9. Mass spectra of labeled (forefront) and unlabeled (background) sAPPα-specific b- and y- product ions from a human CSF sample. The retention time of the quadruply charged unlabeled (PGSGLTNIKTEEVKMDAEFR of m/z = 638.98) and labeled (PGSGL*TNIKTEEVKMDAEFR of m/z = 640.48) sAPPα peptide was approximately 31.5 min.

Page 88: The Processing of Amyloid Precursor Protein in the Central

70

Based on our computer program, SILTmass, calculations of the labeled/unlabeled b- and y- ions

for sAPPβ and sAPPα, we generated standard curves and 36 h time courses for one participant

for each sAPP species (Figure 4.10-4.13). The human 36 h time course was not optimal, but it

was the best that we were able to generate on the LTQ mass spectrometer, which was not the

most favorable instrument for quantitation because it was not sensitive enough to detect minute

changes in labeling.

Figure 4.10. Linear sAPPβ-specific standard curve, using media from CHO cells. The % labeled/% unlabeled sAPPβ was determined by taking the ratio all the b- and y- product ion intensities from the labeled and unlabeled peptides (see Figure 4.3.) found in media with 13C6-leucine label at 0%, 1.25%, 2.5%, 5%, 10%, and 20%. The ratio was computed using a custom Microsoft Excel spreadsheet with macros. This representative sAPPβ standard curve indicated a high slope (m = 0.84) and high accuracy and linearity (r2 = 0.997). The y-intercept was slightly high (0, 0.007) which may be explained, in part, by the natural abundance of labeled leucine in the environment and partly by noisy signal at the lower end of standard curve.

Page 89: The Processing of Amyloid Precursor Protein in the Central

71

Figure 4.11. Linear sAPPα-specific standard curve, using media from CHO cells. The % labeled/% unlabeled sAPPα was determined by taking the ratio of all the b- and y- product ion intensities from the labeled and unlabeled peptides (see Figure 4.5.) found in media with 13C6-leucine label at 0%, 1.25%, 2.5%, 5%, 10%, and 20% . The ratio was computed using a custom Microsoft Excel spreadsheet with macros. This representative sAPPα standard curve indicated a high slope (m = 0.76) and high linearity (r2 = 0.996). The y-intercept was high (0, 0.02) which indicates a difficulty of the LTQ in accurately measuring the lower end of standard curve. Noisy signals in the MS spectra of those standards produced deceptively higher percentage label of peptide in those samples.

Page 90: The Processing of Amyloid Precursor Protein in the Central

72

Figure 4.12. A human CSF labeling curve for sAPPβ. The % labeled/% unlabeled sAPPβ was determined by taking the ratio of all of the b- and y- product ion intensities from the labeled and unlabeled peptides (see Figure 4.8.) found in CSF (collected from 0 to 36 h) of one human participant. The ratio was computed using a custom Microsoft Excel spreadsheet with macros. This human sAPPβ labeling curve was too noisy to produce quantifiable results. Fractional synthesis rate and fractional clearance rate could not be accurately determined from this data.

Page 91: The Processing of Amyloid Precursor Protein in the Central

73

Figure 4.13. A human CSF labeling curve for sAPPα. The % labeled/% unlabeled sAPPα was determined by taking the ratio of all of the b- and y- product ion intensities from the labeled and unlabeled peptides (see Figure 4.9.) found in CSF (collected from 0 to 36 h) of one human participant. The ratio was computed using a custom Microsoft Excel spreadsheet with macros. This human sAPPα labeling curve had extremely high background (CSF collected at t = 1 h already showed labeling at 3.25%) and was too noisy to produce quantifiable results. Fractional synthesis rate and fractional clearance rate could not be accurately determined from this data.

Discussion

Unfortunately, we were unable to determine turnover rates for the individual sAPP metabolites

using the above methods, because of the mass spectrometer’s lack of sufficient sensitivity. Even

though analysis of media samples produced linear standard curves, there was difficulty

producing a robust labeling CSF curve. In the CSF samples, the signal strengths of the peptides

of interest containing labeled leucine were close to the noise measurements. This was likely due

Page 92: The Processing of Amyloid Precursor Protein in the Central

74

in part to the length of our measured peptides (sAPPβ=15 amino acids; sAPPα=20 amino acids).

Peptides with much shorter amino acid sequences would be more favorable for more consistent

fragmentation using an LTQ. A more consistent fragmentation would, in turn, produce higher

signal strength for labeled and unlabeled peptides. Unfortunately the peptides following 8E5 IP

and protease digestion that we detected with the LTQ were long. The most optimal peptides for

signal strength were the peptides discussed in this chapter.

In order to find better peptides we developed a method for simultaneously measuring sAPPα and

sAPPβ from a single CSF sample following serial immunoprecipitation. This method was

developed in a non-human primate model and is outlined in the following chapter (5). It may

now be used to translate into human studies where it could help determine if and by how much β-

secretase activity is altered in AD. Future studies stemming from these results are further

explored in Chapter 6.

Page 93: The Processing of Amyloid Precursor Protein in the Central

75

Chapter 5 β-secretase inhibition on the effect of concentrations and kinetic of sAPPα, sAPPβ, and Aβ in rhesus macaques

Introduction

Amyloid Precursor Protein is a ubiquitous transmembrane protein involved in cell signaling,

development, and gene regulation. Compared to peripheral, non-central nervous system (CNS)

tissues, CNS APP processing demonstrates increased β-secretase activity (Irizarry et al., 2001;

Fukumoto et al., 2002) and different responses to γ-secretase modulation (Cook et al., 2010).

These fundamental differences in APP processing between the CNS and peripheral

compartments are not fully understood (Ortega et al., 2013); but may be important to inform the

design and development of Alzheimer’s disease (AD) therapeutics.

Amyloid Precursor Protein is first proteolyzed by either BACE or α-secretase generating

extracellular soluble APPβ (sAPPβ) or soluble APPα (sAPPα) respectively. Subsequent to β-

secretase cleavage, C99 may be cleaved by γ-secretase producing extracellular amyloid-β (Aβ)

and the APP intracellular domain (AICD). Recent reports (Portelius et al., 2010; Cook et al.,

2010) indicate that there is an alternate pathway of a tandem α-secretase and β-secretase

cleavage of APP which results in APP metabolites such as Aβ1-15/1-16. The relationship of

physiological α-secretase to β-secretase processing in the CNS is not fully understood; although

some insights from secretase inhibition studies of these enzymes suggest that APP can be

shunted to other pathways (Mattsson et. al., 2012).

Based on these and other studies, β-secretase inhibition has been proposed to decrease the

amount of APP processed into Aβ, and shunt APP to the α-secretase pathway. The predominant

Page 94: The Processing of Amyloid Precursor Protein in the Central

76

BACE in the CNS is BACE1, located mainly in the membranes of cellular compartments such as

endosomes and the trans-Golgi Network. BACE1 may be increased approximately 2-fold in

brains (Fukumoto et al., 2002; Yang et al., 2003; Li et al., 2004), or CSF, of AD patients

(Holsinger et. al., 2004; Holsinger et al., 2006; Verheijen et al., 2006; Zetterberg et al., 2008).

Brain penetrant BACE1 inhibitors capable of lowering CNS Aβ in rodent models and in non-

human primates (Takahashi et al., 2010; Malamas et al., 2010; Sankaranarayanan et al., 2008;

Truong et al., 2010.; Sankaranarayanan et al., 2009; Cumming, et al, 2012 Mandal et al, 2012,

Stamford, et al., 2012 ), have been identified and multiple BACE inhibitors have advanced into

early stages of human clinical trials (May et al., 2011; Egan et al., 2012, Forman et al., 2013).

In this study, we sought to determine the kinetic behavior of APP metabolites and the

relationship between the α-secretase and β-secretase pathways during BACE1 inhibition in a

non-human primate model. We utilized Stable Isotope Labeling Kinetics (SILK) in combination

with a novel high affinity, selective and centrally-active BACE inhibitor to determine the

production and turnover rates of APP metabolites. The use of labeling kinetics provides a more

sensitive determination of production changes (Bateman et al., 2009) by distinguishing the newly

generated metabolites in the cerebrospinal fluid from those that previously existed.

Methods and Experimental Design

Cisterna magna ported rhesus monkey model.

Animal use procedures in this study were reviewed and approved by the Institutional Animal

Care and Use Committee at Merck Research Laboratories at West Point. They conform to the

Guide for the Care and Use of Laboratory Animals (Institute of Laboratory Animal Resources,

Page 95: The Processing of Amyloid Precursor Protein in the Central

77

National Research Council, 1996). The catheterization procedures are as described previously

(Gilberto et. al., 2003; and Cook et al. 2010).

13C6-Leucine infusion protocol and sample collection.

At 48 h prior to administration of compound or vehicle, monkeys were restricted to a diet

consisting solely of fruits and vegetables. Their diet lacked protein until after the 12 h time-point

of the study (13 h post dosing of the BACE1 inhibitor). 13C6-leucine infusion procedures

followed by same protocol as described previously for the low dose of leucine (Cook et. al.,

2010). Briefly, a 4 mg/mL 13C6-leucine primed bolus infusion was administered intravenously

for 10 minutes, followed by a steady infusion of 4 mg/mL/h for 12 h. Baseline CSF and blood

samples were collected at 22, 20, and 1 h prior to the onset of the 10 minute primed leucine

infusion. After the leucine bolus infusion, CSF and blood samples were collected at the

following hours post leucine administration: 2, 4, 6, 8, and 12, 15, 18, 21, 24, 27, 30, 48, 54, 57,

72, and 144. For each time-point, 1500 μL CSF and 2000 μL whole blood were collected into

low-binding polypropylene tubes (Axygen). The blood was collected on K2 EDTA BD

vacutainer tubes, spun, and 800 μL of plasma was collected and placed into Axygen tubes. CSF

and plasma samples were separated into aliquots of lesser volume (into Axygen or PK tubes) and

placed immediately on dry ice, then stored in a -70 °C freezer until transfer to various

laboratories for analyses. Prior to analyses, samples were stored in a -80 °C freezer.

β-secretase inhibitor study protocol.

The Merck β-secretase inhibitor, MBI-5, was employed in a 4-way crossover randomized design

administering one of three doses of MBI-5: 10, 30, and 125 mg/kg , or vehicle (0.4%

methylcellulose) to cisterna magna ported (CMP) rhesus monkeys (n=5 male rhesus monkeys,

Page 96: The Processing of Amyloid Precursor Protein in the Central

78

10-13 years old, 8-11.7 kg). Vehicle or MBI-5 (10, 30, or 125 mg/kg) was administered orally to

conscious rhesus monkeys 1 h prior to initiating the primed 12 h 13C6-leucine constant infusion

(4 mg/kg + 4 mg/kg/h). Both CSF and plasma samples were collected as mentioned under 13C6-

leucine infusion protocol to assess the pharmacokinetics and pharmacodynamics of this

compound by quantifying MBI-5 concentrations, absolute concentrations of sAPPα, sAPPβ,

Aβ40, and Aβ42 (as measured by ELISA), or 13C6-leucine labeled sAPPα, sAPPβ, and total Aβ,

and free 13C6-leucine enrichment.

Ki or IC50,

nM*

Soluble BACE1 10 1

Soluble BACE2 12 2

Cathepsin D 2,700 600

Cathepsin E 26,600 11,000

Pepsin ~70,000

Renin 9,000

HEK293 APPswe/lon A40 IC50 72 5

HEK293 APPswe/lon A42 IC50 24 6

HEK293 APPswe/lon sAPP IC50 230

Table 5.1. In vitro pharmacological profile of the BACE inhibitor, MBI-5. In vitro enzyme assays utilized purified recombinant human enzymes, except for Cathepsin D which was purified from human liver. Data shown are mean SEM.

Page 97: The Processing of Amyloid Precursor Protein in the Central

79

Cell Culture and Media Standards.

Human H4 neuroglioma cells stably transfected with human APP751 (H4-APPwt) (courtesy of T.

E. Golde Laboratory, UF-Gainesville) were grown in Dulbecco's Modified Eagle Medium

(DMEM) for two passages. Leucine-free DMEM was obtained and supplemented with a mixture

of labeled leucine solution so that the total final leucine concentration was the same as in

standard DMEM, with leucine levels unaltered (105 mg/L). The final media solutions had: 0%,

1.25%, 2.5%, 5%, 10%, or 20% 13C6-leucine. Labeled DMEM was supplemented with 1:50 B-

27 and 1% each of Penicillin-Streptomycin and Zeocin. At the third passaging of the cells, cells

were pooled and split evenly. The H4-APPwt cells were reconstituted in one of the six prepared

media solutions and cultured for 24 h to allow for sufficient incorporation of label into proteins,

such as APP. Following this incubation with labeled DMEM, the media solutions were collected

and apportioned into Axygen tubes in 1 mL aliquots and immediately frozen and stored at -80 °C

until ready to use.

Aβ, sAPPβ, and sAPPα ELISA Protocols.

The assays for CSF Aβ40, Aβ42, sAPPα, and sAPPβ measurement were described previously

(Sankaranarayanan et al., 2009, Wu et al., 2011) with some modification in dilution factor.

Briefly, CSF was diluted with 3% BSA/PBS at 1:3 for Aβ42, 1:10 for Aβ40, and 1:80 for both

sAPPβ and sAPPα. Diluted CSF (100 μL) was then used for each analyte measurement. The

concentration was calculated based on each standard curve. The absolute concentrations of total

Aβ reported herein are derived by summating the absolute concentrations measured by the two

individual Aβ ELISAs: Aβ40 and Aβ42.

Page 98: The Processing of Amyloid Precursor Protein in the Central

80

Immunoprecipitation and digestion of sAPPβ.

Immunoprecipitation (IP) of sAPPβ from CSF was performed using a rabbit monoclonal

antibody that can specifically recognize the KM-neo-epitope of sAPPβ created following the

cleavage of APP by β-secretase. The generation and specificity characterization of the prepared

anti-sAPPβ neo-epitope antibody Mrk-61 was previously described (Wu et al. 2011; Wu et al,

2012). Mrk-61 binds exclusively with sAPPβ but not to sAPPα in both western blot and direct

immunoassay with coated each recombinant protein. The purified rabbit monoclonal anti-Mrk-61

antibody was conjugated with CNBr-activated SepharoseTM 4B beads (Amersham Biosciences,

Sweden) according to the manufacturer’s protocol. The activity of the SepharoseTM 4B

conjugated Mrk-61 antibody was evaluated in immunoprecipitation efficiency with normal

rhesus CSF following overnight beads incubation and the sAPPβ level in CSF after IP was

measured with a sensitive sAPPβ ELISA (Wu et al., 2011). The characterized Mrk-61-

SepharoseTM 4B beads were then reconstituted in a 50% slurry of 0.02% sodium azide in

phosphate buffered saline (PBS) and stored at 4 °C until use.

From each time-point, 500 μL of CSF, along with H4-APPwt media standards (0%, 1.25%, 2.5%,

5%, 10%, 20%), was diluted 1:1 with 500 μL PBS. Protease inhibitors (40 μg/mL aprotinin and

20 μg/mL leupeptin) (Calbiochem/EMD Millipore) were added to each sample at a volume of

10μL, followed by the addition of 50μL of Mrk-61 antibody

Samples were rotated for approximately 22 h at 4 °C. At the conclusion of the rotation, samples

were centrifuged (16,837 x g) and 925 μL of the supernatant was collected into new

polypropylene tubes and stored at 4 °C for approximately 1.5 h until ready for the sAPPα/Aβ

immunoprecipitation protocol. The Mer61-3 beads were washed three times with 25mM

ammonium bicarbonate (AmBic), with centrifugation between washes. The beads wash was

Page 99: The Processing of Amyloid Precursor Protein in the Central

81

aspirated after the final rinse and 100 μL of neat formic acid was immediately added to each

sample to elute the sAPPβ from the antibody-bead complex. Samples were left for 10 minutes at

25 °C and centrifuged. The formic acid supernatant was transferred to a new polypropylene tube

and evaporated in a rotary evaporator at 37 °C for 30 minutes. The dried samples were

reconstituted with 25 mM AmBic, and 5ng sequencing-grade Metalloendopeptidase (Lys-N)

(Seikagaku Corporation/Associates of Cape Cod Inc.) in 25 mM AmBic was added. Extracts

were digested for approximately 20 h on a shaker at 37 °C and transferred into autosampler vials.

Immunoprecipitation and digestion of sAPPα and Aβ.

Prior to study onset, a mouse monoclonal antibody W0-2 (EMD Millipore, Billerica, MA)

(directed against Aβ4-10) and a mouse monoclonal antibody HJ5.1 (Washington University in St.

Louis) (directed against Aβ13-28) were covalently bound to CNBr SepharoseTM 4B beads

according to the manufacturer’s instructions, then stored in a 50% slurry of 0.02% PBS azide at 4

°C prior to use. The sAPPα/Aβ IP protocol was optimized for maximum sAPPα/Aβ signal from

the liquid chromatography-mass spectrometer system. The above-mentioned CSF and media

standard supernatants were taken from 4 °C and put on ice. To each sample 45 μL W0-2

antibody slurry and 60 μL HJ5.1 antibody slurry were added. Samples were rotated, rinsed,

eluted, and reconstituted in the same way as with the Mer61-3 antibody.

Quantitation of peptides by liquid chromatography-mass spectrometry (LC-MS).

Peptides specific to sAPPα and sAPPβ (KYL(L*)ETPGDENEHAHFQ), and Aβ

(KL(L*)VFFAEDVGSN) were analyzed on a Thermo-Finnigan LTQ equipped with a New

Objective nanoflow ESI source. The peptides were separated by RP HPLC using an Eksigent

2D-LC nanoflow pump operating in 1D mode at a flow rate of 200 nL/min. Sample (5 μL) was

Page 100: The Processing of Amyloid Precursor Protein in the Central

82

injected onto a New Objective picofrit column packed to 12 cm with 5 μm Magic C18aq packing

material (Michrom). Mobile Phase A contained 0.1% formic acid (FA) in water and Mobile

Phase B was 0.1% FA in acetonitrile (ACN).

Page 101: The Processing of Amyloid Precursor Protein in the Central

83

Figure 5.1. Rhesus macaque APP sequence. The rhesus macaque APP sequence has approximately 91% homology with human APP. Non-homologous regions are shown in purple. The non-homologous amino-terminal sequence is absent in humans. The human APP sequence begins with “MLP” at its N-terminus. The second non-homologous sequence found in the mid-domain of APP is such in humans: AMSQSLLKTTQEPLARDPVKLPTTAASTPDAVDKYLETPGDENEHAHFQKAKERLEAKHRERMSQVMREWEEAERQAKNLPKADKKAVI. Various regions of the APP sequence pertinent to this study are indicated, namely the amino acid sequences that this study’s antibodies are directed against, as well as peptides used in MS quantification. Note that even though the sAPPα and sAPPβ MS peptide used in this study’s quantification is located in a region non-homologous to human APP, the actual peptide’s sequence (KYLETPGDENEHAHFQ) is conserved among species.

Free Leucine Quantitation by gas-chromatography-mass spectrometry (GC-MS).

Plasma 13C6-leucine enrichment was determined using gas capillary gas chromatography mass

spectrometry (GC-MS; Agilent 6890N gas chromatograph and Agilent 5973N mass selective

detector) in negative chemical ionization mode as described previously (Yarasheski et al., 1992;

Bateman et al., 2007; Cook et al., 2010). As described in Wolfe et al. (2005), 13C6-leucine

enrichment was quantified as a tracer to trace ratio.

Page 102: The Processing of Amyloid Precursor Protein in the Central

84

Calculation of labeled APP metabolite ratio.

The percentage of labeled metabolite was determined by taking the ratio of b- and y- product ion

intensities from the unlabeled metabolite peptide and the labeled peptide. For Aβ, the peptide

quantified was KL(L*)VFFAEDVGSN. For sAPPα and sAPPβ, the peptide was

KYL(L*)ETPGDENEHAHFQ. Mole fraction labeled was calculated.

Calculations of fractional synthesis rate and monoexponential slope fractional clearance rate.

For each metabolite a fractional synthesis rate (FSR) and monoexponential slope fractional

clearance rate (FCR) was quantified as previously reported (Wolfe et al., 2005). Briefly, FSR

was calculated as the slope of the labeled metabolite during 2-8 h, divided by the plasma 13C6-

leucine enrichment which serves as the precursor pool for APP synthesis. Monoexponential

slope FCR was computed using the natural logarithm of each labeled metabolite during the

clearance phase of labeling, notably 18-30 h.

Calculations of newly generated APP metabolites and Area Under the Curve (AUC) analysis.

Similarly to Bateman et al. (2009) calculation of newly generated APP metabolites was

performed by determining the product of the percentage of metabolite labeled at a particular

time-point (established by LC-MS) and the absolute concentration of metabolite (determined by

ELISA) at that particular time-point.

Page 103: The Processing of Amyloid Precursor Protein in the Central

85

Figure 5.2. Rhesus macaque sample processing study design. Blood and CSF are collected from rhesus macaques for biochemical assessments. Blood is collected, and the plasma portion is used for pharmacokinetic analysis and free labeled leucine measurements by Gas Chromatography-Mass Spectrometry (GC-MS), to determine the leucine enrichment for which to normalize all APP metabolite labeling curves. Cerebrospinal fluid is collected through the cistern magna port. A portion of the CSF is used for ELISA measurements of absolute concentrations of sAPPα, sAPPβ, Aβ40, Aβ42. The remainder of the CSF is employed for SILK analyses by Liquid Chromatography-Mass Spectrometry (LC-MS). To isolate the particular metabolites, a serial immunoprecipitation protocol is used. Media or CSF is first immunoprecipitated with Mrk-61, a KM neo-epitope specific antibody against sAPPβ. After immunoprecipitation, the supernatant is collected and immunoprecipitated a second time with a cocktail of two antibodies: W0-2 (recognizing Aβ4-10, which is within the sAPPα sequence) and HJ5.1 (recognizing Aβ13-28). The sAPPβ/Mrk-61 bead complex from the first immunoprecipitation is digested by Lys-N (proteolytically cleaving the protein on

Page 104: The Processing of Amyloid Precursor Protein in the Central

86

the N-terminus of Lysine residues) and the unlabeled and labeled peptides KYL(L*)ETPGDENEHAHFQ are quantified by LC-MS to determine percentage labeled sAPPβ at each time point and produce an sAPPβ labeling curve. Following the completion of the second immunoprecipitation, the sAPPα/W0-2 bead complex and the Aβ/HJ5.1 bead complex are proteolytically cleaved by Lys-N to produce the unlabeled and labeled peptides KYL(L*)ETPGDENEHAHFQ and KL(L*)VFFAEDVGSN, respectively. These peptides are quantified by LC-MS to determine percentage labeled sAPPα and Aβ at each time point and to produce sAPPα and Aβ labeling curves.

Statistical Analyses.

For each analyte, area under the curve from -1 to 57 h (AUC-1-57) was calculated using the

trapezoidal rule. This time duration was chosen due to the majority of the drug effect and

labeling occurring during this time period. To estimate the effect of each active treatment versus

vehicle, a linear mixed effects model with fixed effects for treatments and random effects for

monkeys was fit to the log (base 2) of AUC-1-57. Estimates of mean differences from vehicle

were back transformed from the log scale to yield percent differences. Exposure was measured

using the area under the concentration-time curve from 0 to 59 h. Estimation of the linear

relationship between each analyte and exposure was performed using a mixed effects model like

the one above, except with the treatment effects replaced by a slope and intercept for exposure.

Results

Macaque Aβ FSR is similar to human FSR & monoexponential slope FCR much higher than human’s.

Turnover of sAPPα and sAPPβ was slower than Aβ in the CNS. The average kinetic curves of

sAPPα, sAPPβ, and Aβ in the vehicle-treated group were evaluated (Figure 5.3.A), comparing

parameters of the three labeling curves maximum mole fraction labeled, time to peak labeling,

and AUC-1-57 analyses. Soluble APPα achieved a maximum of 0.8 ± 0.01 mole fraction labeled,

Page 105: The Processing of Amyloid Precursor Protein in the Central

87

while sAPPβ exhibited a similar labeling maximum (0.8 ± 0.7 mole fraction labeled; p = 0.8)

(Figure 5.3.B). Amyloid-β had a significantly higher peak of mole-fraction labeled (9.7 ± 0.6%

SD) compared with both sAPPα (*p = 0.01) and sAPPβ (*p = 0.01) (Figure 5.3.B). The labeling

peak time-points for sAPPα and sAPPβ were identical (t = 16.2 ± 1.6 h (SD), p = 1.0), with the

Aβ labeling peak preceding both (t = 14.4 ± 1.3 h (SD)) without reaching statistical significance

(p = 0.07) (Figure 5.3.C). There was no difference among metabolites’ AUC-1-57 values (sAPPα

AUC-1-57 = 1.9 ± 0.3 (SD); sAPPβ AUC-1-57 = 1.9 ± 0.2 (SD); Aβ AUC-1-57 = 2.0 ± 0.3 (SD), p =

0.9) (Figure 5.3.D).

To quantify the turnover rates of the APP metabolites, FSR was calculated to estimate

production rate and monoexponential slope FCR was calculated to estimate clearance rate. By

definition, FSRs and monoexponential slope FCRs are only valid when measured in a steady-

state system (Wolfe et. al., 2005), thus kinetic analyses of sAPPα, sAPPβ, and Aβ were

performed exclusively using data collected from the vehicle-treated monkeys. The mean sAPPα

FSR (3.8 ± 0.4%/h (SD)) as measured in the CSF was faster than the mean sAPPβ FSR (3.1 ±

0.5%/h (SD), *p = 0.01) (Figure 5.3.E). The mean Aβ FSR was 7.3 ± 0.9%/h (SD) (Figure

5.3.E) and was comparable to the mean human Aβ FSR (dashed line) of 7.6%/h reported

previously (Bateman et. al., 2006). The mean Aβ FSR was significantly faster than that of either

sAPPβ (***p < 0.0005) or sAPPα (**p < 0.001).

Mean FCRs for sAPPα and sAPPβ were 7.8 ± 0.7%/h (SD) and 8.3 ± 1.4%/h (SD), respectively

(Figure 5.3. F). Mean Aβ FCR was 13.0 ± 1.9%/h (SD) (Figure 5.3.F) and was approximately

twice as fast as mean human Aβ FCR (dashed line) previously reported (Bateman et. al., 2006).

The FCRs of all three metabolites were significantly different (p < 0.002) as assessed by a one-

Page 106: The Processing of Amyloid Precursor Protein in the Central

88

way repeated measures ANOVA. Aβ FCR was significantly faster than FCR of either sAPPβ

(*p < 0.02) or sAPPα (*p < 0.005). The FCRs for sAPPβ and sAPPα were not significantly

different from one another (p = 0.6).

Figure 5.3. CNS metabolism of APP metabolites in vehicle-treated rhesus monkeys. A, 13C6-leucine labeling curve profiles of vehicle-group-averaged (n=5) sAPPα, sAPPβ, and Aβ. B, Maximum mole fraction labeled for each metabolite was determined. Aβ reached a significantly higher maximum labeling as compared to sAPPα (paired t-test, *p = 0.01) and sAPPβ (paired t-test, *p = 0.01). C, The time-point at which maximum labeling occurred was not statistically significant among metabolites. D, AUC-1-57 values for each metabolite did not differ. E, The means of FSRs of APP metabolites were significantly different from one another (Repeated Measures ANOVA, ***p < 0.0001). The dashed line in the Aβ column indicates the previously reported human FSR of Aβ (7.6%/h) (Bateman et. al., 2006). F, The mean of the monoexponential slope FCR of Aβ was significantly higher than the monoexponential slope FCRs of both sAPPα (paired t-test, **p = 0.004) and sAPPβ (paired t-test, *p = 0.01). There was no statistically significant difference between sAPPα and sAPPβ monoexponential slope FCRs. The dashed line in the Aβ column indicates previously reported human FCR of Aβ (8.3%/h) (Bateman et. al., 2006).

Page 107: The Processing of Amyloid Precursor Protein in the Central

89

BACE1 inhibitor dose-dependently decreased sAPPβ and Aβ in rhesus monkey CNS.

Labeling curves of Aβ and sAPPβ, as well as absolute concentrations of both analytes measured

by ELISA, decreased dose-dependently in the presence of a BACE1 inhibitor. Concentrations of

newly generated metabolites were calculated by taking the product of percentage labeled and

absolute concentration of a given metabolite at each time-point. Like the labeling profiles and

absolute concentrations, the newly generated Aβ and sAPPβ reflect a dose-dependent decrease in

the presence of a BACE1 inhibitor. Area under the curve for Aβ labeling for each monkey after

BACE1 inhibition were normalized to that monkey’s vehicle labeling curve and averaged. Dose

dependent decreases were seen in both SILK labeled and ELISA AUC-1-57 concentrations of

both Aβ and sAPPβ.

Page 108: The Processing of Amyloid Precursor Protein in the Central

90

Figure 5.4 Effects of a BACE1 inhibitor on SILK labeling, ELISA concentrations, and newly generated Aβ (A-C), sAPPβ (D-F), and sAPPα (G-I) in CNS of rhesus monkeys. A, D, G) SILK Mole fraction labeled Aβ and sAPPβ decreases dose-dependently with β-secretase inhibitor, and mole fraction labeled sAPPα indicated no difference among vehicle and drug groups (measured by LC-MS). B, E, H) ELISA concentrations of Aβ and sAPPβ decreased dose-dependently and absolute concentrations of sAPPα increased dose-dependently with a BACE1 inhibitor (measured by ELISA). C, F, I) Newly generated Aβ and sAPPβ decreased dose-dependently and newly generated sAPPα increased dose-dependently with a BACE1 inhibitor (measured as product of LC-MS labeling and ELISA absolute concentrations at each time-point).

A

-20 0 20 40 60 80 10

012

014

0

0.02

0.04

0.06

0.08

0.10

0.12

Vehicle

A

10 MPK30 MPK

Hour of Collection

125 MPK

-20 0 20 40 60 80 10

012

014

0

pM

A

-20 20 40 60 80 10

012

014

0-200

200

400

600

800

1000

C

Hour of Collection

New

ly G

ener

ated

A

sAPP

-20 0 20 40 60 80 10

012

014

0

0.02

0.04

0.06

0.08

0.10

D

Hour of Collection-2

0 0 20 40 60 80 100

120

140

pM

-20 20 40 60 80 10

012

014

0

New

ly G

ener

ated

sA

PP

sAPP

-20 0 20 40 60 80 10

012

014

0

0.02

0.04

0.06

0.08

0.10

0.12

G

Hour of Collection-2

0 0 20 40 60 80 100

120

140

pM

-20 20 40 60 80 10

012

014

0

New

ly G

ener

ated

sA

PP

Page 109: The Processing of Amyloid Precursor Protein in the Central

91

We calculated the percent difference of each analyte from vehicle to each drug treatment group.

Mean values from each MBI-5 dosage group for each analyte were compared to the mean value

of that analyte’s vehicle group. The mole fraction labeled Aβ AUC-1-57 indicated a dose-

dependent decrease to approximately 50% at the highest BACE1 inhibitor dose of 125 mg/kg

(Figure 5.5.A, Table 5.1.); approximately 10% of the total Aβ is labeled (see above). Area

under the curves for ELISA concentrations of Aβ normalized to vehicle showed a dose-

dependent response to about 33%, a greater extent than measured by SILK (Figure 5.5.B, Table

5.1.). Stable isotope labeling kinetics mole fraction labeled sAPPβ AUC-1-57 indicated a dose-

dependent decrease to a lesser extent than Aβ: to approximately 70% of vehicle values at the

highest BACE1 inhibitor dose (Figure 5.5.C, Table 5.1.), approximately 8% of the total sAPPβ

is labeled. Similarly to Aβ, AUC-1-57 values for absolute concentrations of sAPPβ measured by

ELISA normalized to vehicle indicated a dose-dependent decrease to a greater extent

(approximately 38% at the highest inhibitor dose) compared to SILK (Figure 5.5.D, Table 5.1.).

Page 110: The Processing of Amyloid Precursor Protein in the Central

92

Figure 5.5. Effects of BACE1 inhibition on APP metabolites’ AUC-1-57. Results are represented as percentage change from vehicle AUC-1-57. Each line represents a particular

Vehicl

e

10M

PK

30M

PK

125 M

PK

% D

iffe

ren

ce f

rom

Veh

icle

Vehicl

e

10M

PK

30M

PK

125 M

PK

% D

iffe

ren

ce f

rom

Veh

icle

Vehicl

e

10M

PK

30M

PK

125 M

PK

% D

iffe

ren

ce f

rom

Veh

icle

Vehicl

e

10M

PK

30M

PK

125 M

PK

% D

iffe

ren

ce f

rom

Veh

icle

Vehicl

e

10M

PK

30M

PK

125 M

PK

% D

iffe

ren

ce f

rom

Veh

icle

Vehicl

e

10M

PK

30M

PK

125 M

PK

% D

iffe

ren

ce f

rom

Veh

icle

Page 111: The Processing of Amyloid Precursor Protein in the Central

93

monkey. A, C, E) Mole fraction labeled Aβ and sAPPβ AUC-1-57 were decreased dose-dependently to a maximum of approximately 48% and 70% of vehicle at the highest dose, respectively. Analyses of mole fraction labeled sAPPα AUC-1-57 indicated that dosing groups did not significantly differ from the vehicle-treated group. B, D, F) AUC-1-57 values for ELISA absolute concentrations of Aβ and sAPPβ were decreased dose-dependently to a maximum of approximately 33% and 38% of vehicle at the highest dose, respectively. Analyses of AUC-1-57 of ELISA absolute concentrations of sAPPα presented a dose-dependent increase to approximately 131% of vehicle at the highest dose.

Table 5.2. Effects of BACE1 inhibition on APP metabolites’ AUC-1-57 from SILK and ELISA assays. Values provided represent the percentage difference of a particular metabolite’s mean AUC-1-57 in each MBI-5 dosage group from that metabolite’s mean AUC-1-57 in the vehicle group.

BACE1 inhibition had no effect on fraction labeled sAPPα but dose-dependently increased total sAPPα concentrations.

The labeling curves of sAPPα for vehicle and all three BACE1 inhibitor groups indicated no

differences in sAPPα fraction labeled (repeated measures ANOVA, p = 1.0) (Figure 5.4.G).

Concentrations measured by sAPPα ELISA indicated a dose-dependent increase with BACE1

Dosage of BACE Inhibitor

SILK Mole Fraction Labeled

ELISA Absolute Concentration

10 mg/kg 77.8 ± 6.4 % 111 ± 12.9%30 mg/kg 70.8 ± 7.0% 58.6 ± 12.3%125 mg/kg 47.5 ± 6.2% 33.2 ± 8.2%

10 mg/kg 84.4 ± 4.4% 88.1 ± 2.1%30 mg/kg 78.3 ± 3.4% 63.0 ± 2.7%125 mg/kg 69.9 ± 5.1% 38.3 ± 4.4%

10 mg/kg 99.6 ± 9.3% 107.8 ± 7.4%

30 mg/kg 101.0 ± 3.2% 113.8 ± 5.8%125 mg/kg 101.5 ± 6.6% 131.4 ± 6.9%

Aβ (mean, SEM)

sAPPβ (mean, SEM)

sAPPα (mean, SEM)

Page 112: The Processing of Amyloid Precursor Protein in the Central

94

inhibition (Figure 5.4.H). Newly generated sAPPα (the product of steady state concentration

multiplied by 0.8% fraction labeled sAPPα), demonstrated dose dependent increases, but to a

lesser degree than the observed sAPPβ decrease (Figure 5.4. I).

Stable isotope labeling kinetics mole fraction labeled sAPPα AUC-1-57 indicated a lack of

significant difference among dosage groups (Figure 5.5.E, Table 5.1.). However, AUC-1-57 for

absolute concentrations of sAPPα normalized to vehicle indicated a modest dose-dependent

increase to approximately 131% at the highest dose (Figure 5.5.F, Table 5.1.).

Discussion

This study provided further evidence that CNS production of Aβ in rhesus monkeys is similar to

human. A prior study from our group (Cook et al., 2010) demonstrated this similarity between

rhesus monkeys and human primates (Bateman et. al., 2006). To date, metabolism of other APP

fragments have not been assessed in rhesus monkeys, although Dobrowolska et al. (2008)

demonstrated that total soluble APP (sAPP) was metabolized approximately two-fold slower

than Aβ in young, healthy human participants. To our knowledge, there are no prior studies

investigating metabolism of sAPPα and sAPPβ. We report that similar to total sAPP metabolism

in humans, metabolism of sAPPα and sAPPβ in rhesus monkeys was equal to one another and

was half the rate of Aβ metabolism. This correspondence between sAPP metabolisms in both

species further indicated that rhesus non-human primates are an appropriate preclinical model for

human CNS APP processing.

Herein, we report the first SILK BACE1 inhibitor study to evaluate in vivo APP metabolite

kinetics. As expected, there was a notable and dose-dependent decrease in Aβ and sAPPβ in the

Page 113: The Processing of Amyloid Precursor Protein in the Central

95

presence of the BACE1 inhibitor, indicating the drug hit its intended target. The finding that

there was a greater percentage reduction in Aβ compared with sAPPβ in the presence of the

inhibitor was consistent with Aβ having a faster turnover rate at steady state (in the vehicle-

treated group). Owing to conflicting results in prior studies, it was unclear whether the kinetics

of α-secretase processing of APP to sAPPα would also be altered. In cell culture assays where

BACE inhibitors were applied to the cells, APP appeared to be shunted down the α-secretase

pathway, resulting in increased sAPPα product. This contrasts with human studies that seemed

to indicate a non-competitive relationship between the α-secretase and β-secretase APP pathways

(Lewczuk et al. 2010, Gabelle et al. 2010, Alexopolous et al. 2011, Dobrowolska, in

preparation). These in vitro studies had suggested that, pharmacological intervention may alter

normal APP processing such that decreased BACE activity diverts APP into the α-secretase

pathway, producing more sAPPα. However, the in vitro setting is quite different from a whole

organism, in which our experiments are set. We sample from a site quite distal from the initial

labeling of leucine and its incorporation into APP. Not only is APP trafficked within the cell,

but its metabolites may move through many compartments in the macaque CNS between release

into the ECF from a neuron and collection within CSF from a cisterna magna port. An in vitro

labeling experiment would not have this added dimension and might not be predictive of what

we may see in vivo. Indeed, this study does not support the in vitro studies as there is no increase

in newly generated sAPPα (SILK study) during the acute period of BACE1 inhibition used in

this study. However, there was a partial dose dependent increase in sAPPα concentration

(ELISA study) at the highest doses, but this is confounded by increases in sAPPα concentrations

in the vehicle treated group. Thus, the ELISA sAPPα results are inconclusive and the SILK

sAPPα results show no significant difference at all between dose groups. Another caveat of the

Page 114: The Processing of Amyloid Precursor Protein in the Central

96

sAPPα ELISA measures is that sAPPα did not increase to same extent that sAPPβ decreased, and

there is no rebound of sAPPβ over baseline following the resumption of BACE activity as the

drug levels fall with metabolism. Thus, a build-up of APP substrate may be degraded through

mechanisms other than traditional α-secretase cleavage activities to account for these

observations. For example, recent immunoprecipitation-studies indicate that alternative α- and

β- secretase processing may occur in vivo in the human CNS, i.e. sAPPαQ686 (sAPPα’),

sAPPαK687, sAPPβM671, and sAPPβY681 (sAPPβ’) from the APP770 splice variant (Brinkmalm et

al., 2013). Conversely, the additional APP substrate may undergo lysosomal degradation.

Measurements using SILK would not reflect APP shunted toward lysosomes. For a model and

further discussion on how the BACE1 inhibitor study in rhesus macaques may be reconciled with

the human studies discussed in Chapter 2, please refer to Chapter 6.

The isotopic labeling profiles of Aβ and sAPPβ were reduced in a dose-dependent manner with

increasing doses of BACE inhibitor, concomitant with decreased CSF concentrations of these

proteins measured by ELISA, whereas the isotopic labeling profile of sAPPα was virtually

unaltered despite a dose-dependent increase of approximately 20-30% in the ELISA

concentration AUC-1-57 in the highest dose group (Figure 5.5., Table 5.1.). This apparent

dichotomy can be explained based on simulations using a non-steady state compartmental model

(BWP, unpublished observations). BACE inhibitor was introduced at around the same time as

the tracer was administered so that the system transitioned from a steady state to a non-steady

state at the time tracer entered the system. We may presume that the production of labeled Aβ

and sAPPβ was immediately decreased in a dose-dependent manner. The newly synthesized

peptides may be transferred into pools of pre-existing CSF peptides that initially remain at basal

concentrations, since turnover of the CSF pools must occur before CSF concentrations decrease

Page 115: The Processing of Amyloid Precursor Protein in the Central

97

in response to BACE inhibition. The decreased appearance of labeled peptides in CSF preceded

the decrease in CSF concentration, thus resulting in a dose-dependent decrease in the isotopic

enrichment profile. BACE inhibition should cause the amount of APP to increase, which leads to

increased production of sAPPα due to mass action. Thus, increasing amounts of labeled sAPPα

should appear in CSF in a dose-dependent manner. However, the concentration of CSF sAPPα

will increase concomitant with and in proportion to the increased appearance of labeled sAPPα,

and thus there is little impact on the ratio of labeled to unlabeled peptide (i.e., isotopic

enrichment). The differential sensitivity of the enrichment profiles to acute inhibition versus an

increase of peptide synthesis results because the peptides are sampled from a downstream

location (CSF) that is remote from the site of BACE activity (brain).

This study has the benefit of measuring changes in APP metabolites by two separate methods:

employing SILK and mass spectrometry, as well as measuring absolute concentrations by

ELISA. SILK is a quite robust and reliable method of quantifying de novo proteins changing

over time following a labeling pulse, whereas the ELISA assays measure the steady state

concentrations of a particular metabolite. SILK is more specific to newly generated proteins and

more sensitive to detect changes at earlier time points in a secretase inhibitor setting (Bateman et

al., 2009; Cook et al., 2010) where protein levels may border near the lower level of detection or

ELISA variance masks relatively smaller changes in concentrations.

The CNS kinetic rates of APP metabolites are an important physiologic measure that is tightly

regulated and consistent across primates in studies to date. Modulation of APP processing is a

key approach in AD therapeutic development with β-secretase inhibition being an attractive

target. This is consistent with recent strong genetic evidence of a protective factor (Jonsson et

al., 2012) in individuals with a mutation at the BACE cleavage site of APP that prevents

Page 116: The Processing of Amyloid Precursor Protein in the Central

98

development of AD. The CNS in vivo study reported here indicates that while BACE inhibition

modulated APP processing in a predictable and dose-dependent fashion, it has also provided a

novel finding into the balance of BACE and α-secretase APP processing following BACE

inhibition.

Page 117: The Processing of Amyloid Precursor Protein in the Central

99

Chapter 6 General Conclusions and Future Directions

The thesis work presented here encompasses the processing of one of the proteins central to

Alzheimer’s disease pathogenesis. This protein, APP, has been implicated in AD due to the

amyloid hypothesis which posits that AD is caused by the aberrant aggregation of Aβ, which is a

direct cleavage product of APP. The study of AD pathogenesis has been around since 1906,

when Alois Alzheimer first described this disease and since then the field has evolved greatly.

However, there are still many unanswered questions about APP processing, both physiological

and pathophysiological. This thesis work provides measures of APP amounts, processing, and

kinetics in both the human and non-human primate CNS.

In this work, I present novel sAPPα and sAPPβ diurnal rhythms that are consistent with the

hypothesis that, to some extent, there exists APP regulation of the diurnal rhythms observed in

Aβ within our study and in previous reports (Bateman et al., 2007; Kang et al., 2009; Huang et

al., 2012a; Huang et al., 2012b). Similarly to Aβ, other APP metabolites also exhibit diurnal

patterns and these patterns are diminished with age. This indicates that at least a part of the

diurnal patterns observed in Aβ is due to diurnal patterns of APP. The cause of the diurnal APP

patterns is yet to be determined. It could be due to APP transcription, translation into protein, or

trafficking of the protein in the CNS. To a lesser degree, a diurnal variation in β-secretase or γ-

secretase may also have some effect on Aβ diurnal rhythms. Owing to our observed positive

correlations between the sAPPα diurnal rhythm and diurnal rhythms of other APP metabolites, I

speculate that the contributions of potential β-secretase and γ-secretase diurnal variation to the

overall Aβ diurnal rhythm are minimal. The source of the diurnal regulation of Aβ is likely

Page 118: The Processing of Amyloid Precursor Protein in the Central

100

upstream of the β-secretase cleavage step since sAPPα, sAPPβ, and Aβ rhythms are similar to

the extent that we have shown in Chapter 2.

In support of other recent publications, we also present that APP metabolites correlate with one

another (Lewczuk et al., 2010; Gabelle et al., 2010; Alexopolous et al., 2011). Most notably,

sAPPα and sAPPβ correlate highly positively and indicate that the β- and α-pathways are non-

competitive in hour-to-hour dynamics under normal physiological control. It appears that

cellular APP protein is not a limiting factor in this setting and that both α-secretase and β-

secretase have plenty of substrate available for cleavage. Thus, when APP protein levels

fluctuate, levels of the direct cleavage products (i.e. sAPPα and sAPPβ) fluctuate in a similar

manner to one another. To test the hypothesis that Aβ diurnal rhythms are regulated by diurnal

changes in APP, one must determine cellular APP protein levels over the time-course. Although

our current study has the benefit of showing physiological APP diurnal rhythms in the human

setting, this also limits us from examining cellular holo-APP rhythms since we do not have brain

samples corresponding to the CSF samples we used in this study. It would be beneficial to back-

translate this study into mice or rats. One robust advantage of a rodent model is that many mice

or rats may be sacrificed across a spectrum of time-points within a 24 hour period so that brains

may be harvested and concentrations of holo-APP, as well as APP metabolites may be

determined by ELISA of brain homogenate samples. In order to avoid complications of inter-

subject variability, one would have to normalize each animal’s APP metabolites to a reference

point, such as to the CSF protein level for that particular animal at the experiment’s onset.

Measuring holo-APP and APP metabolites in brain tissue would also remove the caveat of APP

trafficking from brain to CSF. In our human study, the point of CSF draw is quite distal from the

Page 119: The Processing of Amyloid Precursor Protein in the Central

101

site of APP cleavage. Removing the extra dimension of transport of APP from brain to the CSF

collected at the L4/L5 lumbar space would simplify the system.

Similarly to other studies, we show that taken alone, sAPPα or sAPPβ concentrations are not

highly predictive biomarkers of AD or amyloidosis. They do not segregate the AD group from

the non-AD groups. However, the ratio of these two metabolites (sAPPβ/ sAPPα) distinguished

the group exhibiting amyloidosis from the non-amyloidosis group. The ratio was elevated

significantly in the older amyloid positive group compared to the young and older non-amyloid

groups. This significantly elevated ratio is not particularly surprising as there is an observed

trend, albeit statistically insignificant, for sAPPβ to be elevated with age, and further with

amyloidosis. Inter-subject variability in absolute sAPPβ levels within subject group may make it

difficult to discern potential differences in APP processing among groups. Normalizing a

subject’s sAPPβ concentration to their sAPPα concentration from a particular time-point will

decrease this inter-subject variability within groups. An increase in ratio would be a direct

consequence of increased β-secretase activity and/or decreased APP processing through the α-

secretase pathway. This is consistent with our hypothesis of a shift toward β-secretase

processing of APP in AD. Thus, the sAPPβ/ sAPPα ratio may serve as an additional index to the

currently available CSF biomarkers for AD.

We report the novel finding that human total APP and rhesus macaque sAPPα or sAPPβ turnover

rates are about twice as slow as the Aβ turnover rate. The methods I developed to measure

kinetics of sAPPα, sAPPβ, and Aβ in rhesus macaque CSF may be easily performed on human

CSF that has been serially sampled over 36 hours following labeling with 13C-leucine. Such 36

hour human CSF time-courses are currently stored in the Bateman lab at -80⁰C and may be used

for such a purpose. Performing such a study using CSF from healthy young humans, older

Page 120: The Processing of Amyloid Precursor Protein in the Central

102

healthy humans, and older humans with amyloidosis, would have high implication as it would

test the hypothesis that AD is associated with increased β-secretase activity as a cause of

increased Aβ production.

Our lab was the first to develop a method to measure CNS protein metabolism in humans by

SILK. Using this technique, we discovered that Aβ is rapidly produced and cleared in the human

CNS (Bateman et al., 2006). In a γ-secretase inhibitor study, we detected small changes in Aβ

production rates, indicating SILK is a sensitive measure of CNS protein production rates

(Bateman et al., 2009). Furthermore, we have recently demonstrated an overall impaired

clearance of Aβ (Mawuenyega et al., 2010) in Alzheimer’s disease. However, half of the

subjects have increased Aβ production, while others have decreased Aβ production, suggesting a

sub-population that over-produces Aβ. Taken together, this data is suggestive of an increase in

β-secretase activity in at least some AD participants, but to date has not been directly measured.

These results have led us to hypothesize that approximately half of AD patients over-produce Aβ

owing to increased β-secretase activity, while the other half have decreased Aβ clearance. In

order to address questions of physiological and pathophysiological β-secretase (and α-secretase)

activity, we could measure sAPPβ and sAPPα kinetics in the human CNS using the above

mentioned method that I developed in rhesus macaques. By directly measuring production rates

of sAPPβ and sAPPα, we could determine if, and by how much, β-secretase activity is increased

in AD. These results would elucidate human CNS APP physiology and pathophysiology in AD.

Further, outcomes of this study may prove useful for measuring pharmacodynamic effects of

candidate therapeutics, such as β-secretase inhibitors. β-secretase is currently a high priority

target for AD, and results of altered β-secretase activity in AD are critical for understanding AD

pathophysiology and the development of disease modifying therapeutics. In order to address our

Page 121: The Processing of Amyloid Precursor Protein in the Central

103

hypothesis, we would determine sAPPβ and sAPPα production and clearance rates in at least 24

AD and 24 age-matched control patients. sAPPβ production rate is a direct measure of β-

secretase activity when controlled by sAPPα production rate. We would predict that in a control

subject, sAPPβ and sAPPα production rates would be similar (Figure 6.1.A). However, in AD

changes in β-secretase and/or α secretase activity may occur and would alter sAPPβ and/or

sAPPα production rates. One unlikely prediction is that there may be no change in β- or α-

secretase activity, evidenced by no change in the production rates of sAPPβ or sAPPα (Figure

6.1.B). A more likely scenario would be that increased β-secretase activity in AD leads to

increased sAPPβ production rate with a possible decrease in the sAPPα production rate, owing to

diminishing of the total APP pool (Figure 6.1.C). A third scenario would be an increase in total

APP during disease state, which would drive the increased production of both sAPPβ and sAPPα

(Figure 6.1.D).

A B C D

Figure 6.1. Models of changes in APP processing in AD.

Page 122: The Processing of Amyloid Precursor Protein in the Central

104

Based on evidence of Aβ overproduction in some AD subjects and an average increase in sAPPβ

/sAPPα ratio (Figure 2.10.A), we hypothesize that the most likely of these scenarios would be

that at least half of the AD population will have increased β-secretase activity (Figure 6.1.C).

We expect that about half of AD subjects will have increased sAPPβ production rates and have

increased CSF sAPPβ to sAPPα ratio and Aβ production rate. This increase in sAPPβ

production rates will correlate with increased Aβ production rates in AD. We also hypothesize

that sAPPα production rates will decrease, and clearance rates will remain unchanged. These

results would be important because they could help to inform about the potential cause of AD, as

well as the physiological and pathophysiological processing of APP. We predict that a subgroup

of AD subjects may be affected by overproduction of Aβ and may respond differently to

treatments such as β-secretase inhibitors compared with AD subjects with only impaired Aβ

clearance. This differential response to treatment could decrease the power of a clinical study.

However, a study such as this may provide a method to identify the subgroup of AD patients

with overproduction of Aβ via increased β-secretase activity. Further, knowing the magnitude of

increased β-secretase activity will assist in determining the correct pharmacodynamic dose for a

particular patient. Several recent drug trials have failed due to insufficient efficacy of the drug,

targeting of the wrong population, or incorrect timing of treatment. As many β-secretase

inhibitors (BSIs) are currently under development, results of such a study wouldl be highly

informative to treatment programs targeting β-secretase and may identify participants for clinical

trials of BSIs with the highest chance of success, additionally offering a way to titrate the drug

for the desired pharmacodynamic effect.

We were initially surprised when we discovered the lack of increased α-secretase production

effects of a potent β-secretase (BACE1) inhibitor on APP processing. Our data clearly indicated

Page 123: The Processing of Amyloid Precursor Protein in the Central

105

that the BACE1 inhibitor, MBI-5, dose-dependently lowered both sAPPβ and Aβ as measured

independently by ELISA and SILK methods. This indicated the expected phenomenon: MBI-5

was hitting its target by blocking β-secretase. It was inhibiting β-secretase from cleaving its

substrate, APP, thus preventing APP from being processed through the β-pathway. The

intriguing question that we sought to answer was what effect, if any, would MBI-5 have on APP

processing through the α-pathway. Prior to the BACE1 inhibitor study, and as discussed in

Chapter 2, we had generated a simplistic model of interrelated APP processing pathways due to a

common APP pool from which both the α-secretase and β-secretase gained their substrate, which

they went on to cleave through the α- or β-pathway, respectively. That model was made after the

observations from human APP processing studies we had done previously and which showed a

clear positive correlation between the two direct products of the first step of APP processing by

α-secretase (sAPPα) of β-secretase (sAPPβ). Owing to these prior observations and the

subsequent hypothetical model that we had generated, we hypothesized that if MBI-5 were a

potent β-secretase inhibitor, then the common APP pool would have surplus APP which would,

in turn, be shunted down the α-pathway. Such a shunting would result in an increase in the

direct product of APP processing by α-secretase (sAPPα), which would be detectable by the

methods we were employing. We were surprised to observe that although MBI-5 did decrease

APP processing through the β-pathway, there was absolutely no corresponding change in APP

processing through the α-pathway as measured by SILK. SILK measurements of sAPPα showed

four labeling kinetic curves (for vehicle and the three doses) that were superimposable. By

ELISA, sAPPα measurements showed increases in all four study groups, including the vehicle

group. By ELISA, the sAPPα response to MBI-5 was not dose-dependent, although the highest

dose did show a 35% increase. However, with the substantial error in these measurements, and

Page 124: The Processing of Amyloid Precursor Protein in the Central

106

the observation that there was an apparent, albeit modest, increase in sAPPα in the vehicle group,

we were skeptical whether these results were representative of a truly physiological event or

caused by the ELISA assay not being fully specific for sAPPα. These observations, taken

together with our results that found no change detected in sAPPα by the sensitive SILK method,

which has been able to detect even minor changes in APP metabolites resulting from γ-secretase

inhibition (Cook et al., 2010), sporadic AD (Mawuenyega et al., 2010), and autosomal dominant

AD (Potter et al., 2013), led us to believe that APP was not being shunted to any great degree

through the classic α-secretase pathway during BACE1 inhibition, and thus our model of APP

processing may be overly simplistic. Therefore, here we propose a more sophisticated model

which alludes to both a spatial and a temporal separation of APP processing by α-secretase and

β-secretase. This model better explains the superficially disjoint phenomena observed in the

various studies of this thesis.

First, to preface the model, I will refer to background information (for review, see Chapter 1)

that indicates APP is a transmembrane protein that is abundant throughout subcellular

compartments such as the ER, Golgi apparatus, endosomes, etc. It is also located directly in the

cellular membrane. The family of ADAM proteins which are α-secretases are also ubiquitous

transmembrane proteins located throughout the cell and α-secretase cleavage of APP is evident at

the cell surface (Sisodia 1992). Thus cleavage of APP through the α-pathway may occur in

many areas within the cell but predominantly at the cell surface, and the directly resulting sAPPα

is released into the ECF. β-secretase is a transmembrane aspartyl protease whose activity is

primarily in neural tissues (neurons) (Vassar et al., 1999), although it is found to some extent

throughout most cells and tissues (Haass et al., 1992). At the cellular level, β-secretase

predominantly localizes to endosomes and the late Golgi/TGN. It requires an acidic environment

Page 125: The Processing of Amyloid Precursor Protein in the Central

107

(pH 4.5) for optimal activity. Thus, its function is highest in the acidic subcellular compartments

of the endocytic pathway, namely the early and late endosomes, as well as in the Golgi apparatus

(Koo and Squazzo, 1994; Haass et al., 1995).

In neurons, APP that has been newly synthesized is transported by the Golgi membrane

trafficking down the axon and to secretory vesicles. The secretory vesicles move APP to the cell

surface during the secretory pathway. In the secretory vesicles and at the cell membrane, APP is

predominantly cleaved by α-secretase (Sisodia et al., 1990; Hung et al., 1993; Golde et al.,

1992), releasing primarily sAPPα into the extracellular fluid (ECF). A fraction of the APP that

has not been cleaved at the membrane can be re-internalized into endosomes. At the endosome,

largely β-secretase, and subsequently γ-secretase, may cleave APP to generate sAPPβ and Aβ,

respectively. Endosomes with these β-secretase pathway APP metabolites in the lumen may fuse

with the cell membrane and release these metabolites into the ECF. Endosomes not immediately

recycled to the membrane will be trafficked down the endocytic pathway to late endosomes.

These endosomes, in turn, may fuse with the cell membrane to release any newly generated

sAPPβ and Aβ into the ECF. Conversely, the late endosome may fuse with a lysosome and any

holo-APP or APP metabolites remaining within the endosome’s lumen will subsequently be

degraded.

Based on this complex trafficking, we have generated a schematic (Figure 6.2.) of APP

processing that would reconcile the discrepancy between the positive correlation of sAPPα and

sAPPβ in humans and the phenomenon of APP not being shunted down the α-secretase pathway

in the presence of β-secretase inhibition. There is a common starting pool of translated APP,

which is consistent with sAPPα and sAPPβ being positively correlated when measured in human

CSF. Nevertheless, α- and β-secretase mostly do not have access to concomitantly cleave this

Page 126: The Processing of Amyloid Precursor Protein in the Central

108

APP at the same location within the cell. Initially APP is trafficked through the secretory

pathway to the cell surface, where it becomes available for cleavage by α-secretase.

Subsequently, APP uncleaved by α-secretase may be reinternalized into the endocytic pathway in

which β-secretase processing occurs. Endocytosis is irreversible, thus α-secretase cleavage may

not be in equilibrium with the β-secretase cleavage. When a BACE1 inhibitor is present, levels

of sAPPβ and Aβ are markedly decreased as we observe with the rhesus macaques that had MBI-

5 administered. The APP that remains uncleaved isn’t readily available for α-secretase cleavage

anymore because of the irreversibility of endocytosis, and, therefore, is mostly degraded in the

lysosome. This is consistent with what we observe in the BACE1 inhibitor treated rhesus

macaques: no change in sAPPα.

Page 127: The Processing of Amyloid Precursor Protein in the Central

109

Figure 6.2. Amyloid Precursor Protein processing & trafficking schematic. APP is translated across the membrane of the endoplasmic reticulum and trafficked into the Golgi apparatus. In the secretory pathway, APP is transported to the cell membrane. Once at the membrane, cleaved sAPPα metabolites are released into the extracellular fluid (ECF) from which they may travel to the CSF and be detected by our methods. Uncleaved APP may be re-internalized into the cell in endosomes and go through the endocytic pathway. In the endosome, APP is predominantly cleaved by β-secretase, as endosomes have an acidic environment, which is conducive to β-secretase activity. Endosomes may be recycled to the cell membrane, releasing any cleaved APP products, such as sAPPβ and Aβ, into the ECF, or they may undergo lysosomal degradation. In the latter case, any cleaved APP metabolites, as well as uncleaved APP, is degraded.

Page 128: The Processing of Amyloid Precursor Protein in the Central

110

To determine if this model is accurate it would be necessary to conduct additional experiments of

subcellular compartmentalization and cellular APP and sAPPα which were not feasible in our

present experimental set-up. Measuring cellular holo-APP (in addition to measurements of

individual APP metabolites) would help indicate whether total APP concentrations are increasing

due to β-secretase inhibition but are not being shunted down the α-secretase pathway. Total

cellular APP measurements are only feasible if one uses brain tissue. As our measurements of

APP metabolites are done using CSF and brain tissue was not available, we could not address

this. A future study should incorporate a rodent model of 13C-leucine labeling during BACE1

inhibition. Cerebrospinal fluid or interstitial fluid (ISF) could be collected prior to harvesting the

brain at key time-points over 72 hours. Time-points would be chosen based on a test CSF

labeling curve of APP metabolites done before study onset as turnover of label will likely differ

in a rodent model from our current non-human primate model. Brain tissue homogenates and

CSF could be analyzed by ELISA and SILK for all APP metabolites and the brain homogenates

could also be analyzed for holo-APP by both methods.

Additionally, we could use slices from these brains to examine the extent of subcellular

localization of APP during BACE1 inhibition. We could probe for holo-APP, sAPPβ, Aβ,

sAPPα, and BACE1 using immunofluorescence and a FRET (fluorescence resonance energy

transfer) approach to determine co-localization of these proteins during the course of BACE1

inhibition, as well as after administration of placebo (control).

One other avenue to pursue is to determine whether a potential build-up of APP substrate may be

degraded through a mechanism other than traditional α-secretase cleavage activity. A recent

study indicated that alternative α- and β- secretase processing may occur in vivo in the human

CNS, i.e. sAPPαQ686 (sAPPα’), sAPPαK687, sAPPβM671, and sAPPβY681 (sAPPβ’) from the APP770

Page 129: The Processing of Amyloid Precursor Protein in the Central

111

splice variant (Brinkmalm et al., 2013). I would propose assessing our currently banked rhesus

macaque CSF from this study to precisely determining the contributions of individual sAPP

subspecies (sAPPα, sAPPα’, sAPPβ, and sAPPβ’) to our current “sAPPα” and “sAPPβ”

measurements. Although it is not feasible to determine the amount of these subspecies with

current immunoassay systems, it has been performed with LC-MS/MS in human CSF. We could

use a protocol similar to that outlined in Brinkmalm et al. (2013) and immunoprecipitate rhesus

macaque CSF with P2-1 antibody and digest it with trypsin or Asp-N. Peptides for each sAPP

subspecies would be separated by LC-MS/MS to determine whether, sAPPα’ is increased during

BACE1 inhibition. If that is the case, this would indicate that APP does indeed get shunted

toward a type of α-secretase processing if β-secretase processing is blocked, but our prior study

was unable to determine that because we were measuring only sAPPα (by SILK).

Overall, the studies in this thesis have added to the basic science understanding of the complex

processing of APP in vivo. We have been able to provide evidence for α- and β-secretase

processing of APP from a complex single pool model, which is consistent with positive

correlation of APP metabolites from either of these two processing pathways. However, we

present BACE1 inhibition data that would suggest β-secretase inhibition is downstream from the

majority of α-secretase processing, thus indicating that APP is not shunted down the α-secretase

pathway, but it likely degraded in lysosomes. Future experiments to verify these findings are

discussed above. Further, one could imagine setting up a similar experiment to the BACE1

inhibitor study, but instead of using a BACE1 inhibitor, administering the PKC activator phorbol

12-myristate 13-acetate (PMA) which has been shown to increase secretion of sAPPα (Caporaso

et al., 1992; Buxbaum et al., 1993). If our model is correct, such an experiment would

demonstrate not only an increase in sAPPα, but also decreases in sAPPβ and Aβ if less APP is

Page 130: The Processing of Amyloid Precursor Protein in the Central

112

being trafficked through the endocytic pathway due to overwhelming α-secretase cleavage.

Tumor necrosis factor-α-converting enzyme (TACE) is the ADAM family member attributed to

the observed PMA-stimulated APP cleavage (Buxbaum et al., 1998). Therefore, another

potential experiment could be the administration of a TACE inhibitor, i.e., tumor necrosis factor-

α protease inhibitor (TAPI) or IC-3, which would block the PMA-stimulated secretion of sAPPα.

We could measure the β-secretase pathway APP metabolites in this study and would expect to

see them increase if our model is correct.

The studies in this thesis have also provided important clinical implications for AD patients.

Results discussed herein support the hypothesis that Aβ diurnal rhythms are at least in part due to

APP diurnal regulation. This finding is important for future clinicians measuring APP

metabolites for diagnostic purposes. In order to maintain consistent and accurate measurements,

CSF samples should always be collected at a set time of day because hourly variations of Aβ and

sAPPβ are significant. In this thesis, I also present some evidence for a potential benefit to

adding the sAPPβ/sAPPα ratio to measures for determining AD. Lastly, the BACE1 inhibitor

study in this thesis has potentially profound implications in the clinic. MBI-5 was successful at

lowering sAPPβ and Aβ in non-human primates and could be a promising drug candidate for a

human clinical trial.

Page 131: The Processing of Amyloid Precursor Protein in the Central

cxiii

Bibliography

Alexopoulos P, Tsolakidou A, Roselli F, Arnold A, Grimmer T, Westerteicher C, Leante MR, Förstl H, Livrea P, Kurz A, Perneczky R (2012) Clinical and neurobiological correlates of soluble amyloid precursor proteins in the cerebrospinal fluid. Alzheimers Dement 8: 304-311.

Alzheimer A (1907) Über eine eigenartige Erkrankung der Hirnrinde. Allg Zeitschr Psychiatr 64146-64148.

Alzheimer Disease and Frontotemporal Dementia Mutation Database. Available at http://www.molgen.ua.ac.be/ADMutations/

Alzheimer’s Association (2013) 2013 Alzheimer’s Disease Facts and Figures, Alzheimer’s & Dementia, Volume 9, Issue 2.

Anderson JJ, Holtz G, Baskin PP, Wang R, Mazzarelli L, Wagner SL, Menzaghi F (1999) Reduced cerebrospinal fluid levels of α-secretase-cleaved amyloid precursor protein in aged rats: correlation with spatial memory deficits. Neuroscience 93: 1409-1420.

Andreason N, Hesse C, Davidsson P, Minthon L, Wallin A, Winblad B, Vanderstichele H,

Vanmechelen E, Blennow K (1999) Cerebrospinal fluid β-amyloid(1-42) in Alzheimer disease:

differences between early- and late-onset Alzheimer disease and stability during the course of disease. Arch Neurol 56: 673-680.

Bateman RJ et al. (2012) Clinical and biomarker changes in dominantly inherited Alzheimer's disease. N Engl J Med 367: 795-804.

Bateman RJ, Aisen PS, De Strooper B, Fox NC, Lemere CA, Ringman JM, Salloway S, Sperling RA, Windisch M, Xiong C (2011) Autosomal-dominant Alzheimer's disease: a review and proposal for the prevention of Alzheimer's disease. Alzheimers Res Ther 3: 1.

Bateman RJ, Siemers ER, Mawuenyega KG, Wen G, Browning KR, Sigurdson WC, Yarasheski KE, Friedrich SW, Demattos RB, May PC, Paul SM, Holtzman DM (2009) A γ-secretase inhibitor decreases amyloid-β production in the central nervous system. Ann Neurol 66: 48-54.

Bateman RJ, Wildsmith KR, Dobrowolska J, Munsell L, Mawuenyega KG (2009) Methods for Simultaneously Measuring the in vivo Metabolism of Two or More Isoforms of a Biomolecule. U.S. Patent Application 2011-0294138 A1, filed November 12, 2009, published December 1, 2011. Patent Pending Pub. No.: WO/2010/056815

Bateman RJ, Munsell LY, Chen X, Holtzman DM, Yarasheski KE (2007a) Stable isotope labeling tandem mass spectrometry (SILT) to quantify protein production and clearance rates. J Am Soc Mass Spectrom 18: 997-1006.

Page 132: The Processing of Amyloid Precursor Protein in the Central

cxiv

Bateman RJ, Wen G, Morris JC, Holtzman DM (2007b) Fluctuations of CSF amyloid-β levels: implications for a diagnostic and therapeutic biomarker. Neurology 68: 666-669.

Bateman RJ, Munsell LY, Morris JC, Swarmn R, Yarasheski KE, Holtzman DM (2006) Human amyloid-β synthesis and clearance rates as measured in cerebrospinal fluid in vivo. Nat Med 12: 856-861.

Brinkmalm G, Brinkmalm A, Bourgeois P, Persson R, Hansson O, Portelius E, Mercken M, Andreasson U, Parent S, Lipari F, Ohrfelt A, Bjerke M, Minthon L, Zetterberg H, Blennow K, Nutu M (2013) Soluble amyloid precursor protein α and β in CSF in Alzheimer's disease. Brain Res 1513: 117 - 126. doi: 10.1016/j.brainres.2013.03.019. Epub 2013 Mar 27.

Busciglio J, Gabuzda DH, Matsudaira P, Yankner BA (1993) Generation of β-amyloid in the secretory pathway in neuronal and nonneuronal cells. Proc Natl Acad Sci USA 90: 2092-2096.

Buxbaum JD, Koo EH, and Greengard P (1993) Protein phosphorylation inhibits production of Alzheimer amyloid beta/A4 peptide. PNAS 90: 9195 – 9198.

Buxbaum JD, Liu K-N, Luo Y, Slack JL, Stocking KL, Peschon JJ, Johnson RS, Castner BJ, Cerretti DP, Black RA (1998) Evidence That Tumor Necrosis Factor α Converting Enzyme Is Involved in Regulated α-Secretase Cleavage of the Alzheimer Amyloid Protein Precursor. JBC 273: 27765 - 27767.

Caporaso GL, Gandy SE, Buxbaum JD, Ramabhadran TV, Greengard P (1992) Protein phosphorylation regulates secretion of Alzheimer beta/A4 amyloid precursor protein. PNAS 89: 3055 – 3059.

Cole SL and Vassar R (2007) The Alzheimer's disease β-secretase enzyme, BACE1. Mol Neurodegener 2:22.

Cook JJ, Wildsmith KR, Gilberto DB, Holahan MA, Kinney GG, Mathers PD, Michener MS, Price EA, Shearman MS, Simon AJ, Wang JX, Wu G, Yarasheski KE, Bateman RJ (2010) Acute γ-secretase inhibition of nonhuman primate CNS shifts Amyloid Precursor Protein (APP) metabolism from amyloid-β production to alternative APP fragments without Amyloid-β rebound. J Neurosci 30: 6743-6750.

Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 261: 921-923.

Cumming JN et al. (2012) Structure based design of iminohydantoin BACE1 inhibitors: identification of an orally available, centrally active BACE1 inhibitor. Bioorg Med Chem Lett 22: 2444-2449.

Page 133: The Processing of Amyloid Precursor Protein in the Central

cxv

Cupers P et al. (2001) The discrepancy between presenilin subcellular localization and γ-secretase processing of amyloid precursor protein. J Cell Biol 154: 731-740.

De Strooper B (2007) Loss-of-function presenilin mutations in Alzheimer disease. Talking point on the role of presenilin mutations in Alzheimer disease. EMBO Rep 8: 141-146.

Dobrowolska J, Mawuenyega KG, Bateman RJ (2008) The metabolism of amyloid precursor protein as measured in human cerebrospinal fluid. Program No. 138.2. Neuroscience Meeting Planner. Washington D.C.: Society for Neuroscience, Online.

Egan MF, Forman MS, Palcza J, Tseng J, Leempoels J, Ramael S, Han D, Jhee S, Ereshefsky L, Tanen M, Laterza O, Dockendorf M, Krishna G, Ma L, Wagner JA, Troyer MD (2012) OC4 The Novel BACE Inhibitor MK-8931 Dramatically Lowers CSF Aβ Peptides in Healthy Subjects Following Single and Multiple Dose Administration. 5th conference clinical trials on Alzheimer's disease: October 29-31, 2012, Grimaldi Forum, Convention Center, Monte Carlo. J Nutr Health Aging 16: 797. Fagan AM, Mintun MA, Mach RH, Lee SY, Dence CS, Shah AR, LaRossa GN, Spinner ML, Klunk WE, Mathis CA, DeKosky ST, Morris JC, Holtzman DM (2006) Inverse relation between

in vivo amyloid imaging load and cerebrospinal fluid Aβ42 in humans. Ann Neurol 59: 512-519.

Fagan AM, Roe CM, Xiong C, Mintun MA, Morris JC, Holtzman DM. (2007) Cerebrospinal

Fluid tau/β-Amyloid42 Ratio as a Prediction of Cognitive Decline in Nondemented Older Adults.

Arch Neurol 64: 343-349.

Fagan AM, Head D, Shah AR, Marcus D, Mintun M, Morris JC, Holtzman DM (2009)

Decreased cerebrospinal fluid Aβ42 correlates with brain atrophy in cognitively normal elderly.

Ann Neurol 65: 176–183.

Fellgiebel A, Kojro E, Müller MJ, Scheurich A, Schmidt LG, Fahrenholz F (2009) CSF APPsα and phosphorylated tau protein levels in mild cognitive impairment and dementia of Alzheimer's type. J Geriatr Psychiatry Neurol 22: 3-9.

Ferri CP, Prince M, Brayne C, Brodaty H, Fratiglioni L, Ganguli M, Hall K, Hasegawa K, Hendrie H, Huang Y, Jorm A, Mathers C, Menezes Pr, Rimmer E, Scazufca M, Alzheimer’s Disease International (2005) Global Prevalence of dementia: a Delphi consensus study. Lancet 366: 2112-2117.

Forman MS et al. (2013) The Novel BACE Inhibitor MK-8931 Dramatically Lowers CSF Aβ

Peptide in Patients with Mild to Moderate Alzheimer's Disease. AD/PDTM 2013, the 11th

International Conference on Alzheimer’s and Parkinson’s Diseases, Florence, March 2013.

Fukumoto H, Cheung BS, Hyman BT, Irizarry MC (2002) β-secretase protein and activity are increased in the neocortex in Alzheimer disease. Arch Neurol 59: 1381-1389.

Page 134: The Processing of Amyloid Precursor Protein in the Central

cxvi

Furukawa K, Sopher BL, Rydel RE, Begley JG, Pham DG, Martin GM, Fox M, Mattson MP (1996) Increased activity-regulating and neuroprotective efficacy of α-secretase-derived secreted amyloid precursor protein conferred by a C-terminal heparin-binding domain. J Neurochem 67: 1882-1896.

Gabelle A, Roche S, Gény C, Bennys K, Labauge P, Tholance Y, Quadrio I, Tiers L, Gor B, Chaulet C, Vighetto A, Croisile B, Krolak-Salmon P, Touchon J, Perret-Liaudet A, Lehmann S (2010) Correlations between soluble α/β forms of amyloid precursor protein and Aβ38, 40, and 42 in human cerebrospinal fluid. Brain Res 1357: 175-183.

Gagnon JF, Petit D, Latreille V, Montplaisir J (2008) Neurobiology of sleep disturbances in neurodegenerative disorders. Curr Pharm Des 14: 3430-3445.

Games D et al. (1995) Alzheimer-type neuropathology in transgenic mice overexpressing V717F β-amyloid precursor protein. Nature 373: 523-527.

Gilberto DB, Zeoli AH, Szczerba PJ, Gehret JR, Holahan MA, Sitko GR, Johnson CA, Cook JJ, Motzel SL (2003) An alternative method of chronic cerebrospinal fluid collection via the cisterna magna in conscious rhesus monkeys. Contemp Top Lab Anim Sci 42: 53-59.

Goate A et al. (1991) Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 349: 704-706.

Golde TE, Estus S, Younkin LH, Selkoe DJ, Younkin SG (1992) Processing of the amyloid protein precursor to potentially amyloidogenic derivatives. Science 255: 728-730.

Goodman Y and Mattson MP (1994) Secreted forms of β-amyloid precursor protein protect hippocampal neurons against amyloid β-peptide-induced oxidative injury. Exp Neurol 128: 1-12.

Grimmer T, Alexopoulos P, Tsolakidou A, Guo L-H, Henriksen G, Yousefi BH, Förstl H, Sorg C, Kurz A, Drzezga A, Perneczky R (2012) Cerebrospinal Fluid BACE1 Activity and Brain Amyloid Load in Alzheimer's Disease. ScientificWorldJournal 2012: 712048. doi: 10.1100/2012/712048. Epub 2012 Jan 3.

Guo Q, Wang Z, Li H, Wiese M, Zheng H (2012) APP physiological and pathophysiological functions: insights from animal models. Cell Res 22: 78-89.

Gustafson DR, Skoog I, Rosengren L, Zetterberg, H, and Blennow K (2007) Cerebrospinal fluid β-amyloid 1–42 concentration may predict cognitive decline in older women. J Neurol Neurosurg Psychiatry 78: 461-464.

Haass C, Lemere CA, Capell A, Citron M, Seubert P, Schenk D, Lannfelt L, Selkoe DJ (1995) The Swedish mutation causes early-onset Alzheimer's disease by β-secretase cleavage within the secretory pathway. Nat Med 1: 1291-1296.

Page 135: The Processing of Amyloid Precursor Protein in the Central

cxvii

Hardy J and Selkoe DJ (2002) The Amyloid Hypothesis of Alzheimer’s Disease: Progress and Problems on the Road to Therapeutics. Science 297: 353-356.

Hassold T, Hunt PA, Sherman S (1993) Trisomy in humans: incidence, origin and etiology. Curr Opin Genet Dev 3: 398-403.

Holsinger RM, McLean CA, Beyreuther K, Masters CL, Evin G (2002) Increased expression of the amyloid precursor β-secretase in Alzheimer's disease. Ann Neurol 51: 783-786.

Holsinger RMD, McLean CA, Collins SJ, Masters CL, Evin, G (2004) Increased β-secretase activity in cerebrospinal fluid of Alzheimer’s disease subjects. Ann Neurol 55: 898–899.

Holsinger RM, Lee JS, Boyd A, Masters CL, Collins SJ (2006) CSF BACE1 activity is increased in CJD and Alzheimer disease versus other dementias. Neurology 67: 710-712.

Holtzman DM, Morris JC, Goate AM (2011) Alzheimer’s Disease: the challenge of the second century. Sci Transl Med 3: 77sr1.

Huang Y, Potter R, Sigurdson W, Kasten T, Connors R, Morris JC, Benzinger T, Mintun M, Ashwood T, Ferm M, Budd SL, Bateman RJ (2012b) β-amyloid dynamics in human plasma. Arch Neurol 69: 1591-1597. doi:10.1001/archneurol.2012.1810. Epub 2012 Sep 10.

Huang Y, Potter R, Sigurdson W, Santacruz A, Shih S, Ju Y-E, Kasten T, Morris JC, Mintun M, Duntley S, Bateman RJ (2012a) Effects of age and amyloid deposition on Aβ dynamics in the human central nervous system. Arch Neurol 69: 51-58. doi: 10.1001/archneurol.2011.235. Epub 2011 Sep 12.

Hung AY, Haass C, Nitsch RM, Qiu WQ, Citron M, Wurtman RJ, Growdon JH, Selkoe DJ (1993) Activation of protein kinase C inhibits cellular production of the amyloid beta-protein. J Biol Chem 268: 22959-22962.

Hung AY and Selkoe DJ (1994) Selective ectodomain phosphorylation and regulated cleavage of β-amyloid precursor protein. EMBO J 13: 534-542.

Irizarry MC, Locascio JJ, Hyman BT (2001) β-site APP cleaving enzyme mRNA expression in APP transgenic mice: anatomical overlap with transgene expression and static levels with aging. Am J Pathol 158: 173-177.

Johnston JA, Liu WW, Todd SA, Coulson DT, Murphy S, Irvine GB, Passmore AP (2005) Expression and activity of β-site cleaving enzyme in Alzheimer’s disease. Biochem Soc Trans 33: 1096-1100.

Jonsson T et al. (2012) A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature 488: 96-99.

Page 136: The Processing of Amyloid Precursor Protein in the Central

cxviii

Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, Fujiki N, Nishino S, Holtzman DM (2009) Amyloid-β dynamics are regulated by orexin and the sleep-wake cycle. Science 326: 1005-1007.

Kim KS, Miller DL, Sapienza VJ, Chen C-MJ, Bai C, Gundke-Iqbal I, Currie JR, Wisniewski HM (1988) Production and characterization of monoclonal antibodies reactive to synthetic cerebrovascular amyloid peptide. Neurosci Res Commun 2: 121–130.

Kim M, Suh J, Romano D, Truong MH, Mullin K, Hooli B, Norton D, Tesco G, Elliott K, Wagner SL, Moir RD, Becker KD, Tanzi RE (2009) Potential late-onset Alzheimer's disease-associated mutations in the ADAM10 gene attenuate α-secretase activity. Hum Mol Genet 18: 3987-3996.

Klunk et al. (2004) Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol 55: 306-319.

Koo EH and Squazzo SL (1994) Evidence that production and release of amyloid β-protein involves the endocytic pathway. J Biol Chem 269: 17386-17389.

Koo EH, Squazzo SL, Selkoe DJ, Koo CH (1996) Trafficking of cell-surface amyloid β-protein precursor. I. Secretion, endocytosis and recycling as detected by labeled monoclonal antibody. J Cell Sci 109: 991-998.

Lannfelt L, Basun H, Wahlund LO, Rowe BA, Wagner SL (1995) Decreased α-secretase-cleaved amyloid precursor protein as a diagnostic marker for Alzheimer's disease. Nat Med 1: 829-832.

Lemere CA, Blusztajn JK, Yamaguchi H, Wisniewski T, Saido TC, Selkoe DJ (1996) Sequence of deposition of heterogeneous amyloid β-peptides and APO E in Down syndrome: implications for initial events in amyloid plaque formation. Neurobiol Dis 3: 16-32.

Levy-Lahad E et al. (1995) Candidate gene for the chromosome 1 familial Alzheimer's disease locus. Science 269: 973-977.

Lewczuk P et al. (2010) Soluble amyloid precursor proteins in the cerebrospinal fluid as novel potential biomarkers of Alzheimer’s disease: a multicenter study. Mol Psychiatry 15: 138-145.

Li R, Lindholm K, Yang LB, Yue X, Citron M, Yan R, Beach T, Sue L, Sabbagh M, Cai H, Wong P, Price D, Shen Y (2004) Amyloid β peptide load is correlated with increased β-secretase activity in sporadic Alzheimer’s disease patients. Proc Natl Acad Sci USA 101: 3632-3637.

Malamas MS, Robichaud A, Edei J, Quagliato D, Solvibile W, Zhou P, Morris K, Turner J, Wagner E, Fan K, Olland A, Jacobsen S, Reinhart P, Riddell D, Pangalos M (2010) Design and synthesis of aminohydantoins as potent and selective human β-secretase (BACE1) inhibitors with enhanced brain permeability. Bioorg Med Chem Lett 20: 6597-6605.

Page 137: The Processing of Amyloid Precursor Protein in the Central

cxix

Malamud N (1972) Neuropathology of organic brain syndromes associated with aging. In Gaitz CM (ed): Aging and the Brain. New York: Plenum Press, pp 63-87.

Mandal M et al. (2012) Design and validation of bicyclic iminopyrimidinones as β amyloid cleaving enzyme-1 (BACE1) inhibitors: conformational constraint to favor a bioactive conformation. J Med Chem 55: 9331-9345.

Mattsson N, Rajendran L, Zetterberg H, Gustavsson M, Andreasson U, Olsson M, Brinkmalm G, Lundkvist J, Jacobson LH, Perrot L, Neumann U, Borghys H, Mercken M, Dhuyvetter D, Jeppsson F, Blennow K, Portelius E (2012) BACE1 Inhibition Induces a Specific Cerebrospinal Fluid β-Amyloid Pattern That Identifies Drug Effects in the Central Nervous System. PLoS ONE 7(2): e31084.

Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, Yarasheski KE, Bateman RJ (2010) Decreased clearance of CNS β-amyloid in Alzheimer's disease. Science 330: 1774.

May PC et al. (2011) Robust central reduction of Amyloid-β in humans with an orally available, non-peptidic β-secretase inhibitor. J Neurosci 31: 16507-16516.

Mintun MA, Larossa GN, Sheline YI, Dence CS, Lee SY, Mach RH, Klunk WE, Mathis CA,

DeKosky ST, Morris JC (2006) [11C]PIB in a nondemented population: potential antecedent

marker of Alzheimer disease. Neurology 67: 446-452.

Morris JC, Roe CM, Xiong C, Fagan AM, Goate AM, Holtzman DM, Mintun MA (2010) APOE predicts amyloid-β but not tau Alzheimer pathology in cognitively normal aging. Ann Neurol 67: 122-131.

Morris JC, Roe CM, Grant EA, Head D, Storandt M, Goate AM, Fagan AM, Holtzman DM, Mintun MA (2009) Pittsburgh compound B imaging and prediction of progression from cognitive normality to symptomatic Alzheimer disease. Arch Neurol 66: 1469–1475.

Müller U, Winter P, Graeber MB (2013) A presenilin 1 mutation in the first case of Alzheimer's disease. Lancet Neurol 12: 129-130. doi: 10.1016/S1474-4422(12)70307-1. Epub 2012 Dec 14.

Nishitomi K, Sakaguchi G, Horikoshi Y, Gray AJ, Maeda M, Hirata-Fukae C, Becker AG, Hosono M, Sakaguchi I, Minami SS, Nakajima Y, Li HF, Takeyama C, Kihara T, Ota A, Wong PC, Aisen PS, Kato A, Kinoshita N, Matsuoka Y (2006) BACE1 inhibition reduces endogenous Aβ and alters APP processing in wild-type mice. J Neurochem 99: 1555-1563.

Olsson A, Höglund K, Sjögren M, Andreasen N, Minthon L, Lannfelt L, Buerger K, Möller HJ, Hampel H, Davidsson P, Blennow K (2003) Measurement of α- and β-secretase cleaved amyloid precursor protein in cerebrospinal fluid from Alzheimer patients. Exp Neurol 183: 74-80.

Page 138: The Processing of Amyloid Precursor Protein in the Central

cxx

Oltersdorf T, Ward PJ, Henriksson T, Beattie EC, Neve R, Lieberburg I, Fritz LC (1990) The Alzheimer amyloid precursor protein. Identification of a stable intermediate in the biosynthetic /degradative pathway. J Biol Chem 265: 4492-4497.

Ortega F, Stott J, Visser SAG, Bendtsen C (2013) Interplay between α-, β-, and γ-secretases determines biphasic amyloid-β protein level in the presence of a γ-secretase inhibitor. J Biol Chem 288: 785-792.

Parvathy S, Hussain I, Karran EH, Turner AJ, Hooper NM (1999) Cleavage of Alzheimer’s amyloid precursor protein by α-secretase occurs at the surface of neuronal cells. Biochemistry 38: 9728–9734.

Pasternak SH et al. (2003) Presenilin-1, nicastrin, amyloid precursor protein, and γ-secretase activity are co-localized in the lysosomal membrane. J Biol Chem 278: 26687–26694.

Pierrot N and Octave J-N (2008) Processing of amyloid precursor protein and amyloid peptide neurotoxicity. Curr Alzheimer Res 5: 92-99.

Portelius E, Price E, Brinkmalm G, Stiteler M, Olsson M, Persson R, Westman-Brinkmalm A, Zetterberg H, Simon AJ, Blennow K (2011) A novel pathway for amyloid precursor protein processing. Neurobiol Aging 32: 1090-1098.

Postina R, Schroeder A, Dewachter I, Bohl J, Schmitt U, Kojro E, Prinzen C, Endres K, Hiemke C, Blessing M, Flamez P, Dequenne A, Godaux E, van Leuven F, Fahrenholz F (2004) A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest 113: 1456-1464.

Potter R, Patterson BW, Elbert DL, Ovod V, Kasten T, Sigurdson W, Mawuenyega K, Blazey T, Goate A, Chott R, Yarasheski KE, Holtzman DM, Morris JC, Benzinger TL, Bateman RJ (2013) Increased in vivo amyloid-β42 production, exchange, and loss in presenilin mutation carriers. Sci Transl Med 5:189ra77. doi: 10.1126/scitranslmed.3005615.

Querfurth HW, Wijsman EM, St George-Hyslop PH, Selkoe DJ (1995) βAPP mRNA transcription is increased in cultured fibroblasts from the familial Alzheimer's disease-1 family. Mol Brain Res 28: 319-337.

Ray WJ et al. (1999) Cell surface presenilin-1 participates in the γ-secretase-like proteolysis of Notch. J Biol Chem 274: 36801-36807.

Redline S, Kirchner HL, Quan SF, Gottlieb DJ, Kapur V, Newman A (2004) The effects of age, sex, ethnicity, and sleep-disordered breathing on sleep architecture. Arch Intern Med 164: 406-418.

Page 139: The Processing of Amyloid Precursor Protein in the Central

cxxi

Roh JH, Huang Y, Bero AW, Kasten T, Stewart FR, Bateman RJ, Holtzman DM (2012) Disruption of the sleep-wake cycle and diurnal fluctuation of β-amyloid in mice with Alzheimer’s disease pathology. Sci Transl Med 4: 150ra122.

Sakurai T (2007) The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci. 8: 171-181. Epub 2007 Feb 14.

Sankaranarayanan S et al. (2009) First demonstration of cerebrospinal fluid and plasma Aβ lowering with oral administration of a β-site amyloid precursor protein-cleaving enzyme 1 inhibitor in nonhuman primates. J Pharmacol Exp Ther 328: 131-140.

Sankaranarayanan S, Price EA, Wu G, Crouthamel MC, Shi XP, Tugusheva K, Tyler KX, Kahana J, Ellis J, Jin L, Steele T, Stachel S, Coburn C, Simon AJ (2008) In vivo β-secretase 1 inhibition leads to brain Aβ lowering and increased α-secretase processing of amyloid precursor protein without effect on neuregulin-1. J Pharmacol Exp Ther 324: 957-969.

Sennvik K, Fastbom J, Blomberg M, Wahlund LO, Winblad B, Benedikz E (2000) Levels of α- and β-secretase cleaved amyloid precursor protein in the cerebrospinal fluid of Alzheimer’s disease patients. Neurosci Lett 278: 169-172.

Sherrington R et al. (1995) Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature 375: 754-760.

Sisodia SS, Koo EH, Beyreuther K, Unterbeck A, Price DL (1990) Evidence that beta-amyloid protein in Alzheimer’s disease is not derived by normal processing. Science 248: 492-495.

Sisodia SS (1992) β-amyloid precursor protein cleavage by a membrane-bound protease. Proc Natl Acad Sci 89: 6075 - 6079.

Skoog I, Davidsson P, Aevarsson O, Vanderstichele H, Vanmechelen E, Blennow K (2003) Cerebrospinal fluid beta-amyloid 42 is reduced before the onset of sporadic dementia: a population-based study in 85-year-olds. Dement Geriatr Cogn Disord 15: 169-176.

Skovronsky DM, Moore DB, Milla ME, Doms RW, Lee VM (2000) Protein Kinase C-dependent α-secretase competes with β-secretase for cleavage of amyloid-β precursor protein in the Trans-Golgi Network. J Biol Chem 275: 2568-2575.

Smith-Swintosky VL, Pettigrew LC, Craddock SD, Culwell AR, Rydel RE, Mattson MP (1994) Secreted forms of β-amyloid precursor protein protect against ischemic brain injury. J Neurochem 63: 781-784.

Stamford AW et al. (2012) Discovery of an orally available, brain penetrant BACE1 inhibitor that affords robust CNS Aβ reduction. ACS Med Chem Lett 3: 897-902.

Page 140: The Processing of Amyloid Precursor Protein in the Central

cxxii

Stomrud E, Hansson O, Blennow K, Minthon L, and Londos E (2007) Cerebrospinal fluid biomarkers predict decline in subjective cognitive function over 3 years in healthy elderly. Dement Geriatr Cogn Disord 24: 118–124.

Takahashi H, Fukumoto H, Maeda R, Terauchi J, Kato K, Miyamoto M (2010) Ameliorative effects of a non-competitive BACE1 inhibitor TAK-070 on Aβ peptide levels and impaired learning behavior in aged rats. Brain Res 1361: 146-156.

Tranah GJ, Blackwell T, Stone KL, Ancoli-Israel S, Paudel ML, Ensrud KE, Cauley JA, Redline S, Hillier TA, Cummings SR, Yaffe K, SOF Research Group (2011) Circadian activity rhythyms and risk of incident dementia and mild cognitive impairment in older women. Ann Neurol 70: 722-732.

Truong et al. (2010) Design of an orally efficacious hydroxyethylamine (HEA) BACE-1 inhibitor in a preclinical animal model. Bioorg Med Chem Lett 20: 6231-6236.

Verheijen JH, Huisman LG, van Lent N, Neumann U, Paganetti P, Hack CE, Bouwman F, Lindeman J, Bollen EL, Hanemaaijer R (2006) Detection of a soluble form of BACE-1 in human cerebrospinal fluid by a sensitive activity assay. Clin Chem 52: 1168–1174.

Vuletic S, Li G, Peskind ER, Kennedy H, Marcovina SM, Leverenz JB, Petrie EC, Lee VM, Galasko D, Schellenberg GD, Albers JJ (2008) Apolipoprotein E highly correlates with AβPP- and tau-related markers in human cerebrospinal fluid. J Alzheimers Dis 15: 409-417.

Walter J, Capell A, Hung AY, Langen H, Schnölzer M, Thinakaran G, Sisodia SS, Selkoe DJ, Haass C (1997) Ectodomain phosphorylation of β-amyloid precursor protein at two distinct cellular locations. J Biol Chem 272: 1896-1903.

Weidemann A, König G, Bunke D, Fischer P, Salbaum JM, Masters CL, Beyreuther K (1989) Identification, biogenesis, and localization of precursors of Alzheimer's disease A4 amyloid protein. Cell 57: 115-126.

Wisniewski KE, Wisniewski HM, Wen GY (1985) Occurrence of neuropathological changes and dementia of Alzheimer's disease in Down's syndrome. Ann Neurol 17: 278-282.

Wolfe RR, Chinkes DL, Wolfe RR (2005) Isotope tracers in metabolic research: principles and practice of kinetic analysis, Ed 2. Hoboken, NJ: Wiley.

Wu G, Sankaranarayanan S, Hsieh SH, Simon AJ, Savage MJ (2011) Decrease in Brain Soluble Amyloid Precursor Protein β (sAPPβ) in Alzheimer’s Disease Cortex. J Neurosci Res 89: 822-832.

Wu G, Sankaranarayanan S, Wong J, Tugusheva K, Michener MS, Shi X, Cook JJ, Simon AJ, Savage MJ (2012) Characterization of plasma β-secretase (BACE1) activity and soluble amyloid

Page 141: The Processing of Amyloid Precursor Protein in the Central

cxxiii

precursor proteins as potential biomarkers for Alzheimer's disease. J Neurosci Res 90: 2247-2258.

Yaffe K, Laffan AM, Harrison SL, Redline S, Spira AP, Ensrud KE, Ancoli-Israel S, Stone KL (2011) Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in olrder women. JAMA 306: 613-619.

Yamazaki T, Koo EH, Selkoe DJ (1996) Trafficking of cell-surface amyloid beta-protein precursor. II. Endocytosis, recycling and lysosomal targeting by immunolocalization. J Cell Sci 109: 999-1008.

Yang LB, Lindholm K, Yan R, Citron M, Xia W, Yang XL, Beach T, Sue L, Wong P, Price D, Li R, Shen Y (2003) Elevated β-secretase expression and enzymatic activity detected in sporadic Alzheimer disease. Nat Med 9: 3-4.

Yarasheski KE, Smith K, Rennie MJ, Bier DM (1992) Measurement of muscle protein fractional synthetic rate by capillary gas chromatography/combustion isotope ratio mass spectrometry. Biol Mass Spectrom 21: 486-490.

Yasuda M, Maeda K, Ikejiri Y, Kawamata T, Kuroda S, Tanaka C (1997) A novel missense mutation in the presenilin-1 gene in a familial Alzheimer's disease pedigree with abundant amyloid angiopathy. Neurosci Lett 232: 29-32.

Yu G et al. (2000) Nicastrin modulates presenilin-mediated notch/glp-1 signal transduction and βAPP processing. Nature 407: 48-54.

Zetterberg H, Andreasson U, Hansson O, Wu G, Sankaranarayanan S, Andersson ME, Buchhave P, Londos E, Umek RM, Minthon L, Simon AJ, Blennow K (2008) Elevated cerebrospinal fluid BACE1 activity in incipient Alzheimer disease. Arch Neurol 65: 1102–1107.

Zhang YW, Thompson R, Zhang H, Xu H (2011) APP processing in Alzheimer’s disease. Mol Brain 4:3.

Zheng H and Koo EH (2011) Biology and pathophysiology of the amyloid precursor protein. Mol Neurodegener 6:27.

Zhong Z, Ewers M, Teipel S, Bürger K, Wallin A, Blennow K, He P, McAllister C, Hampel H, Shen Y (2007) Levels of β-secretase (BACE1) in cerebrospinal fluid as a predictor of risk in mild cognitive impairment. Arch Gen Psychiatry 64: 718-726.

Zigman W, Silverman W, Wisniewski HM (1996) Aging and Alzheimer’s disease in Down syndrome: Clinical and pathological changes. Ment Retard Dev Disabil Res Rev 2: 73–79.