@2020 Yang Yang ALL RIGHTS RESERVED

158
@2020 Yang Yang ALL RIGHTS RESERVED

Transcript of @2020 Yang Yang ALL RIGHTS RESERVED

Page 1: @2020 Yang Yang ALL RIGHTS RESERVED

@2020

Yang Yang

ALL RIGHTS RESERVED

Page 2: @2020 Yang Yang ALL RIGHTS RESERVED

AUTOPHAGY SUPPRESSES OXIDATIVE STRESS AND

P53 FOR ADULT MICE SURVIVAL AND IS ESSENTIAL FOR

TELOCYTES SURVIVAL AND INTESTINAL HOMEOSTASIS

by

YANG YANG

A dissertation submitted to the

School of Graduate Studies

Rutgers, The State University of New Jersey

In partial fulfillment of the requirements

For the degree of

Doctor of Philosophy

Graduate Program in Biochemistry

Written under the direction of

Eileen White

And approved by

New Brunswick, New Jersey

JANUARY, 2020

Page 3: @2020 Yang Yang ALL RIGHTS RESERVED

ii

ABSTRACT OF THE DISSERTATION

Autophagy Suppresses Oxidative Stress and p53

for Adult Mice Survival and Is Essential for

Telocytes Survival and Intestinal Homeostasis

By YANG YANG

Dissertation Director:

Eileen White

Autophagy captures intracellular components and delivers them to lysosomes

for degradation and recycling. Conditional autophagy deficiency in adult mice

causes liver damage, and shortens lifespan to three months due to

neurodegeneration. As autophagy deficiency causes p53 induction and cell

death in neurons, we sought to test if p53 mediates the toxic effects of

autophagy deficiency. Here we conditionally deleted Trp53 (p53 hereafter)

and/or the essential autophagy gene Atg7 throughout adult mice. Compared to

Atg7Δ/Δ mice, life span of Atg7Δ/Δp53Δ/Δ mice was extended due to delayed

neurodegeneration and resistance to death upon fasting. Atg7 also limited

apoptosis induced by the p53 activator Nutlin-3, suggesting that autophagy

inhibited p53 activation. To test if increased oxidative stress in Atg7Δ/Δ mice was

Page 4: @2020 Yang Yang ALL RIGHTS RESERVED

iii

responsible for p53 activation, Atg7 was deleted in the presence or absence of

the master regulator of antioxidant defense Nuclear factor erythroid 2-related

factor 2 (Nrf2). Nrf2-/-Atg7Δ/Δ mice died rapidly due to small intestine damage,

which was not rescued by co-deletion of p53. Thus, autophagy limits p53

activation and p53-mediated neurodegeneration. In turn, NRF2 mitigates lethal

intestine degeneration upon autophagy loss. These findings illustrate the

tissue-specific roles for autophagy and functional dependencies on the p53 and

NRF2 stress response mechanisms.

Atg7 and Atg5 are both essential autophagy genes (ATG) involved in

autophagosome formation and deleting these genes leads to autophagy

deficiency. As essential autophagy genes can function differently, we then

sought to test whether whole-body conditional Atg5 deletion in adult mice

(Atg5Δ/Δ mice) would have similar phenotype to that of Atg7 deletion. In contrast

to Atg7Δ/Δ mice, Atg5Δ/Δ mice surprisingly lived for less than five days. Atg5Δ/Δ

mice showed selective damage in the ileum part of intestine, with marked

epithelial damage and loss of barrier function. In comparison to Atg7Δ/Δ mice,

the ileum of Atg5Δ/Δ showed evidence of more rapid loss of autophagy, and loss

of stem cells and malfunction of Paneth cells. Furthermore, Atg5Δ/Δ mice had

decreased active β-catenin in the ileum, the key transcription factor for Wnt

signaling that is essential for intestinal stem cell renewal. Atg5Δ/Δ mice lost

PDGFRα+ mesenchymal cells (telocytes) in the ileum, which are required to

Page 5: @2020 Yang Yang ALL RIGHTS RESERVED

iv

provide Wnt signals to stem cells. Deletion of Atg5 more gradually overcame

the loss of ileum telocytes and stem cells and resulted in death much later from

neurodegeneration similar to deletion of Atg7 or Atg12. Atg5Δ/Δ telocytes

displayed significantly decreased aspartate and nucleotides, which caused

their loss. These findings reveal a novel function of autophagy in maintenance

of telocytes, Wnt signaling, and thereby stem cells essential for intestinal

homeostasis and the survival of adult mice. As impaired autophagy is

associated with Inflammatory Bowel Diseases, this suggests that failure to

maintain telocyte function is involved in development of this disease.

Page 6: @2020 Yang Yang ALL RIGHTS RESERVED

v

PREFACE

Part of this research was originally submitted to Genes&Development and is

being reviewed.

GENESDEV/2019/335570

Yang Yang, Gizem Karsli Uzunbas, Laura Poillet-Perez, Akshada Sawant,

Zhixian Sherrie Hu, Yuhan Zhao, Wenwei Hu, and Eileen White. Autophagy

Promotes Mammalian Survival by Suppressing Oxidative Stress and p53.

Page 7: @2020 Yang Yang ALL RIGHTS RESERVED

vi

ACKNOWLEDGEMENTS

This dissertation would never exist without the support from lots of people: my

mentors, my colleagues, my friends and my family members. I would like to

thank all these people for their contributions to my projects and the start of my

academic roads.

First of all, I would like to specially thank my thesis advisor, Eileen White, for

her kind guidance on my project. Her advice was really valuable to me and

helped me a lot during my experiments. It is challenging to work in the lab

because of her high requirements and expectations, but I really learnt a lot

during my stay in the lab. It is really fortunate for me to have a chance to join

the lab and worked on these two great projects, and her enthusiasm on

science really motivated me during my graduate study. Thank you for all the

knowledge and research skills you taught me and it is of great help for my

future academic career.

I would like to also thank my committee members: Michael Verzi, for all the

guidance for experiments on identifying the intestinal phenotype of the Atg5Δ/Δ

mice, especially for the suggestion on the telocytes. Without your knowledge

on gastrointestinal area we wound not be able to identify such an interesting

phenotype. I would like to acknowledge Dr. Wenwei Hu for her help on

identifying the Atg7Δ/Δp53Δ/Δ phenotype. I would like to acknowledge my other

Page 8: @2020 Yang Yang ALL RIGHTS RESERVED

vii

committee members: Bing Xia and Darren Carpizo for their time, valuable

feedbacks and suggestions on both of my projects, your constructive ideas

were really helpful to me.

I feel so lucky to have a chance to work with my great lab members and

colleagues. First, I want to mention Laura and Akshada. After they joined the

lab, they gave me a lot of valuable suggestions on lab skills, including

techniques, writing skills and also lots of valuable ideas on interpreting the

results. I would also like to thank Sherrie, who is helping me so much on

mouse organization and mouse experiments with her knowledge and

experience. My thanks also go to Eduardo, for his help on the techniques and

the valuable suggestions on how to plan for a future academic career. I also

want to thank Jessie, who taught me when I joined the lab as a rotation student

on lots of basic skills, and her lab member Vrushank and Khoosheh for the

technical support. Without their help, this thesis cannot be completed.

I also need to acknowledge all the CINJ staff; in particular, Wen Xie for

guidance on confocal microscope, Shafiq from the histology lab for preparation

of the slides from paraffin blocks. Yuhan previously in CINJ for real time PCR. I

would also thank for all the outside members who helped with these projects;

specially Jayanta Debnath and his lab member Tim on providing tissues

materials to help us identify the gut phenotype of Atg5Δ/Δ mice, Shalev Itzkovitz

Page 9: @2020 Yang Yang ALL RIGHTS RESERVED

viii

for the single cell RNA sequencing data, and Shawn Davidson and his lab

member RaElle and Noel for MALDI imaging mass spec.

I owe many thanks to my family members too. My precious cousin and best

bro, Bokan Bao; my supportive elder cousin, Xiaomin Yang and my two cute

nieces Yi and Yue; my younger cousins, Jiayin and Junyi. My special thanks

go to my beloved parents, Huanchun Yang and Xiaoqiu Shi, for always being

with me for all these years. I thank them for their unconditional support and

constant listening to all my happiness and sorrow.

Page 10: @2020 Yang Yang ALL RIGHTS RESERVED

ix

TABLE OF CONTENTS

ABSTRACT OF THE DISSERTATION ........................................................... ii

PREFACE ....................................................................................................... v

ACKNOWLEDGEMENTS .............................................................................. vi

TABLE OF CONTENTS ................................................................................. ix

LIST OF FIGURES ....................................................................................... xiii

CHAPTER 1

INTRODUCTION ............................................................................................. 1

Autophagy Signaling Network .................................................................. 3

Regulation of Autophagy Machinery ........................................................ 9

Transcriptional Regulation ........................................................................ 9

Post-translational Regulation .................................................................. 11

Epigenetic and microRNAs Regulation ................................................... 13

Physiological Role of Autophagy ........................................................... 14

p53 and Autophagy .................................................................................. 21

CHAPTER 2

MATERIALS AND METHODS ...................................................................... 23

Mouse models .......................................................................................... 24

TAM administration .................................................................................. 25

Page 11: @2020 Yang Yang ALL RIGHTS RESERVED

x

Survival ..................................................................................................... 25

Fasting ...................................................................................................... 26

Histology ................................................................................................... 26

Nutlin-3a administration .......................................................................... 27

Real-time PCR .......................................................................................... 27

Western blot ............................................................................................. 29

Organoid culture ...................................................................................... 29

In vivo Wnt ligand supplementation ....................................................... 30

Glucose supplementation ....................................................................... 30

FITC dextran supplementation ................................................................ 31

Single cell RNA sequencing for PDGFRα+ mesenchymal cells ........... 31

Matrix-assisted laser desorption ionization coupled to imaging mass

spectrometry (MALDI-IMS) ...................................................................... 31

CHAPTER 3

AUTOPHAGY PROMOTES MAMMALIAN SURVIVAL BY SUPPRESSING

OXIDATIVE STRESS AND P53 .................................................................... 34

Introduction .............................................................................................. 35

Results ...................................................................................................... 37

Loss of p53 delays neurodegeneration and prolongs survival of

Atg7-deficient mice. ................................................................................ 37

p53 deficiency reduces tissue damage in Atg7Δ/Δ mice ........................... 38

ATG7 is required for tumorigenesis driven by p53 deletion .................... 39

Page 12: @2020 Yang Yang ALL RIGHTS RESERVED

xi

p53 deficiency prevents lethality in Atg7Δ/Δ mice during fasting .............. 40

ATG7 is required to protect liver and brain from p53-mediated damage . 43

ATG7 limits p53 activation by Nutlin-3a .................................................. 47

Atg7 deficiency is synthetically lethal in the absence of Nrf2 .................. 52

Discussion ................................................................................................ 58

Supplementary Figures ........................................................................... 61

CHAPTER 4

AUTOPHAGY IS ESSENTIAL FOR TELOCYTE SURVIVAL, WNT

SIGNALING AND INTESTINAL STEM CELL HOMEOSTASIS ................... 70

Introduction .............................................................................................. 71

Results ...................................................................................................... 73

Conditional, whole-body Atg5 deletion in adult mice leads to ileum damage

and death ................................................................................................ 73

Acute whole body Atg5 deletion decreases blood glucose and causes loss

of intestinal barrier function ..................................................................... 77

Deletion of Atg5 causes more rapid loss of autophagy in the Ileum than

deletion of Atg7 ....................................................................................... 78

Ileum damage in Atg5Δ/Δ mice is caused by loss of stem cells ................ 79

In vitro deletion of Atg5 lead to growth inhibition and can be rescued by

supplementation of Wnt ligands .............................................................. 83

Atg5 is required to maintain PDGFRα+ mesenchymal cells. ................... 87

Telocyte specific deletion of Atg5 causes similar ileum damage phenotype.

Page 13: @2020 Yang Yang ALL RIGHTS RESERVED

xii

................................................................................................................ 91

Deleting Atg5 more gradually rescues intestinal ileum function. ............. 95

Autophagy is required to maintain mitochondria function, TCA cycle and

nucleotide metabolism in PDGFRα+ mesenchymal cells ...................... 100

Discussion .............................................................................................. 106

Supplemental Figures ............................................................................ 111

CHAPTER 5

REFERENCES ............................................................................................ 131

Page 14: @2020 Yang Yang ALL RIGHTS RESERVED

xiii

LIST OF FIGURES

Fig. 3.1 Atg7Δ/Δ, p53Δ/Δ mice have extended life span, delayed tissue damage

and neurodegeneration compared to Atg7Δ/Δ mice……………….....................41

Fig. 3.2. Autophagy is required to protect liver and brain from p53

accumulation, DNA damage response activation and apoptosis………..........45

Fig. 3.3 Activation of p53 by MDM2 antagonist Nutlin-3a in Atg7Δ/Δ mice leads to

further increased DNA damage response and apoptosis in liver and

brain…………………………………………………………………………...........50

Fig. 3.4 Atg7 deficiency is synthetically lethal in the absence of Nrf2….........55

Figure 3.5. Mechanism by which autophagy interacts with the p53 and NRF2

stress response mechanisms to protect tissues in a tissue specific

manner…………………………………………………………………..................57

Figure S3.1. Related to Figure 3.1………………………………………….........61

Figure S3.2. Related to Figure 3.2……………………………………………......63

Figure S3.3. Related to Figure 3.3……………………………………………......66

Figure S3.4. Related to Figure 3.3……………………………………………......67

Figure S3.5. Related to Figure 3.4.…………………………………………….....68

Figure. 4.1. Conditional, whole-body Atg5 deletion in adult mice leads to ileum

damage and death………….......………………………………………...............75

Figure 4.2. Ileum damage in Atg5Δ/Δ mice is caused by loss of stem cells.

……………………………………………………………………………………….81

Figure 4.3. In vitro deletion of Atg5 lead to growth inhibition and can be

Page 15: @2020 Yang Yang ALL RIGHTS RESERVED

xiv

rescued by supplementation of Wnt ligands....................................................85

Figure 4.4. Atg5 is required to maintain PDGFRα+ mesenchymal cells...........89

Figure 4.5. Telocyte specific deletion of Atg5 causes similar ileum damage

phenotype.......................................................................................................93

Figure 4.6. Deleting Atg5 more gradually rescues intestinal ileum

function...........................................................................................................98

Fig.4.7. Autophagy is required to maintain mitochondria function, TCA cycle

and nucleotide metabolism in PDGFRα+ mesenchymal cells…....................103

Fig.4.8. Mechanism by which autophagy is required for telocytes survival to

maintain intestinal homeostasis....................................................................105

Figure S4.1. Related to Figure 4.1.…………………………………………......111

Figure S4.2. Related to Figure 4.1.…………………………………………......113

Figure S4.3. Related to Figure 4.2.…………………………………………......115

Figure S4.4. Related to Figure 4.2.…………………………………………......117

Figure.S4.5. Related to Figure 4.3.…………………………………………......119

Figure S4.6. Related to Figure 4.4.…………………………………………......122

Figure S4.7. Related to Figure 4.4.…………………………………………......124

Figure S4.8. Related to Figure 4.5.…………………………………………......126

Figure S4.9. Related to Figure 4.6.…………………………………………......128

Figure S4.10. Related to Figure 4.7.…………………………………………....130

Page 16: @2020 Yang Yang ALL RIGHTS RESERVED

1

CHAPTER 1

INTRODUCTION

Page 17: @2020 Yang Yang ALL RIGHTS RESERVED

2

Autophagy is a dynamic catabolic process by which cells direct their own

intracellular proteins, lipids, organelles and pathogens to the lysosomal

compartment for degradation (Mizushima, 2010). Autophagy and the

ubiquitin-proteosome system (UPS) are the two major cellular quality control

systems which is responsible for clearance of proteins and organelles. These

two systems formed an interactive network, where decision of pathway is self

organized and controlled by different biophysical markers and

compartmentalization. Usually, the UPS is in charge of the misfolded soluble

proteins and short lived proteins, sending them to the proteosome for

degradation; while autophagy deals with larger cytosolic structure and

organelles such as damaged mitochondria and ER, long lived protein, protein

aggregates and bacteria invading into the cell (Pohl and Dikic, 2019).

There are three types of autophagy: macroautophagy, microautophagy and

chaperone-mediated autophagy. Macroautophagy (hereafter autophagy) is the

major type and is responsible for most of the protein aggregates and

organelles degradation. Generally, the autophagy pathway involves the

formation of double membrane-bound vesicles called autophagosomes that

capture cargos including cytoplasmic proteins, organelles and bacteria.

Autophagosomes with cargo then fuse with lysosomes to form autolysosomes

where the cargo is degraded (Kaur and Debnath, 2015). The breakdown

products are then released into the cytoplasm where they are recycled, and

Page 18: @2020 Yang Yang ALL RIGHTS RESERVED

3

specifically used as substrates for central carbon metabolism to sustain

survival (Guo et al., 2016; Rabinowitz and White, 2010). By doing this,

autophagy also serves as the quality control system by eliminating protein

aggregates and damaged organelles(Mizushima and Komatsu, 2011).

Autophagy Signaling Network

Autophagy is controlled by the autophagy-related proteins (ATG) and other

proteins that enable the formation of autophagosomes and recognition and

capture of cargos (Mizushima and Komatsu, 2011). Autophagy is usually

upregulated by different kinds of stress such as oxidative stress, hypoxia,

nutrient deprivation and infection. During nutrient deprivation when cells are

low in energy supply, autophagy is activated to recycle substrates to replenish

the cellular ATP levels. Low ATP: AMP levels triggers the 5’ AMP activated

protein kinase (AMPK), which is the main energy sensor and regulator of

cellular metabolism for energy homeostasis, induced autophagy(Kim et al.,

2011). In contrast to AMPK, another anabolic metabolic stress sensor,

mammalian target of rapamycin multi protein complex 1(mTORC1), which can

be activated by mitogen-activated protein kinase (MAPK) and

Phosphoinositide 3-kinase (PI3K)/ Protein Kinase B (AKT) pathway inhibits

autophagy. Under nutrient deprivation, AMPK induction by metabolic stress

either inhibits or bypasses mTORC1 to initiate autophagy(Jung et al., 2010).

Page 19: @2020 Yang Yang ALL RIGHTS RESERVED

4

Autophagy machinery is progressed in several steps: autophagosome

formation (including initiation, nucleation and elongation), fusion with lysosome

and degradation/cargo recycling. These steps are controlled by ATGs and

other proteins. The core protein complex for initiation step is the

uncoordinated-51-like kinase (ULK) complex, which is composed of ULK1,

ATG13, ATG101 and focal adhesion kinase family interacting protein of

200-kDa (FIP200). During sufficient nutrient condition, mTORC1 binds to and

phosphorylate the ULK1 at Serine (Ser) 757 and ATG13, prevents their

activation and interaction to AMPK (Hosokawa et al., 2009; Kim et al., 2011).

Upon starvation, following inhibition of mTORC1, ULK1 is dephosphorylated

on the mTORC1 binding site, dissociated from mTORC1, and then undergoes

autophosphorylation. The autophosphorylation activates ULK1 followed by

phosphorylation of ATG13 and FIP200, thus the full protein complex is

activated (Hosokawa et al., 2009; Jung et al., 2010).

Following ULK complex activation, it triggers nucleation of the phagophore by

phosphorylating components of the nucleation complex class III PI3K (PI3KC3)

complex, which is composed of PI3KC3, vacuolar protein sorting 34 (VPS34),

BECN1 (Beclin-1, BCL-2 interacting protein) and general vesicular transport

factor (p115), ATG14 for PI3KC3 complex I or UV radiation resistance

associated gene protein (UVRAG) in PI3KC3 complex II. The main function for

the nucleation complex is to generate local phosphatidylinositol-3-phosphate

Page 20: @2020 Yang Yang ALL RIGHTS RESERVED

5

(PI3P) at the phagophore assembly site (PAS), which is usually on the

endoplasmic reticulum(ER) emanating membrane domains called the

omegasome. However, the ER-mitochondria and ER- plasma membrane

contact sites, the Golgi complex, plasma membrane and recycling endosomes

are also proposed as PAS (Abada and Elazar, 2014) .PI3P then recruits the

PI3P effector proteins WD repeat domain phosphoinositide interacting proteins

2(WIPI2) and zinc-finger FYVE domain containing protein 1 (DFCP1) to the

omegasome via interaction with their PI3P binding domains (Dikic and Elazar,

2018). Afterwards, ATG9 containing vesicles generated by the cellular

secretory pathway were recruited to the PAS, which delivers additional lipids

and proteins for the upcoming membrane elongation (Karanasios et al., 2013;

Nishimura et al., 2017) .

Autophagosome elongation requires two ubiquitin-like systems. The first

system is ATG8 family proteins, in mammals including Microtubule-associated

proteins 1A/1B light chain 3B (LC3) and γ- aminobutyric acid receptor-

associated proteins (GABARAPs). Nascent pro-ATG8 is processed by the

cysteine protease ATG4 at its C-terminal to expose a glycine residue essential

for the binding of phosphatidylethanolamine (PE). Processed ATG8s are

activated by the E1-like ubiquitin activating enzyme ATG7 (for LC3 it is

referred to as LC3-I), then conjugated to the PE with the help of ATG3 (for LC3

referred to as LC3-II) (Hamasaki et al., 2013). The other conjugation system is

Page 21: @2020 Yang Yang ALL RIGHTS RESERVED

6

ATG5-ATG12-ATG16L1 protein complex. ATG12 is activated by ATG7 and

then binds to the ATG5 by E2 like ubiquitin conjugating enzyme ATG10 to form

ATG12-ATG5 conjugate, which is working as an E3 like ubiquitin ligating

enzyme essential for the efficient conjugating of PE to ATG8s (Hamasaki et al.,

2013). ATG16L1 directly crosslinks with ATG12-ATG5 conjugate, or forms

homooligomers by its coiled-coil domain, then crosslink several ATG12-ATG5

conjugates into a large protein complex, then the ATG16L1-ATG12-ATG5

complex is localized to the PAS by interaction of ATG16L1 with the WIPI2

(Dooley et al., 2014; Kaufmann et al., 2014). Interestingly, besides the

processing of C terminal of ATG8 proteins, ATG4 is also capable of

deconjugating ATG8s from PE to release them from the ER membrane and

prevent phagophore formation (Satoo et al., 2009). Therefore, PE-conjugated

ATG8s are protected from ATG4, possibly through inhibition of ATG4 by

phosphorylation of ULK1 or mitochondria generated reactive oxygen species

(ROS) (Pengo et al., 2017; Scherz-Shouval et al., 2007).

Before sealing the phagophore, one important step is to identify and capture

the cargo in the autophagosome. Selective autophagy requires the labeling of

cargo with capturing signals (prominently ubiquitin chains), which is

recognized by autophagy cargo receptors that link cargo to the ATG8 family

proteins autophagosome membrane (Dikic and Elazar, 2018). There are

several autophagy cargo receptors identified, including SQSTM1

Page 22: @2020 Yang Yang ALL RIGHTS RESERVED

7

(Sequestosome 1, or p62), NIP-like protein X (NIX), FuN14 domain- containing

protein 1 (FUNDC1), BCl2/adenovirus e1B 19 kDa protein- interacting protein

3 (BNIP3), calcium- binding and coiled- coil domain-containing protein 2

(NDP52), Optineurin (OPTN), and neighbor of BRCA1 gene 1(NBR1) (Dikic

and Elazar, 2018; Johansen and Lamark, 2011; Wild et al., 2011). All these

cargo receptors have a conserved LC3-interacting region (LIR) essential for

cargo interaction with the autophagosome membrane. Besides, p62 and NBR1

contains a ubiquitin associated (UBA) domain, OPTN has a ubiquitin binding in

ABIN and NEMO(UBAN) domain and a zinc finger domain, and NDP52 has a

zinc finger domain; all the domains are involved in recognition and uptake of

these ubiquitinated cargos(Johansen and Lamark, 2011). To degrade protein

aggregates, for example, p62 interacts with protein labeled with ubiquitin

chains and recognize the LC3 through LIR to enable selective autophagic

degradation and is degraded in the lysosome along with cargo (Johansen and

Lamark, 2011). Damaged mitochondrion is another important type of cargo.

Ubiquitin dependent degradation of mitochondria requires the participation of

PTEN-induced putative kinase 1 (PINK1), a mitochondrial kinase; and Parkin,

an E3 ubiquitin ligase. When mitochondria are damaged and then depolarized,

PINK1 is stabilized and accumulates on the outer membrane of the

mitochondria and followed by recruitment of Parkin (Narendra et al., 2010).

Parkin ubiquitinates mitochondrial outer membrane proteins, then the

damaged mitochondria are recognized by p62, OPTN or NDP52 and brought

Page 23: @2020 Yang Yang ALL RIGHTS RESERVED

8

to the autophagosome. Mitophagy can also occur in a Ubiquitin independent

manner, which utilizes the NIX, FUNC1 or BNIP3 as cargo receptors. These

receptors are mitochondria proteins which contain a LIR domain without UBA

domain, therefore can directly function as a cargo receptor for mitophagy

(Dikic and Elazar, 2018). Some lipids and phospholipids such as cardiolipin

and ceramide also serve as cargo receptors for mitophagy. Cardiolipin is

located in the inner membrane of mitochondria. However, it is externalized on

the mitochondria surface upon mitochondrial damage, where it can be

recognized by LC3 and recruited to autophagosome (Chu et al., 2013; Sentelle

et al., 2012). Thus, autophagy receptors can recognize damaged organelles

and bring them to autophagosome for degradation (Okamoto, 2014). Other

autophagy receptors such as OPTN and NDP52 can similarly mediate the

degradation of invading bacteria by ubiquitination (Wild et al., 2011).

The next step following elongation, cargo recruitment and sealing of

phagophore by LC3-II is the maturation of autophagosome and fusion with

lysosome. These steps require the clearance of ATGs on the outer membrane

of autophagosome, recruitment of the lysosomal delivery machinery, and the

machinery for fusion with the lysosome, which are working in a coordinated

manner but the detailed mechanism are poorly characterized (Stolz et al.,

2014). ATG8 proteins link autophagosome to kinesins through FYVE and

coiled- coil domain containing protein 1(FYCO1) to drive its maturation (Olsvik

Page 24: @2020 Yang Yang ALL RIGHTS RESERVED

9

et al., 2015). The fusion machinery that is studied is the SNAREs, which are

protein that mediate the fusion of vesicles with target membrane. There are

SNARE protein on the vesicle (v-SNARE) and the target membrane

(t-SNARE), which combine together to form a trans-SNARE which provides

the force for membrane fusion. For autophagosome fusion with the lysosome,

Syntaxin 17 (STX17) and synaptosomal- associated protein 29 (SNAP29) are

localized on the autophagosome as v-SNARE, while vesicle-associated

membrane protein 8 (VAMP8) is on the lysosome as t-SNARE. With the help

of homotypic fusion and protein sorting (HOPS) complex, autophagosome and

lysosome membrane are tethered together for SNARE- mediated fusion (Diao

et al., 2015; Itakura et al., 2012).

Regulation of Autophagy Machinery

Transcriptional Regulation

Autophagy genes are first regulated at the transcriptional level. One

evolutionary conserved protein that regulates autophagy is the forkhead box O

(FOXO) transcriptional factor. FOXO proteins are activated in response to

cellular stress, and regulate genes involved in energy production, oxidative

stress, cell viability and proliferation (Dikic and Elazar, 2018). Upon cellular

stress, FOXO proteins are phosphorylated by AKT and translocated to the

nucleus to upregulate the Atg genes transcriptionally in hepatocytes,

Page 25: @2020 Yang Yang ALL RIGHTS RESERVED

10

cardiomyocytes, neurons and primary renal proximal tubular cells(Webb and

Brunet, 2014; Zhao et al., 2007).

Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is

another transcription factor that upregulates autophagy, which induces BECN1,

BCL2 and p62 expression by binding to their antioxidant response elements

(AREs) in the promoter region (Copetti et al., 2009; Jain et al., 2010; Niture

and Jaiswal, 2012). Another key regulator for autophagy genes is the

transcription factor EB (TFEB). TFEB control the mTORC1 lysosome

recruitment for degradation or activity by regulating expression of the mTOR

activating Rag GTPase complex component Ras-related GTP binding protein

D (RagD)(Di Malta et al., 2017). Thus, TFEB is inhibited by mTORC1 under

nutrient sufficient condition, and released upon starvation to trigger the

expression level of autophagy related proteins including WIPI, p62 and

UVRAG (Settembre et al., 2011).

Recently, the epigenetic reader bromodomain- containing protein 4 (BRD4)

were reported as repressors of autophagic genes needed for autophagosome

biogenesis at transcriptional level. BRD4 is a member of the bromodomain and

extraterminal (BET) family proteins characterized by two N-terminal

bromodomains and an extraterminal (ET) domain, which binds to acetylated

histones and transcription factors through bromodomains and recruits

Page 26: @2020 Yang Yang ALL RIGHTS RESERVED

11

transcriptional regulators involved in the activation of genes for cell growth and

cell cycle progression. BRD4 suppresses the expression of a series of

autophagy genes by binding to the promoter regions under normal growth

conditions, and the repression is alleviated when autophagy is upregulated

(Sakamaki et al., 2017).

Post-translational Regulation

Autophagy can be triggered by nutrient starvation through inhibition of

mTORC1 and activation of AMPK. Low amino acids upregulate autophagy

through inhibition of mTORC1 but not mTORC2 (Condon and Sabatini, 2019;

Saxton and Sabatini, 2017). In glucose starvation, low cellular energy level and

decreased ATP: AMP ratio is sensed by AMPK and serine/threonine- protein

kinase STK11 (LKB1). LKB1 activates autophagy through AMPK, which

inhibits mTORC1 by activation of mTORC1inhibitor Tuberous Sclerosis

Complex 2 (TSC2) and inhibition of Raptor by phosphorylation, an essential

component of the mTORC1 (Tripathi et al., 2013). When LKB1 is deficient,

autophagy is activated to maintain metabolic flexibility by modulating lipid

metabolism in lung tumors (Bhatt et al., 2019).

ATG proteins are mainly regulated by post-translational modification including

phosphorylation, lipidation (i.e. LC3 lipidation of PE, discussed in the previous

Page 27: @2020 Yang Yang ALL RIGHTS RESERVED

12

section), oxidation and acetylation. During initiation stage, ULK1, ATG13 and

FIP200 are inhibited or activated by phosphorylation at different regions as

discussed in the previous section. At phagophore nucleation step, BECN1 can

be phosphorylated by ULK1 or AMPK. And it can promote autophagy by

interacting with the activating molecule in Beclin 1-regulated autophagy protein

1 (AMBRA1). Phosphorylated AMBRA1 by ULK1 is released from

microtubules to allow binding of BECN1 and consequent activation of the full

nucleation complex PI3KC3-C1 (Di Bartolomeo et al., 2010). At

autophagosome elongation, maturation and fusion steps, phosphorylation of

LC3 by protein kinase A (PKA) inhibits its activity to negatively regulate

autophagy (Cherra et al., 2010). It was recently discovered that

phosphorylation of LC3 at Thr50 by the Ste20 Hippo kinase orthologues

serine/threonine-protein kinase 3 (STK3) and STK4 is required for fusion of

autophagosome with lysosome (Wilkinson et al., 2015). Oxidation also

modulates the function of ATGs. During nutrient starvation, increased ROS

specifically generates hydrogen peroxide, which activates ATG4 for

processing of ATG8 proteins through direct oxidation (Scherz-Shouval et al.,

2007). Moreover, ATGs are regulated by acetylation and deacetylation

process. ATG5, ATG7 and ATG8s are acetylated in autophagy inactive

conditions. During starvation, silent mating type information regulation 2

homolog 1(SIRT1), a deacetylase, directly interacts with these ATGs and

deacetylates them to induce autophagosome formation (Lee et al., 2008).

Page 28: @2020 Yang Yang ALL RIGHTS RESERVED

13

Besides, VPS34 is acetylated by histone acetyltransferase p300, which either

diminishes the VPS34 affinity to the PI3KC3-C1 or prevents formation of the

nucleation complex. During fasting, p300 inactivates and causes VPS34

deacetylation, coordinates with the activation of autophagy in mouse liver (Su

et al., 2017).

Epigenetic and microRNAs Regulation

Histone modification of Atg genes by histone acteyltransferases (HATs) and

histone deacetylases (HDACs) haven’t been experimentally shown yet.

However, these histone modifying proteins are related in regulating autophagy.

HDAC inhibitors induce autophagy in yeast, mouse embryonic fibroblasts

(MEFs), skeletal muscle cells, HeLa cells and glioblastoma cells; but they

inhibit autophagy in cardiomyocytes. HAT counterparts the function of HDAC.

Esa1, for example, can acetylate ATG3 to induce autophagy in yeast

(Fullgrabe et al., 2014).

MicroRNAs (miRNA) are also important regulators of autophagy process from

initiation, nucleation and elongation. Currently, the identified miRNA targets

are: ULK1 and FIP200 in initiation step; BECN1 and UVRAG in nucleation;

ATG3, ATG7, ATG10, ATG5, ATG12, ATG16, p62, LC3 during elongation.

There are no miRNAs identified yet to regulate proteins involved in fusion step

Page 29: @2020 Yang Yang ALL RIGHTS RESERVED

14

(Dikic and Elazar, 2018; Fullgrabe et al., 2014).

Physiological Role of Autophagy

Autophagy maintains organelle function, prevents the accumulation of toxic

cellular waste products, and sustains cell metabolism and survival during

starvation (Poillet-Perez and White, 2019). Autophagy is required to prevent

the accumulation of damaged mitochondria, which is particularly important in

the liver, muscle and brain. In fact, the buildup of damaged mitochondria can

lead to oxidative stress and perturbation of metabolism (Rabinowitz and White,

2010). Autophagy is also important for removal of damaged proteins,

functioning in coordination with proteasome degradation for protein quality

control(Pohl and Dikic, 2019). Autophagy defects lead to endoplasmic

reticulum (ER) stress and accumulation of chaperone proteins due to loss of

the ability to remove the unfolded protein and properly remodel the proteome in

response to stress (Mathew et al., 2009; Mathew et al., 2014). Under stress

conditions such as nutrient starvation, autophagy is dramatically induced and

essential for stress adaptation (Mizushima et al., 2004). Cargo-selective

autophagy is also important, for example, to recycle iron from ferritin, which is

critical for the iron homeostasis (Mancias et al., 2014).

Page 30: @2020 Yang Yang ALL RIGHTS RESERVED

15

Autophagy also has a critical role in mouse survival. Constitutively Atg5- or

Atg7-deficient mice are born developmentally normal but fail to survive the

neonatal starvation period when the trans-placental nutrient supply is

interrupted but not yet restored by milk (Komatsu et al., 2005; Kuma et al.,

2004). Force feeding only extends survival to neonatal starvation by 24 hours.

In contrast to newborn mice, adult mice have a greater tolerance to the loss of

autophagy. Conditional whole body ablation of the essential autophagy gene

Atg7 in adult mice shortens life span to two to three months due to susceptibility

to infection and neurodegeneration (Karsli-Uzunbas et al., 2014). Autophagy

also suppresses liver, brain and muscle damage. While adult mice tolerate

autophagy deficiency in the short term and in the fed state, fasting is lethal

within 16 hours due to hypoglycemia (Karsli-Uzunbas et al., 2014). These

findings demonstrate that autophagy is required to maintain systemic

metabolism and survival by mitigating metabolic stress during nutrient

deprivation (Karsli-Uzunbas et al., 2014). Moreover, there are remarkable

tissue-specific dependencies on autophagy with brain, liver, muscle and WAT

being particularly autophagy-dependent (Karsli-Uzunbas et al., 2014).

Autophagy also regulates intracellular lipid storage by breaking down

triglycerides and cholesterol stored in lipid droplets, which is lipophagy.

Impaired autophagy in liver causes accumulation of lipid droplets; in addition to

that, neurons, stellate cells and fibroblasts also require autophagy for lipid

Page 31: @2020 Yang Yang ALL RIGHTS RESERVED

16

breakdown(Liu and Czaja, 2013). Lipophagy activity varies in response to the

extracellular supply of nutrients. Free fatty acids and high-fat diet reduces

lipophagy through feedback inhibition (Singh et al., 2009). On the other hand,

autophagy plays a distinct role in regulating adipose lipid physiology.

Autophagy is essential to maintain fat mass and large lipid droplet in adipose

tissue instead of breaking down lipids. Adipocyte-specific Atg7 knockout mice

have decreased WAT and increased brown adipose tissue (BAT); and these

mice are resistant to high fat diet-induced obesity (Singh et al., 2009).

Autophagy is also required to maintain WAT in the whole body Atg7 knockout

mice (Karsli-Uzunbas et al., 2014).

Autophagy plays an important role in maintaining the tissue homeostasis

(Karsli-Uzunbas et al., 2014), and dysregulation of autophagy contributes to

different types of organ specific diseases. Neurodegenerative diseases are

tightly related with autophagy. The first hall mark for neurodegenerative

diseases is the aggregates of misfolded or unfolded proteins that accumulate

inside neuronal cells including Parkinson’s disease, Alzheimer’s disease and

Huntington’s disease (Rubinsztein et al., 2012). In healthy cells, misfolded

proteins are labeled with Ub and degraded by the proteasome. However,

proteasomal activity is impaired by various internal and external stresses and

declines with age. When the capacity of proteasome is overloaded, autophagy

is upregulated to compensate for the proteasome function and remove protein

Page 32: @2020 Yang Yang ALL RIGHTS RESERVED

17

aggregates as well as organelles that are damaged by these non-functional

aggregates (Dikic, 2017). Indeed, neurodegeneration accompanied with

accumulation of these undegraded ubiquitylated protein aggregates causes

death of autophagy deficient mice (Karsli-Uzunbas et al., 2014; Komatsu et al.,

2006). Besides, numerous mutated genes identified in neurodegenerative

diseases are related to autophagy (Gan-Or et al., 2015) and lysosome

function(Moors et al., 2016). p62, as a cargo receptor for autophagy, plays a

key role in bringing misfolded protein together for sequestration and then

transport them to the autophagosome (Pankiv et al., 2007; Seibenhener et al.,

2004). The second hallmark for neurodegenerative disease is the

accumulation of dysfunctional mitochondria, which elevates the ROS level that

damages the proteome and genome of the cell (Dikic and Elazar, 2018).

Damaged mitochondria can be removed by mitophagy using

ubiquitin-dependent PINK1-Parkin pathway as described in the previous

section, and mutations in PINK1 or Parkin are associated with the early onset

of Parkinson’s disease(Walden and Muqit, 2017). NIX functions as an

alternative ubiquitin independent cargo for mitophagy. Overexpression of NIX

restores mitophagy in Parkin- or PINK1-deficient cell lines derived from

patients with Parkinson disease (Koentjoro et al., 2017). Another protein

involved in regulating mitophagy in Parkinson’s disease is the TANK- binding

kinase 1 (TBK1). TBK1 phosphorylates ubiquitylated autophagy cargo

receptors including p62, OPTN, NDP52, enhances their binding affinity to Ub

Page 33: @2020 Yang Yang ALL RIGHTS RESERVED

18

on the cargo (here damaged mitochondria), LC3 on the autophagosome or

both, which contributes to the increased efficiency on recruitment of damaged

mitochondria to the autophagosome for degradation (Lazarou et al., 2015;

Matsumoto et al., 2015; Richter et al., 2016). Therefore, autophagy is a

beneficial machinery that counteracts the neurodegenerative diseases, and its

induction is currently being explored as a strategy for prevention or treatment

of neurodegenerative disease (Dikic and Elazar, 2018).

Other than neurodegenerative diseases, there’s evidence that autophagy is

related to cardiovascular disorders including ischemia, cardiomyopathy,

congestive heart failure and Danon’s disease. Autophagosome accumulation

was detected in myocardial biopsy samples from cardiovascular patients

(Terman and Brunk, 2005). Genetic defects in autophagy exacerbate the

propensity of mice to spontaneously develop cardiodegenerative disorders,

and alteration in autophagic flux affects disease outcome in rodent models of

several cardiovascular disorders including various types of cardiomyopathy,

myocardial infarction and atherosclerosis (Bravo-San Pedro et al., 2017).

Other organ-specific diseases such as cystic fibrosis muscle and liver

disorders are related to altered autophagy, which were mostly based on

decreased clearance of abnormal accumulation of defective organelles,

proteins and lipids (Schneider and Cuervo, 2014).

Page 34: @2020 Yang Yang ALL RIGHTS RESERVED

19

Moreover, autophagy regulates mammalian differentiation and development.

The very initial autophagic events in development happen in fertilized oocytes,

which are ATG5 dependent (Tsukamoto et al., 2008). After fertilization,

autophagy is profoundly activated in all the tissues to degrade maternal

mRNAs and proteins, except in the brain (Kuma et al., 2004). Autophagy is

upregulated during the neonatal period due to starvation, which is consistent

with the results that knockouts of Atg3, Atg5, Atg7, Atg9, and Atg16L1 cause

neonatal lethality. Interestingly, restoration of ATG5 in the brain is sufficient to

rescue Atg5 knockout mice, even though these mice display some

abnormalities including hypogonadism and iron-deficiency anemia, suggesting

that neuronal loss of Atg5 is the primary cause of death of these mice (Yoshii

et al., 2016). Absence of upstream autophagy regulators causes more severe

phenotypes. For example, beclin1, Ambra1, and FIP200 deficiency induce

embryonic lethality, probably due to autophagy-independent functions

(Mizushima and Levine, 2010). In addition to development, autophagy is

required for lymphocyte differentiation through NIX-dependent mitochondrial

clearance. Autophagy deficiency reduces T and B lymphocyte counts with

mitochondrial accumulation, and lymphocytes tend to increase superoxide and

sensitivity to apoptosis (Mortensen et al., 2010; Novak et al., 2010). Autophagy

is also required for the differentiation and survival of regulatory T cells (Wei et

al., 2016).

Page 35: @2020 Yang Yang ALL RIGHTS RESERVED

20

Autophagy has been demonstrated to involve in immune functions, including

inflammation control, inflammatory cytokine secretion, intracellular bacteria

removal and antigen presentation (Dikic and Elazar, 2018). Autophagy

deficient animals are susceptible to bacterial infection, and autophagy defects

are widely displayed in autoimmune diseases, such as diabetes, multiple

sclerosis, Crohn’s disease, systemic lupus erythematosus, rheumatoid arthritis,

and psoriasis (Karsli-Uzunbas et al., 2014; Rioux et al., 2007; Rockel and

Kapoor, 2016) . One of the major signaling pathway on inflammation which

crosstalks with autophagy is the IκB kinase complex (IKK)-NF-κB pathway.

NF-κB induced autophagy by transactivating BECN1, BCL2 and p62 (Copetti

et al., 2009; Jain et al., 2010; Niture and Jaiswal, 2012), while IKK induces

autophagy in the presence of stress signals (Criollo et al., 2010). However,

NF- κB pathway may inhibit autophagy in macrophages infected by E.coli and

tumor necrosis factor- α (TNFα)-induced cell death (Djavaheri-Mergny et al.,

2006; Schlottmann et al., 2008). On the other hand, autophagy can suppress

NF-κB signaling by the autophagic degradation of active IKKβ mediated either

by E3 ubiquitin- protein ligase Kelch- like ECH- associated protein 1(KEAP1)

or RO52(Kim et al., 2010; Niida et al., 2010).

Autophagy can also eliminate a variety of invading bacteria including

Mycobacteria , Salmonella, Shigella, Legionella, Listeria and group A

streptococcus(Gomes and Dikic, 2014). Once these bacteria invade into the

Page 36: @2020 Yang Yang ALL RIGHTS RESERVED

21

cytosol, they are labeled with various types of Ub chain and galectin and

sequestered by autophagic membranes utilizing the cargo receptors such as

p62, NDP52 and OPTN (Gomes and Dikic, 2014). The Ub modifications on the

bacteria coat is not only the target of autophagy cargo receptors, it also

transforms the bacteria surface into a signaling platform to attract other

proteins. The linear Ub chains, for example, activate the NF-κB signaling as

well as attracting autophagy cargo receptors to maximize the antibacterial

response (Noad et al., 2017; van Wijk et al., 2017). Interestingly, some

pathogens develop their strategies to avoid the autophagy machinery by

blocking the fusion of autophagosome with lysosome (Nguyen et al., 2005),

competing with host LC3-interacting proteins for LC3 binding (Real et al.,

2018), or secreting factors to activate host kinases to interfere autophagosome

maturation (Neumann et al., 2016). Some bacteria are even able to take use of

the autophagy machinery and replicate in the autophagosome-like

vesicles(Devenish and Lai, 2015). However, generally autophagy is still

considered a valid way to combat bacterial infections.

p53 and Autophagy

Many major stress responses are controlled by p53, and there is mounting

evidence for a functional interaction between the p53 and the autophagy

pathways. p53 is a transcription factor and tumor suppressor that responds to

Page 37: @2020 Yang Yang ALL RIGHTS RESERVED

22

diverse types of stresses including DNA damage, oncogene activation,

oxidative stress and hypoxia (Fischer, 2017). In response to stress p53 can

induce apoptosis, senescence, and cell cycle arrest, and alter cell metabolism

by regulating multiple p53 target genes (Toledo and Wahl, 2006). It is generally

thought that the p53 stress response can provide either protection and facilitate

adaptation and recovery (e.g. cell cycle arrest) in the case of mild stress, or can

eliminate cells (e.g. apoptosis) with excessive damage in the setting of high

levels of stress (Kruiswijk et al., 2015). p53 thereby controls the nature of the

stress response and its outcome.

p53 can also regulate autophagy. Under nutrient deprivation, a low ATP: AMP

ratio activates AMPK, which then induces p53. Induction of p53 activates the

transcription of genes in the AMPK pathway including TSC2 and AMPK itself,

and leads to the inhibition of mTOR and activation of autophagy (Feng et al.,

2007). Some p53 target genes like BCL2-associated X protein (BAX) and

p53-upregulated modulator of apoptosis (PUMA) can directly activate

autophagy in MEF cells (Yee et al., 2009). p53 can also directly turn on the

expression of essential autophagy genes or induce autophagy via

transcriptional activation of Damage-Regulated Autophagy Modulator

(DRAM-1) in human and mouse cell lines (Crighton et al., 2006; Kenzelmann

Broz et al., 2013; Mah et al., 2012).

Page 38: @2020 Yang Yang ALL RIGHTS RESERVED

23

CHAPTER 2

MATERIALS AND METHODS

Page 39: @2020 Yang Yang ALL RIGHTS RESERVED

24

Mouse models

All animal care was carried out in compliance with Rutgers University

Institutional Animal Care and Use Committee guidelines. Ubc-CreERT2/+ mice

(Ruzankina et al., 2007)(The Jackson Laboratory) and Atg7flox/flox

mice(Komatsu et al., 2005) (provided by Dr. M. Komatsu, Tokyo Metropolitan

Institute of Medical Science) were cross-bred to generate the Ubc-Cre ERT2/+;

Atg7flox/flox mice as previously described (Karsli-Uzunbas et al., 2014). To

generate Ubc-CreERT2/+; Atg7flox/flox; p53flox/flox mice, p53flox/flox mice (Marino et

al., 2000) (The Jackson Laboratory) were cross-bred with our previously

created Ubc-Cre ERT2/+; Atg7flox/flox mice. To generate Nrf2-/-; Ubc-CreERT2/+ mice

and Nrf2-/-; Ubc-CreERT2/+; Atg7flox/flox mice, Nrf2-/- mice (Chan et al., 1996)

(provided by YW Kan, UCSF) were cross-bred with Ubc-CreERT2/+ mice and our

previously generated Ubc-CreERT2/+; Atg7flox/flox mice. To generate Ubc-CreERT2

/+; Atg5flox/flox mice, Ubc-CreERT2 mice was cross-bred with Atg5flox/flox mice (Hara

et al., 2006). To generate Ubc-CreERT2 /+; Atg5flox/flox; Trp53flox/flox mice,

Trp53flox/flox mice (Marino et al., 2000)(The Jackson Laboratory) were

crossbred with previously generated Ubc-CreERT2 /+; Atg5flox/flox mice.

Cag-CreERT2; Atg5flox/flox mice and Cag-CreERT2; Atg12flox/flox mice were provided

by Dr.J.Debnath (University of California San Francisco). To generate

PDGFRα-CreERT/+; Atg5flox/flox mice and PDGFRα-CreERT/+ ; Atg7flox/flox mice,

PDGFRα-CreERT/+ mice(provided by Dwight E Bergles, Johns Hopkins School

of Medicine)(The Jackson Laboratory) was cross-bred with Atg5flox/flox mice and

Page 40: @2020 Yang Yang ALL RIGHTS RESERVED

25

Atg7flox/flox mice.

TAM administration

For acute deletion of Atg7 and/or p53, detailed rationale and TAM preparation

is described as previously published (Karsli-Uzunbas et al., 2014). For TAM

delivery to the adult mice, 200 μl of the suspended solution per 20 g of body

weight equals (20mg/kg) were injected intraperitoneally (IP) into 8-10 weeks

old Ubc-CreERT2/+; Atg7flox/flox mice and Ubc-CreERT2/+; Atg7flox/flox; p53flox/flox mice

once per day for 5 consecutive days to delete the floxed gene systematically.

Additionally, same dosage of TAM were given to Ubc-CreERT2/+ mice and

Ubc-CreERT2/+; p53flox/flox mice and these mice were examined as control

groups. For Atg5 related experiment, same amount of TAM were injected

intraperitoneally (IP) into 8-10 weeks old Ubc-CreERT2/+; Atg5flox/flox mice,

Ubc-CreERT2 /+; Atg7flox/flox mice, PDGFRα-CreERT/+; Atg5flox/flox mice and

PDGFRα-CreERT/+; Atg7flox/flox mice once per day for 5 consecutive days. For

slow deletion, same amount of TAM was used once per week for 4 shots to

delete the floxed gene systematically.

Survival

For mouse Kaplan-Meyer survival curve, mice were monitored daily until they

reached the endpoint. Cag-CreERT2; Atg12flox/flox mice were sacrificed 70 days

Page 41: @2020 Yang Yang ALL RIGHTS RESERVED

26

after the last TAM injection because they met the criteria for euthanization with

a body condition score of 2 and defects in grooming.

Fasting

Fasting was conducted as previous described (Karsli-Uzunbas et al., 2014).

Histology

Mouse tissues were collected and fixed in 10% formalin solution

(Formaldehyde Fresh, Fisher Scientific, SF94-4). Tissues were fixed overnight

and then transferred to 70% ethanol for paraffin-embedded sections or 15%

sucrose following by 30% sucrose for frozen sections. For Alcian blue staining,

paraffin sections were processed with the Alcian blue stain kit (Abcam)

following the manufacturer’s protocol. For Bodipy C11 (Thermo Scientific,

D3861) stain, 5μM of Bodipy C11 dye were used to stain the frozen sections of

intestine for 30 minutes and counterstained with DAPI. For IHC, paraffin

sections were stained with antibodies against p53 (Novus Biologicals,

NB200-103, 1:400), Active caspase-3 (Cell Signaling, 9661, 1:300), γ-H2AX

(Cell Signaling, 9718, 1:480), MDA (Cosmo Bio USA, NOF-N213530-EX,

1:200), OLFM4 (Cell Signaling, 39141, 1:2000), Lysozyme (Agilent, A0099,

1:2000), Active β-Catenin (Cell Signaling, 8814, 1:3000). For quantification of

IHC on p53, Active caspase-3 and γ-H2AX, the liver and brain tissue were

Page 42: @2020 Yang Yang ALL RIGHTS RESERVED

27

analyzed by quantifying at least 10 images at 60X magnification using ImageJ.

A minimum of 100 cells in liver and brain were scored for each image. For

Immunofluorescence, paraffin sections were stained with antibodies against

p62 (Novus 00008878, 1:1000), OLFM4 (cell signaling, 39141, 1:2000), CD34

(Abcam, 81289, 1:150), PDGFRα (R&D systems, AF1062, 1:100) and pictures

were taken using confocal microscope.

Nutlin-3a administration

TAM were injected via IP into 8-10 weeks old Ubc-CreERT2/+; Atg7flox/flox mice

and Ubc-CreERT2/+; Atg7flox/flox; p53flox/flox mice once per day for 5 consecutive

days. Additionally, same dosage of TAM were given to Ubc-CreERT2/+ mice and

Ubc-CreERT2/+; p53flox/flox mice. After two weeks, 200mg/kg of Nutlin-3 (Cayman

Chemicals) resolved in 50% DMSO were delivered to the mice by oral gavage

once per day for 7 consecutive days. Mice were sacrificed one day after last

administration of Nutlin-3 and tissues were collected for histology and

snap-frozen for western blot and real-time PCR.

Real-time PCR

Total RNA were isolated from tissue by Trizol (Invitrogen). cDNA were then

reverse transcribed from the total RNA by MultiScribe RT kit (Thermo Fisher).

Real-time PCR were performed on Applied Biosystems StepOne Plus

Page 43: @2020 Yang Yang ALL RIGHTS RESERVED

28

machine. Atg7 and p53 were performed using SYBR green for deletion

detection (Atg7: Forward 5’ ACTTGACCGGTCTTACCCTG 3’; Reverse 5’

TACTCCTGAGCTGTGGTTGC 3’ p53: Forward: 5’

CGACTACAGTTAGGGGGCAC 3’; Reverse: 5’

GGAGGAAGTAGTTTCCATAAGCCT 3’; Actin: Forward 5’

GAACCCTAAGGCCAACCGTGAAAAGATGAC 3’; Reverse 5’

GCAGGATGGCGTGAGGGAGAGCA 3’). Besides that, all the other genes

were detected using predesigned commercial Taqman primers for each gene

accordingly (Cdkn1a: Mm00432448-m1; Actin: Mm00607939-s1; Uvrag:

Mm00724370-m1; Ulk1: Mm0437238-m1; Ulk2: Mm03048846-m1; Vmp1:

Mm00774656-m1; Atg2b: Mm00620760-m1; Atg4a: Mm04214755-s1; Atg4c:

Mm00558175-m1; Atg10: Mm00470550-m1). Results were calculated using

ΔΔCT method and then normalized to actin. For real time PCR on stem cell

markers and Wnt target genes, primers (information kindly from Dr. Michael

Verzi, Rutgers University) are:

OLFM4(Forward:5’ GCCAGATCTTGGCTCTGAAG 3’; Reverse: 5’

GCCAGTTGAGCTGAATCACA 3’)

Lgr5(Forward:5’ ACGTCTTGCTGGAAATGCTT 3’; Reverse: 5’

CACGTAGCTGATGTGGTTGG 3’)

Axin2(Forward:5’ AGGAGCAGCTCAGCAAAAAG 3’; Reverse: 5’

GCTCAGTCGATCCTCTCCAC 3’)

Sox9(Forward:5’ AGTCGGTGAAGAACGGACAA 3’; Reverse: 5’

Page 44: @2020 Yang Yang ALL RIGHTS RESERVED

29

CCCTCTCGCTTCAGATCAAC 3’)

actin (Forward:5’ TGTTACCAACTGGGACGACA 3’; Reverse: 5’

CCATCACAATGCCTG TGGTA 3’)

Western blot

Different tissues were grounded in a Cryomill machine (Retsch) and then total

protein extracts were isolated by Tris lysis buffer (1 mol/L Tris HCl, 1 mol/L

NaCl, 0.1 mol/L EDTA, 10% NP40). Separated proteins were probed with

antibodies against ATG7 (Sigma; A2856, 1:2000), ATG5 (Abcam, ab108327,

1:1000), LC3 (Novus Biologicals; NB600-1384, 1:1500), p62 (American

Research Products, 03-GP62-C, 1:2000), GAPDH (Santa Cruz; sc-365062,

1:1000) and β-actin (Sigma; A1978, 1:5000).

Organoid culture

Organoid culture was performed following standard protocol with modifications

(Sato et al., 2009). Generally, crypts from the ileum part of the murine intestine

were incubated for 40 min at 4℃ in PBS containing 2mM EDTA. Then crypts

were pelleted and then plated in 48 well plates with a total of 250 crypts in 25μl

of Matrigel (Trevigen, 3433-05). After gel solidified in incubator, 250μl of

culture medium (Advanced DMEM/F12 (Thermo Scientific, 12634-010))

containing 50ng/mL of EGF(Peprotech,315-09), 500ng/mL of human

Page 45: @2020 Yang Yang ALL RIGHTS RESERVED

30

R-spondin 1(Peprotech, 120-38) and 100ng/ml of Noggin (Peprotech, 250-38)

were added. For in vitro Atg5 and Atg7 deletion, 1μM of 4-hydroxy-TAM

(Sigma, H7905) were added to the 48 well plates for 15 hours and then

changed back to normal culture medium. For in vitro Wnt ligand

supplementation, Wnt3a (Abcam, ab81484) or Wnt2b (R&D systems,

3900-WN-025) were added with a concentration of 100ng/ml.

In vivo Wnt ligand supplementation

Mice at the age of 8-10 weeks were first treated with TAM as mentioned before.

Wnt3a or Wnt 2b were administered to the mice by intraperitoneal injection

with an amount of 50mg/kg per mice, 2 shots per day starting from the last day

of TAM injection as previously described (Valenta et al., 2016). Mice were then

monitored twice per day for survival curve and sacrificed to collect tissues

when they reached the end point.

Glucose supplementation

To assess if the glucose can rescue the lethality caused by acute ATG5

deletion, mice were supplemented with 1mg/g of glucose twice per day via

intraperitoneal injection starting from the last day of TAM treatment. Blood

glucose level was measured daily using blood glucometer (One Touch Ultra 2).

Page 46: @2020 Yang Yang ALL RIGHTS RESERVED

31

FITC dextran supplementation

In order to test the intestinal barrier function, mice were supplemented with

FITC-dextran and the blood serum was collected to measure the FITC-dextran

concentration using the fluorescence plate reader as previous described

(Gupta and Nebreda, 2014).

Single cell RNA sequencing for PDGFRα+ mesenchymal cells

Single cell RNA sequencing for telocytes were conducted as previously

described (Halpern et al., 2019).

Matrix-assisted laser desorption ionization coupled to imaging mass

spectrometry (MALDI-IMS)

Ileum tissues are snap frozen in liquid nitrogen and stored at -80C until

imaging mass-spectrometry and/or IHC/IF staining. For MALDI sectioned

slides are desiccated and coated with a matrix compatible with MALDI-IMS

(e.g. 9-AA or DHB) and mass spectrometry data is acquired using a Bruker

solariX FTMS at 30-50µM spatial resolution. Conventional procedures for

IHC/IF are used and data acquired from both modalities are used to determine

metabolite levels using antibodies directed against PDGFRα+ mesenchymal

Page 47: @2020 Yang Yang ALL RIGHTS RESERVED

32

cells.

We will confirm the results from imaging spectrometry by alternative

techniques, including LC-MS and fluorescence imaging. For LC-MS we will

dissect comparable regions as in IMS to perform quantitative analysis of

candidate metabolites identified by MALDI and global metabolomics.

Qualitative data analysis will be conducted using Bruker FlexImaging software.

Relative intensity of individual metabolites of interest will be determined for

preliminary studies and standards. For quantitative data analysis approaches,

we have developed an analysis pipeline that analyzes IMS data in three stages:

1) Pre-processing, 2) Peak picking, and 3) Post-processing. In the

Pre-processing stage, we subtract background noise and matrix peaks, scan

for potential adducts of analytes of interest, and perform spectrum alignment.

In the peak picking stage, human-assisted automatic integration is performed

to quantitate (relative quantification where no standards are run and absolute

quantitation where standards are run) the peak area and intensity of the

labeled and unlabeled metabolites of interest. All selected peak areas will be

corrected for natural isotope abundance. In the post-processing stage, IMS

data is overlaid with Hematoxylin and eosin (H&E) or

immunohistochemical/immunofluorescence data. In addition, the post

processing stage employed image fusion algorithms which infer structural

information and metabolite localization from the overlaid images. Finally, we

Page 48: @2020 Yang Yang ALL RIGHTS RESERVED

33

will use the data generated in the post-processing stage to obtain spatial

resolution of metabolic fluxes using metabolites (labeled and unlabeled).

Relative quantification of metabolites is performed as part of the normal

procedure, and absolute quantification of metabolites will be performed for key

metabolites of interest using chemical standards at known concentration

dispensed on tissue sections.

Page 49: @2020 Yang Yang ALL RIGHTS RESERVED

34

CHAPTER 3

AUTOPHAGY PROMOTES MAMMALIAN SURVIVAL BY SUPPRESSING

OXIDATIVE STRESS AND P53

Page 50: @2020 Yang Yang ALL RIGHTS RESERVED

35

Introduction

Autophagy deficiency can cause p53 induction in mouse models of lung and

pancreatic and breast cancer, and also neurons, correlating with more

apoptosis when p53 is intact, suggesting that autophagy may suppress p53

activation in some but not all cancer settings (Guo et al., 2013; Huo et al., 2013;

Rosenfeldt et al., 2013; Strohecker et al., 2013; Yang and Kimmelman, 2014;

Yang et al., 2011; Zhang et al., 2009). As autophagy loss promotes p53

activation, and this p53 activation can be damaging, we sought to test the

hypothesis that p53 was responsible for degenerative phenotypes induced by

conditional autophagy loss in vivo.

To address how p53 and autophagy functionally interact in vivo and to

determine the role that p53 plays in limiting the survival of mice without

autophagy, we developed genetically engineered mouse models (GEMMs) to

conditionally delete Atg7 and/or p53 systemically with tamoxifen (TAM).

Whereas conditional, systemic Atg7 deletion (Atg7Δ/Δ) in adult mice limited their

survival to two to three months, co-deletion of p53 and Atg7 (Atg7Δ/Δ p53Δ/Δ)

remarkably extended lifespan to up to six months. Atg7Δ/Δp53Δ/Δ mice showed

decreased tissue damage, apoptosis, and DNA damage in liver and brain in

comparison to Atg7Δ/Δ mice. Activation of p53 by Nutlin-3 was inhibited by

autophagy, which prevented liver and brain from p53 hyperactivation and

apoptosis, suggesting that autophagy may be a resistance mechanism to

Page 51: @2020 Yang Yang ALL RIGHTS RESERVED

36

p53-activators. NRF2, in turn, is a resistance mechanism to loss of autophagy

as conditional deletion of both Nrf2 and Atg7 in adult mice was synthetically

lethal. Mice deficient for both Atg7 and Nrf2 (Nrf2-/-Atg7Δ/Δ) succumbed to

damage to the small intestine, which was independent of p53 function. Thus,

Atg7 protects against excessive p53 activation and damage in the liver and

brain, whereas NRF-2 protects the intestine from damage upon loss of Atg7,

demonstrating the functional interdependence and tissue specificity of stress

response pathways.

Page 52: @2020 Yang Yang ALL RIGHTS RESERVED

37

Results

Loss of p53 delays neurodegeneration and prolongs survival of

Atg7-deficient mice.

To test whether p53 plays a role in limiting the survival in mice without

autophagy, adult mice were engineered with or without floxed alleles of Atg7

(Kuma et al., 2004), p53 (Marino et al., 2000) and a transgene expressing a

TAM-regulated Cre recombinase under the control of Ubiquitin C promoter that

is ubiquitously expressed in the whole-body (Ubc-CreERT2) (Ruzankina et al.,

2007). Injecting TAM activates Cre throughout these mice and the floxed alleles

of Atg7 and/or p53 are deleted separately or together (Fig. 3.1A). Mice with

systemic loss of Atg7 or p53 or both in all tissues are thereby generated and

gene deletion was confirmed by qRT-PCR at two, five and eight weeks

following the five consecutive days of TAM administration (Fig. S3.1A). Loss of

ATG7 protein expression was also associated with accumulation of

unprocessed form of microtubule-associated protein 1A/1B light chain 3

(LC3-I), decreased or absence of the processed (active) form of LC3 (LC3-II),

and accumulation of the autophagy substrate protein p62, in both Atg7Δ/Δ and

Atg7Δ/Δp53Δ/Δ mice, indicating blockage of autophagy function (Fig. 3.1B).

Atg7Δ/Δ mice had a lifespan of about two to three months primarily due to

susceptibility to infection early, and to neurodegeneration later, which is

consistent with our previous findings (Karsli-Uzunbas et al., 2014). Similar to

constitutively deficient p53-/- mice, p53Δ/Δ mice died from lymphoma, which

Page 53: @2020 Yang Yang ALL RIGHTS RESERVED

38

limited lifespan to up to six months (Fig. 3.1C, D) (Donehower et al., 1995). In

contrast to Atg7Δ/Δ mice, a third of the Atg7Δ/Δp53Δ/Δ mice lived longer than three

months and up to six months post TAM, while all the Atg7Δ/Δ mice died before

three months post TAM (Fig. 3.1C, D). Although Atg7Δ/Δp53Δ/Δ lived longer than

Atg7Δ/Δ mice, death was still predominantly from neurodegeneration (Fig. 3.1D).

As loss of p53 did not alter survival to Atg7 deficiency early post deletion where

death is due to susceptibility to infection (Karsli-Uzunbas et al., 2014), the role

of p53 was specific to promoting death due to neurodegeneration (Fig. 3.1C).

Therefore, p53 promotes neurodegeneration in mice deleted for Atg7.

p53 deficiency reduces tissue damage in Atg7Δ/Δ mice

Histological examination (H&E) of tissues from wild type, Atg7Δ/Δ, p53Δ/Δ, and

Atg7Δ/Δp53Δ/Δ mice revealed no differences two weeks post TAM (Fig. S3.1B).

At five weeks post TAM, Atg7Δ/Δ mice began to show early evidence of loss of

hepatocytes in liver, pyramidal neurons in the cerebrum, Purkinje cells in the

cerebellum, and depletion of lipid in WAT as reported previously

(Karsli-Uzunbas et al., 2014), which was not observed in the p53Δ/Δ or

Atg7Δ/Δp53Δ/Δ mice (Fig. S3.1C). Two months post TAM, Atg7Δ/Δ mice showed

severe loss of hepatocytes, pyramidal neurons, Purkinje cells, and WAT, and

muscle wasting, whereas kidney and lung were not affected (Fig. 3.1E, S3.1D)

(Karsli-Uzunbas et al., 2014). In contrast, these tissue damage phenotypes

Page 54: @2020 Yang Yang ALL RIGHTS RESERVED

39

resulting from Atg7 deficiency were not observed in wild type, p53Δ/Δ and

Atg7Δ/Δp53Δ/Δ mice (Fig. 3.1E, S3.1D). These results suggest that tissue

damage caused by autophagy deficiency is induced by p53. Atg7Δ/Δp53Δ/Δ mice

did display the same phenotype as the Atg7Δ/Δ mice at three to six months post

depletion (data not shown), indicating that loss of p53 delays but does not

prevent lethal neurodegeneration caused by autophagy deficiency.

ATG7 is required for tumorigenesis driven by p53 deletion

Constitutive p53 deficiency leads to the development of lethal thymic

lymphomas, which limits lifespan to about six months (Donehower et al., 1995).

Here, conditional p53 deficiency in adult mice (p53Δ/Δ) produced the same

phenotype as 36 out of 39 mice died of thymic lymphoma (Fig. 3.1D).

Atg7Δ/Δp53Δ/Δ mice showed similar lifespan limitation as the p53Δ/Δ mice,

however, the vast majority of the mice died from neurodegeneration without

tumor development as 18 out of 21 mice died of neurodegeneration (Fig. 3.1D).

Of the three Atg7Δ/Δp53Δ/Δ mice that died from cancer, two died from thymic

lymphomas and one died from a sarcoma (Fig. 3.1D). Thus, autophagy is

required to enable development of lethal thymic lymphoma driven by deletion of

p53.

Page 55: @2020 Yang Yang ALL RIGHTS RESERVED

40

p53 deficiency prevents lethality in Atg7Δ/Δ mice during fasting

While Atg7Δ/Δ mice survive in the short term, fasting is immediately lethal due to

hypoglycemia (Karsli-Uzunbas et al., 2014). Since p53 deficiency extended the

lifespan and attenuated tissue damage in Atg7Δ/Δ mice, we sought to test

whether p53 contributes to the death of Atg7Δ/Δ mice during fasting. In contrast

to Atg7Δ/Δ mice where fasting was lethal, none of the Atg7Δ/Δp53Δ/Δ mice died

upon fasting, suggesting that p53 was responsible for fasting-induced death of

Atg7-deficient mice (Fig. 3.1F).

Page 56: @2020 Yang Yang ALL RIGHTS RESERVED

41

Page 57: @2020 Yang Yang ALL RIGHTS RESERVED

42

Fig. 3.1 Atg7Δ/Δ, p53Δ/Δ mice have extended life span, delayed tissue

damage and neurodegeneration compared to Atg7Δ/Δ mice. A.

Experimental design for generation of Atg7Δ/Δ mice, p53Δ/Δ mice, and Atg7Δ/Δ

p53Δ/Δ mice. Ubc-CreERT2/+, Ubc-CreERT2/+; Atg7flox/flox mice, Ubc-CreERT2/+;

p53flox/flox, Ubc-CreERT2/+; p53flox/flox; Atg7flox/flox mice were treated with TAM at 8

to 10 weeks age and analyzed at certain time points afterwards. B. Western

blot for ATG7, p62, and LC3 at the indicated time points of the indicated tissues

from wild type mice, Atg7Δ/Δ mice, p53Δ/Δ mice, and Atg7Δ/Δp53Δ/Δ mice. β-Actin

is used as a loading control. C. Kaplan–Meier survival curve of wild type mice,

Atg7Δ/Δ mice, p53Δ/Δ mice, and Atg7Δ/Δp53Δ/Δ mice. * p<0.05, ** p<0.01, ****

p<0.0001(Log-rank (Mantel-Cox) test and Gehan-Breslow-Wilcoxon test as

indicated).D. Percentage distribution for the cause of death of Atg7Δ/Δ, p53Δ/Δ,

and Atg7Δ/Δp53Δ/Δ mice. The cause of death was analyzed at 30-90 days post

TAM and 90-180 days post TAM. E. Representative histology of liver, muscle,

cerebrum, cerebellum, pancreas, white adipose tissue (WAT) and lung by

Hematoxylin & Eosin stain (H&E) from wild type, Atg7Δ/Δ, p53Δ/Δ, and

Atg7Δ/Δp53Δ/Δ mice at 8 weeks time point. F. Kaplan–Meier survival curve of wild

type mice, Atg7Δ/Δ mice, p53Δ/Δ mice, and Atg7Δ/Δp53Δ/Δ mice during starvation

at 10 days post TAM. * p<0.05. See also Figure S3.1

Page 58: @2020 Yang Yang ALL RIGHTS RESERVED

43

ATG7 is required to protect liver and brain from p53-mediated damage

Autophagy deficiency causes p53 induction in neurons and in some cancer

models and promotes cell death. Therefore, we tested whether p53 induction

occurred in the whole-body after Atg7 deletion. Immunohistochemistry (IHC) for

p53 protein revealed that p53 accumulation was detectable at two weeks post

TAM administration, and was maintained at five and eight weeks post TAM in

the livers and brains of Atg7Δ/Δ mice, while p53 activation was not apparent in

wild type, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice (Fig. 3.2A, S3.2A, S3.2B). qRT-PCR

for the p53 target gene cyclin-dependent kinase inhibitor 1A (Cdkn1a, or p21)

showed increased Cdkn1a expression in Atg7Δ/Δ mice at two, five and eight

weeks post TAM in liver and brain compared to wild type, p53Δ/Δ and

Atg7Δ/Δp53Δ/Δ mice (Fig. 3.2B, C). These data suggest that loss of Atg7

promotes activation of p53.

Whole-body ATG7 deficiency leads to DNA damage and apoptosis in liver and

cerebrum (Karsli-Uzunbas et al., 2014), and p53 is known to be activated by

different stress signals including DNA damage, oxidative stress and triggers

cell cycle arrest and apoptosis (Fischer, 2017). Therefore, we hypothesized

that p53 induction in Atg7Δ/Δ mice may promote apoptosis. IHC for the DNA

damage response activation marker γ-H2AX revealed accumulation of γ-H2AX

in Atg7Δ/Δ liver and cerebrum starting two weeks that was apparent through

eight weeks post TAM (Fig. 3.2D, S3.2C, S3.2D). In contrast, γ-H2AX

accumulation was not detected in wild type, p53Δ/Δ and Atg7Δ/Δp53Δ/Δ mice (Fig.

Page 59: @2020 Yang Yang ALL RIGHTS RESERVED

44

S3.2C, S3.2D). As a likely consequence of p53 activation in Atg7Δ/Δ mice, these

mice also showed more apoptosis marked by increased active caspase-3 in

liver and brain in comparison to Atg7Δ/Δp53Δ/Δ mice (Fig. 3.2D, S3.2E, S3.2F).

These data indicated that p53 induction caused apoptosis in Atg7Δ/Δ mice.

Atg7Δ/Δ mice also displayed increased Malondialdehyde (MDA) in liver by IHC

compared to wild type, p53Δ/Δ and Atg7Δ/Δp53Δ/Δ mice, indicating that p53

induction was associated with increased oxidative stress in Atg7Δ/Δ mice (Fig.

3.2E).

Page 60: @2020 Yang Yang ALL RIGHTS RESERVED

45

Page 61: @2020 Yang Yang ALL RIGHTS RESERVED

46

Fig. 3.2. Autophagy is required to protect liver and brain from p53

accumulation, DNA damage response activation and apoptosis. A.

Representative liver and cerebrum IHC staining of p53 and quantification at

indicated time points from wild type and Atg7Δ/Δ mice. 2w, 2 weeks time point.

5w, 5 weeks time point. 8w, 8 weeks time point. B-C. Quantitative real-time

PCR of Cdkn1a for liver and brain tissues from wild type, Atg7Δ/Δ, p53Δ/Δ, and

Atg7Δ/Δp53Δ/Δ mice at indicated time points. D. Representative liver and

cerebrum IHC staining for γ-H2AX and active caspase-3 with quantification at

indicated time points from wild type and Atg7Δ/Δ mice. 2w, 2 weeks. 5w, 5

weeks. 8w, 8 weeks. *p<0.05; **p<0.01, ***p<0.001, ****p<0.0001(Unpaired t

test) E. Representative liver IHC staining for MDA at indicated time points from

wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice. See also Figure S3.2.

Page 62: @2020 Yang Yang ALL RIGHTS RESERVED

47

ATG7 limits p53 activation by Nutlin-3a

Regulation of p53 activity relies on the essential p53 antagonist MDM2, which

is a direct transcriptional target of p53 and is upregulated when p53 is activated

by phosphorylation at specific serine and threonine residues (Bode and Dong,

2004). MDM2 binds to p53, and the ubiquitin E3 ligase of MDM2 ubiquitinylates

p53, which decreases its stability by targeting it to the proteasome for

degradation (Honda et al., 1997; Kubbutat et al., 1997; Matsumine et al., 1997).

Under stress conditions, p53 is released from MDM2, and is thereby stabilized

and activated. In this way, p53 and MDM2 form a negative feedback loop

resulting from p53-dependent induction of MDM2 and MDM2-dependent

suppression of p53 activity, which helps the cell to deal with stress without

hyperactivation of p53 (Dotto, 2009; Marine and Lozano, 2010; Montes de Oca

Luna et al., 1995). Nutlin-3 works as an MDM2 antagonist and upregulates the

cellular p53 level by competing for the binding site of MDM2 for p53, and is

being assessed clinically to promote p53 activation for cancer therapy (Vassilev

et al., 2004). Since Atg7 limits p53 accumulation and activation we sought to

test if Atg7 also limited the ability of Nutlin-3 to activate p53 (Khoury and

Domling, 2012) as a potential resistance mechanism.

Following deletion of Atg7 and/or p53, mice were either treated with vehicle or

Nutlin-3 (200mg/kg) once per day for one week (Fig. 3.3A). Deletion of Atg7

and p53 were confirmed by qRT-PCR (Fig. S3.3A). Western blot for loss of

Page 63: @2020 Yang Yang ALL RIGHTS RESERVED

48

ATG7 protein, accumulation of LC3-I and loss of LC3-II, and accumulation of

p62 in liver and brain from Atg7Δ/Δ and Atg7Δ/Δp53Δ/Δ mice indicated blockage of

autophagy (Fig. 3.3B). As previously described, liver damage and neuron loss

in Atg7Δ/Δ mice were confirmed by H&E, which was not affected by Nutlin-3 (Fig.

S3.3B). IHC of liver and brain from Atg7Δ/Δ mice revealed increased p53

compared to wild type mice, which was further increased in Nutlin-3 treated

Atg7Δ/Δ mice, suggesting that Nutlin-3 induced p53 in the absence but not in the

presence of autophagy. As expected, Nutlin-3 did not affect p53 levels in p53Δ/Δ

mice and Atg7Δ/Δp53Δ/Δ mice (Fig. 3.3C, S3.3C). qRT-PCR for the p53 target

gene Cdkn1a showed increased Cdkn1a expression in untreated Atg7Δ/Δ mice,

which was further increased in Nutlin-3 treated Atg7Δ/Δ mice in liver and brain

compared to vehicle or Nutlin-3 treated wild type mice (Fig. 3.3D). This

confirmed that p53 was activated by Nutlin-3 specifically in the absence of

autophagy. IHC of liver and brain revealed increased γ-H2AX and active

caspase-3 in Atg7Δ/Δ mice compared to wild type mice, and activation of p53 by

Nutlin-3 greatly increased γ-H2AX and active caspase-3 levels. Induction of

γ-H2AX and active caspase-3 were not observed in p53Δ/Δ and Atg7Δ/Δ p53Δ/Δ

mice, suggesting that loss of autophagy induced apoptosis through p53

activation (Fig. 3.3E, Fig. S3.3D, S3.3E). Therefore, autophagy is essential to

protect tissues from apoptosis by limiting p53 activation.

Since p53 can induce a series of essential autophagy genes including Atg7 in

Page 64: @2020 Yang Yang ALL RIGHTS RESERVED

49

MEF cells (Kenzelmann Broz et al., 2013), we hypothesized that upregulation

of p53 by Nutlin-3 can turn on essential autophagy genes and protect tissues

from damage caused by p53 induction in wild type mice. Real-time PCR on a

series of autophagy essential genes indicated no significant difference in the

autophagy gene mRNA levels, suggesting that the autophagy transcription

program is not detectably induced by p53 at the times the tissues were

collected (Supplementary Fig. S3.4).

Page 65: @2020 Yang Yang ALL RIGHTS RESERVED

50

Page 66: @2020 Yang Yang ALL RIGHTS RESERVED

51

Fig. 3.3 Activation of p53 by MDM2 antagonist Nutlin-3a in Atg7Δ/Δ mice

leads to further increased DNA damage response and apoptosis in liver

and brain. A. Experimental design for generation of Atg7Δ/Δ, p53Δ/Δ, and

Atg7Δ/Δp53Δ/Δ mice and Nutlin-3 administration. Nutlin-3 was administered to

mice by oral gavage 2 weeks post TAM administration at a dosage of 200mg/kg

for one week. B. Western blot for ATG7, p62 and LC3 for liver and brain tissues

from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice treated with vehicle or

Nutlin-3. β-Actin is used as a loading control. V, treated with Vehicle; N, treated

with Nutlin-3. C. Representative liver and cerebrum IHC staining for p53 and

quantification at indicated time points from wild type and Atg7Δ/Δ mice treated

with vehicle or Nutlin-3. V, Vehicle; N, Nutlin-3. * p<0.05; **** p<0.0001; n.s.,

not significant (Unpaired t test). D. Quantitative real-time PCR of Cdkn1a for

liver and brain tissues from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice

treated with vehicle or Nutlin-3. V, Vehicle; N, Nutlin-3. *p<0.05; **p<0.01; n.s.,

not significant (Unpaired t test). E Representative liver and cerebrum IHC

staining for γ-H2AX and active caspase-3 with quantification at indicated time

points from wild type and Atg7Δ/Δ mice treated with vehicle or Nutlin-3. V,

Vehicle; N, Nutlin-3. * p<0.05; ***p<0.001; ****p<0.0001; n.s., not significant

(Unpaired t test). See also Figure S3.3, S3.4

Page 67: @2020 Yang Yang ALL RIGHTS RESERVED

52

Atg7 deficiency is synthetically lethal in the absence of Nrf2

Autophagy can reduce ROS by removing damaged mitochondria and unfolded

protein, and autophagy deficiency leads to increased ROS and accumulation of

unfolded protein (Manjithaya et al., 2010; Mizushima, 2010). Since we found

induction of oxidative stress markers in Atg7Δ/Δ mice (Fig. 3.2E), we

investigated whether the increased oxidative stress was responsible for p53

activation and/or tissue damage. NRF2 is the master regulator of the

antioxidant defense and is ubiquitinated by an E3 ubiquitin ligase Kelch-like

ECH-associated protein 1 (KEAP1) and degraded by the proteosome pathway

under normal conditions (Kensler et al., 2007). With increased ROS, NRF2 is

released from KEAP1 and triggers expression of a series of antioxidant genes,

and NRF2 is induced by autophagy deficiency (Lau et al., 2010; Levonen et al.,

2014). To examine the role of antioxidant defense in mice lacking autophagy,

mice with constitutive deficiency in Nrf2 (Chan et al., 1996) were crossed with

Ubc-CreERT2/+; Atg7flox/flox mice to generate Nrf2-/-; Ubc-CreERT2/+; Atg7flox/flox

mice (Fig. 3.4A). TAM administration was then used to delete Atg7 in the

presence and absence of Nrf2.

In contrast to Atg7Δ/Δ mice that survive for two to three months, Nrf2-/-Atg7Δ/Δ

mice had a lifespan of less than seven days (Fig. 3.4B). Histological

examination of tissues by H&E surprisingly showed no damage to liver, brain,

pancreas, lung and kidney in Nrf2-/-Atg7Δ/Δ mice (Fig. S3.5A). The only tissue

Page 68: @2020 Yang Yang ALL RIGHTS RESERVED

53

with significant damage was the intestine (duodenum, jejunum and ileum),

which may be the cause of increased muscle wasting and loss of WAT (Fig.

3.4C, Fig. S3.5B). Bodipy C11 staining of the whole small intestine was

significantly increased in Nrf2-/-Atg7Δ/Δ intestine, indicating increased lipid

peroxidation that results from ROS (Fig. 3.4D). IHC for active caspase-3

displayed increased staining in Nrf2-/-Atg7Δ/Δ intestine in both the crypt and

villus, indicating increased apoptosis (Fig. S3.5C-S3.5E). Thus, conditional

deletion of the essential autophagy gene Atg7 in adult mice is synthetically

lethal in the absence of Nrf2 due to damage to the intestine.

We then investigated which cell type in the intestine was most affected by

deficiency in Atg7 in the absence of Nrf2. Alcian blue staining of paraffin

sections from intestine tissues was significantly decreased in the Nrf2-/-Atg7Δ/Δ

mouse intestine, suggesting loss of goblet cells (Fig. 3.4E). IHC for the stem

cell marker OLFM4 revealed loss of OLFM4 staining in Nrf2-/-Atg7Δ/Δ but not in

wild type, Atg7Δ/Δ and Nrf2-/- mouse intestine, suggesting loss of stem cells (Fig.

3.4F). IHC for the Paneth cell marker Lysozyme revealed diffuse staining in the

Nrf2-/-Atg7Δ/Δ mouse intestine in comparison to wild type, Atg7Δ/Δ and Nrf2-/-

intestine, which suggests that Paneth cell function is abnormal (Supplementary

Fig. S3.5F) (Cadwell et al., 2008). We then investigated whether this

deleterious phenotype in the intestine was induced by p53 activation. p53flox/flox

mice were crossed with Nrf2-/-; Ubc-CreERT2/+ mice and Nrf2-/-; Ubc-CreERT2/+;

Atg7flox/flox mice to generate Nrf2-/-; Ubc-CreERT2/+; p53flox/flox mice and Nrf2-/-;

Page 69: @2020 Yang Yang ALL RIGHTS RESERVED

54

Ubc-CreERT2/+; p53flox/flox; Atg7flox/flox mice. After TAM administration, we found

that Nrf2-/-p53Δ/ΔAtg7Δ/Δ mice did not survive longer than Nrf2-/-Atg7Δ/Δ mice,

suggesting that the intestinal damage in the Nrf2-/-Atg7Δ/Δ mice was not caused

by p53 (Supplementary Fig. S3.5G).

Page 70: @2020 Yang Yang ALL RIGHTS RESERVED

55

Page 71: @2020 Yang Yang ALL RIGHTS RESERVED

56

Fig. 3.4 Atg7 deficiency is synthetically lethal in the absence of Nrf2. A.

Experimental design for generation of Atg7Δ/Δ mice, Nrf2-/- mice, and

Nrf2-/-Atg7Δ/Δ mice. B. Kaplan–Meier survival curve of wild type, Atg7Δ/Δ, Nrf2-/-,

and Nrf2-/-Atg7Δ/Δ mice. ****p<0.0001(Log-rank (Mantel-Cox) test). C.

Representative histology of duodenum, jejunum and ileum by H&E at indicated

time points from wild type, Atg7Δ/Δ, Nrf2-/-, and Nrf2-/-Atg7Δ/Δ mice. D.

Representative Bodipy C11 stain of duodenum, jejunum and ileum at indicated

time points from wild type, Atg7Δ/Δ, Nrf2-/-, and Nrf2-/-Atg7Δ/Δ mice. E.

Representative Alcian blue stain of duodenum, jejunum and ileum at indicated

time points from wild type, Atg7Δ/Δ, Nrf2-/-, and Nrf2-/-Atg7Δ/Δ mice. F.

Representative duodenum, jejunum and ileum IHC stain of OLFM4 at indicated

time points from wild type, Atg7Δ/Δ, Nrf2-/-, and Nrf2-/-Atg7Δ/Δ mice. See also

Figure S3.5.

Page 72: @2020 Yang Yang ALL RIGHTS RESERVED

57

Figure 5. Mechanism by which autophagy interacts with the p53 and

NRF2 stress response mechanisms to protect tissues in a tissue specific

manner

See text for explanation.

Page 73: @2020 Yang Yang ALL RIGHTS RESERVED

58

Discussion

Both autophagy and p53 can protect tissues from stress such as DNA damage,

oxidative stress, and hypoxia (Fischer, 2017; Mizushima and Komatsu, 2011),

and their overlapping functions have suggested that these two pathways

interact. p53 upregulates the expression of essential autophagy genes and

autophagy function in vitro (Crighton et al., 2006; Feng et al., 2007;

Kenzelmann Broz et al., 2013; Mah et al., 2012; Zhang et al., 2009). In turn,

autophagy inhibits p53 in some tumors providing a negative feedback loop

(Guo et al., 2013; Rosenfeldt et al., 2013; Strohecker et al., 2013; Yang et al.,

2014). Whether autophagy can regulate p53 in normal tissues in vivo, however,

was not clear. We found that autophagy suppresses p53 activation in liver and

brain, without which hyperactivation of p53 is responsible for damage to these

tissues. Thus, autophagy is a tissue-specific negative regulator of p53 and

contributes to a negative feedback loop to limit p53 activation in vivo (Fig.

3.5A). Remarkably, eliminating p53 also rescued the survival of Atg7-deficient

mice during fasting, suggesting that autophagy restricts p53 activation in

response to exogenous as well as endogenous stress (Fig. 3.5). Even when

p53 activation is forced by Nutlin-3, autophagy prevents these tissues from

p53-mediated damage (Fig. 3.5A, B). These findings also suggest that

autophagy may limit the effectiveness of MDM2 antagonists, and this should be

tested in the cancer setting.

Page 74: @2020 Yang Yang ALL RIGHTS RESERVED

59

The NRF2 and autophagy pathways both contribute to antioxidant defense.

NRF2 is activated by autophagy deficiency in vitro and in tumors (Lau et al.,

2010; Saito et al., 2016; Strohecker et al., 2013). The autophagy substrate p62,

which accumulates when autophagy is blocked, interacts with KEAP1, thereby

releasing and stabilizing NRF2 and promoting expression of its target genes

(Ichimura et al., 2013; Levonen et al., 2014). We found that the protective

function of NRF2 is essential for the survival of mice with loss of ATG7, as

Nrf2-/-; Atg7Δ/Δ mice die rapidly, specifically from damage to the small intestine.

Atg5 deficiency in intestine epithelia causes decreased numbers of intestinal

stem cells, and these stem cells have a higher ROS level compared to wild type

mice, which can be rescued by treating mice with antioxidant N-Acetyl Cysteine

(Asano et al., 2017). Atg16L1 is also required to protect the intestinal epithelium

from necroptosis induction in response to virus-induced intestinal bowel

disease by maintaining mitochondrial homeostasis (Matsuzawa-Ishimoto et al.,

2017). We report here that knockout of NRF2 is synthetically lethal with loss of

Atg7, as NRF2 is specifically required to protect the survival of intestinal stem

cells (Fig. 3.5C). The compensatory protective effect of NRF2 to loss of

autophagy may be broad as recent cell-based screens identified NRF2

activation as a resistance mechanism selected for in cancer cells deleted for

essential autophagy genes (Towers et al., 2019). In conclusion, autophagy

limits p53 activation and damage in liver and brain, while NRF2 limits intestinal

stem cells damage due to loss of autophagy by a p53-independent mechanism.

Page 75: @2020 Yang Yang ALL RIGHTS RESERVED

60

These findings demonstrate the functional interaction and tissue specificity of

these stress regulated pathways (Fig. 3.5A-C).

Page 76: @2020 Yang Yang ALL RIGHTS RESERVED

61

Supplementary Figures

Page 77: @2020 Yang Yang ALL RIGHTS RESERVED

62

Figure S3.1. Related to Figure 3.1. A. Quantitative real-time PCR of Atg7

and p53 for liver, brain, kidney, pancreas, muscle, lung at indicated time points

from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice. 2w, 2 weeks time point.

5w, 5 weeks time point. 8w, 8 weeks time point. B-C. Representative histology

of liver, muscle, cerebrum, cerebellum, pancreas, WAT and lung by H&E at 2

weeks and 5 weeks time point from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ

mice. D. Representative histology of kidney by H&E at indicated time points

from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice. E. Blood glucose level

of wild type, Atg7Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice at the end of starvation. *p<0.05;

**p<0.01(Unpaired t test)

Page 78: @2020 Yang Yang ALL RIGHTS RESERVED

63

Page 79: @2020 Yang Yang ALL RIGHTS RESERVED

64

Figure S3.2. Related to Figure 3.2. A. Representative liver IHC staining of p53

at indicated time points from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice.

1-4, corresponding to images in Fig. 3.2A. B. Representative cerebrum IHC

staining of p53 at indicated time points from wild type, Atg7Δ/Δ, p53Δ/Δ, and

Atg7Δ/Δp53Δ/Δ mice. 1-4, corresponding to images in Fig. 3.2A. C.

Representative liver IHC staining for γ-H2AX at indicated time points from wild

type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice. 1-4, corresponding to images in

Fig. 3.2D. D. Representative cerebrum IHC staining for γ-H2AX at indicated

time points from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice. 1-4,

corresponding to images in Fig. 3.2D. E. Representative liver IHC staining for

active caspase-3 at indicated time points from wild type, Atg7Δ/Δ, p53Δ/Δ, and

Atg7Δ/Δp53Δ/Δ mice. 1-4, corresponding to images in Fig. 3.2D. F.

Representative cerebrum IHC staining for active caspase-3 at indicated time

points from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice. 1-4,

corresponding to images in Fig. 3.2D.

Page 80: @2020 Yang Yang ALL RIGHTS RESERVED

65

Page 81: @2020 Yang Yang ALL RIGHTS RESERVED

66

Figure S3.3. Related to Figure 3.3. A. Quantitative real-time PCR of Atg7 and

p53 for liver and brain from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice

treated with vehicle or Nutlin-3. B. Representative histology of liver and

cerebrum by H&E from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice

treated with vehicle or Nutlin-3. C. Representative liver and cerebrum IHC

staining for p53 from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice treated

with vehicle or Nutlin-3. 1-4, corresponding to images in Fig. 3.3C. D.

Representative liver and cerebrum IHC staining for γ-H2AX and quantification

from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice treated with vehicle or

Nutlin-3. 1-4, corresponding to images in Fig. 3.3E. E. Representative liver and

cerebrum IHC staining for active caspase-3 and quantification at indicated time

points from wild type, Atg7Δ/Δ, p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice treated with

vehicle or Nutlin-3. 1-4, corresponding to images in Fig. 3.3E.

Page 82: @2020 Yang Yang ALL RIGHTS RESERVED

67

Figure S3.4. Related to Figure 3.3. Quantitative real-time PCR of Uvrag, Ulk1,

Ulk2, Vmp1, Atg2b, Atg4a, Atg4c and Atg10 for liver from wild type, Atg7Δ/Δ,

p53Δ/Δ, and Atg7Δ/Δp53Δ/Δ mice treated with vehicle or Nutlin-3. V, treated with

Vehicle; N, treated with Nutlin-3.

Page 83: @2020 Yang Yang ALL RIGHTS RESERVED

68

Page 84: @2020 Yang Yang ALL RIGHTS RESERVED

69

Figure S3.5. Related to Figure 3.4. A. Representative histology of liver,

cerebrum, cerebellum, pancreas, lung and kidney by H&E at 2 days and 4 days

time point from wild type, Atg7Δ/Δ, Nrf2-/-, and Nrf2-/-Atg7Δ/Δ mice. B.

Representative histology of muscle and WAT by H&E at 2 days and 4 days time

point from wild type, Atg7Δ/Δ, Nrf2-/-, and Nrf2-/-Atg7Δ/Δ mice. C-E.

Representative duodenum, jejunum and ileum IHC stain of active caspase-3 at

indicated time points from wild type, Atg7Δ/Δ, Nrf2-/-, and Nrf2-/-Atg7Δ/Δ mice. F.

Representative duodenum, jejunum and ileum IHC stain of Lysozyme at

indicated time points from wild type mice, Atg7Δ/Δ mice, Nrf2-/- mice, and

Nrf2-/-Atg7Δ/Δ mice. G. Kaplan–Meier survival curve of wild type, Atg7Δ/Δ, Nrf2-/-,

and Nrf2-/-Atg7Δ/Δ mice, Nrf2-/-p53Δ/Δ and Nrf2-/-p53Δ/ΔAtg7Δ/Δ mice.

****p<0.0001. n.s., not significant. (Log-rank (Mantel-Cox) test).

Page 85: @2020 Yang Yang ALL RIGHTS RESERVED

70

CHAPTER 4

AUTOPHAGY IS ESSENTIAL FOR TELOCYTE SURVIVAL, WNT

SIGNALING AND INTESTINAL STEM CELL HOMEOSTASIS

Page 86: @2020 Yang Yang ALL RIGHTS RESERVED

71

Introduction

Autophagosome formation requires three steps: initiation, nucleation and

elongation. During the elongation stage, there are two protein complexes

involved: the ATG12-ATG5-ATG16 complex (formed with the help of ATG7

and ATG10) and the LC3 complex (formed with the help of ATG4, ATG7 and

ATG3). The ATG12-ATG5-ATG16 complex is recruited to the autophagosome

membrane, and facilitates the lipidation of LC3 with (PE), which is required for

the elongation and final formation of autophagosomes. Therefore, both Atg5

and Atg7 are essential for the formation of functional autophagosomes and the

process of autophagy.

Conditional, whole-body deletion of essential autophagy gene Atg7 in adult

mice (Atg7Δ/Δ) leads to liver damage, muscle wasting, and loss of adipose

tissue with lifespan decreased to three months due primarily to

neurodegeneration (Karsli-Uzunbas et al., 2014). Constitutively Atg5- or

Atg7-deficient mice are born normal but unable to survive neonatal starvation

when the trans-placental nutrient supply is interrupted but not yet restored by

milk (Komatsu et al., 2005; Kuma et al., 2004). Consistent with the role of

autophagy in promoting survival in starvation, fasting Atg7Δ/Δ but not wild type

mice is lethal (Karsli-Uzunbas et al., 2014). Fasted Atg7Δ/Δ mice rapidly deplete

dedicated nutrient stores of glycogen and lipids and fail to maintain circulating

glucose levels and die from hypoglycemia (Karsli-Uzunbas et al., 2014).

Page 87: @2020 Yang Yang ALL RIGHTS RESERVED

72

Since both Atg7 and Atg5 are essential autophagy genes, we generated the

genetically engineered mouse model (GEMM) for conditional, whole-body

Atg5 deletion to address whether loss of Atg5 would have similar phenotype as

loss of Atg7. In contrast to Atg7Δ/Δ mice which tolerate loss of ATG7 for 2-3

months in the fed state, we found that adult mice with conditional whole-body

deletion of Atg5 (Atg5Δ/Δ) survive only four days post deletion. Atg5Δ/Δ mice

display severe damage in the ileum part of intestine with loss of barrier function.

In contrast to Atg7Δ/Δ mice, Atg5Δ/Δ mice showed more rapid loss of autophagy

in the ileum with loss of stem cells and also PDGFRα+ mesenchymal cells

(telocytes), which provide Wnt ligands to the stem cells for their maintenance.

However, a gradual deletion of Atg5 rescued this phenotype and these mice

generated tissue damage and neurodegenerative phenotype similar as whole

body loss of Atg7 and Atg12. Loss of telocytes in the Atg5Δ/Δ ileum caused

significantly decreased aspartate and increased nucleotide degradation

products. Therefore, Atg5 is required in maintenance of PDGFRα+

mesenchymal cells which contribute Wnt ligands to the intestinal stem cells for

maintenance of intestinal homeostasis.

Page 88: @2020 Yang Yang ALL RIGHTS RESERVED

73

Results

Conditional, whole-body Atg5 deletion in adult mice leads to ileum

damage and death

As both Atg7 and Atg5 function at the same step in the autophagy pathway, we

hypothesized that conditional whole-body deletion of either gene will behave

similarly. To address this, we generated the inducible Ubc-CreERT2/+; Atg5flox/flox

GEMM, treated these adult mice (eight weeks of age) with 5 consecutive daily

injections of tamoxifen (TAM) as we described previously to generate Atg7Δ/Δ

mice (Karsli-Uzunbas et al., 2014) to generate Atg5Δ/Δ mice (Fig. 4.1A).To our

surprise, Atg5Δ/Δ mice showed a significantly decreased lifespan of only about

four days post TAM, while Atg7Δ/Δ mice have a lifespan of about two to three

months (Karsli-Uzunbas et al., 2014) (Fig. 4.1B). In order to confirm the

phenotype is specifically caused by loss of Atg5, we did the TAM deletion in

Cag-CreERT2/+ Atg12flox/flox and Cag-CreERT2/+ Atg5flox/flox mice, which is another

inducible whole body deletion model (Fig. S4.1A), and Cag-CreERT2/+

Atg12flox/flox mice was previously published to have similar lifespan and

neurodegeneration phenotype as the whole body Atg7Δ/Δ mice (Malhotra et al.,

2015). We observed similar lifespan of 4 days for the Atg5Δ/Δ mice, while

Atg12Δ/Δ survived as long as 10 weeks post TAM, which is similar as Atg7Δ/Δ

mice (Fig. S4.1B). To determine why Atg5Δ/Δ mice fail to survive immediately

post deletion, we compared the histology of wild type, Atg7Δ/Δ and Atg5Δ/Δ

mouse tissues, and the primary damage site is the ileum part of intestine. The

Page 89: @2020 Yang Yang ALL RIGHTS RESERVED

74

wild type ileum showed normal villous architecture without inflammation or

significant apoptosis. The Atg7Δ/Δ ileum maintained the normal villous

architecture, but with slightly increased apoptosis in the glandular epithelium.

However, Atg5Δ/Δ ileum demonstrated marked epithelial damage exemplified

by the shortening of villi, cytoplasmic vacuolation, numerous apoptosis, and

sloughing of the surface epithelium especially at Day three post TAM but no

significant inflammatory cell infiltration. Duodenum and jejunum didn’t display

those damage phenotypes (Fig. 4.1C, S4.1C, S4.1D). Similar damage is

observed in Cag-Cre Atg5Δ/Δ ileum but not the duodenum or jejunum (Fig.

S4.1E-G).

Page 90: @2020 Yang Yang ALL RIGHTS RESERVED

75

Page 91: @2020 Yang Yang ALL RIGHTS RESERVED

76

Figure. 4.1. Conditional, whole-body Atg5 deletion in adult mice leads to

ileum damage and death. A. Experimental design for generation of Atg5Δ/Δ

mice and Atg7Δ/Δ mice. Ubc-CreERT2/+, Ubc-CreERT2/+; Atg5flox/flox mice and

Ubc-CreERT2/+; Atg7flox/flox mice were treated with TAM at 8-10 weeks of age

with 5 shots of TAM every consecutive day and analyzed. B. Kaplan-Meier

survival curve of TAM-treated wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. ****

p<0.0001 (log-rank Mantel-Cox test). C. Representative ileum

Hematoxyin&Eosin stained (H&E) histology at indicated time points from wild

type, Atg7Δ/Δ and Atg5Δ/Δ mice. D. Blood glucose concentration measured by

mg/dL from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice at indicated time points.

*p<0.05, ***p<0.001(log-rank Mantel-Cox test) E. FITC-dextran concentration

in the blood serum from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice at indicated time

points. *p<0.05, **p<0.01, ***p<0.001(log-rank Mantel-Cox test) F. Western

blotting for ATG5, ATG7, LC3 at indicated time points from the ileum tissue of

wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. β-ACTIN is used as the loading control.

See also Figure S4.1, Figure S4.2.

Page 92: @2020 Yang Yang ALL RIGHTS RESERVED

77

Acute whole body Atg5 deletion decreases blood glucose and causes

loss of intestinal barrier function

As the ileum functions in dietary nutrient absorption, and nutritional deprivation

of Atg7Δ/Δ mice causes lethal hypoglycemia, we sought to test if Atg5Δ/Δ mice

were nutritionally compromised due to loss of ileum function. Atg5Δ/Δ mice had

decreased blood glucose levels starting from day two post TAM which further

decreased on day three in comparison to wild type and Atg7Δ/Δ mice (Fig. 4.1D).

Supplementation of glucose by intraperitoneal (IP) injection restored the blood

glucose levels but did not rescue the survival of Atg5Δ/Δ mice, suggesting that

this was not solely responsible for death (Fig. S4.2A, S4.2B). Intestinal barrier

function is important in defending from bacterial invasion and required for

mouse survival. To test the barrier function of Atg5Δ/Δ mice, we performed

FITC-dextran dietary supplementation to wild type, Atg7Δ/Δ and Atg5Δ/Δ mice

and compared the concentration of FITC-dextran in the circulating blood

supply. The FITC-dextran concentration in serum was low with no difference at

day one post TAM in wild type control, Atg7Δ/Δ mice and Atg5Δ/Δ mice. However,

Atg5Δ/Δ mice began to show increased FITC-dextran in the circulation at day

two post TAM, which further increased at day three in comparison to wild type

and Atg7Δ/Δ mice (Fig. 4.1E). These results suggest that Atg5Δ/Δ mice lost

intestinal barrier function and likely also efficient nutritional absorption causing

hypoglycemia, which led to death of Atg5Δ/Δ mice.

Page 93: @2020 Yang Yang ALL RIGHTS RESERVED

78

Deletion of Atg5 causes more rapid loss of autophagy in the Ileum than

deletion of Atg7

Deletion of Atg5 in Atg5Δ/Δ mice and deletion of Atg7 in Atg7Δ/Δ mice were

detected, but Atg5 deletion occurred more rapidly and thoroughly compared to

Atg7 (Fig. S4.2C, S4.2D). Loss of ATG5, and more importantly the

ATG5-ATG12 complex, was apparent in the ileum on day one post TAM in

Atg5Δ/Δ mice, whereas the ATG5-ATG12 complex persisted in Atg7Δ/Δ mice

despite the loss of ATG7 (Fig. 4.1F). This indicates that ATG5-ATG12 complex

may be stable and functional longer in the absence of ATG7 but not ATG5. In

contrast, accumulation of the unprocessed form of LC3 (LC3-I) and decreased

levels of the processed form of LC3 (LC3-II) occurred earlier in the Atg7Δ/Δ

ileum compared to the Atg5Δ/Δ ileum, consistent with the direct role of ATG7 in

conjugation of PE to LC3 to generate LC3-II (Fig. 4.1F). Similar results were

observed in duodenum and jejunum (Fig. S4.2E, S4.2F). These results

suggest that whole body deletion of Atg5 and Atg7 can both cause autophagy

defect in the intestine, but Atg5Δ/Δ mice lose autophagy more rapidly than

Atg7Δ/Δ mice due to more rapid and complete loss of the ATG5-ATG12

complex, which may explain the lethal phenotype of Atg5Δ/Δ mice.

Page 94: @2020 Yang Yang ALL RIGHTS RESERVED

79

Ileum damage in Atg5Δ/Δ mice is caused by loss of stem cells

The intestinal epithelium is mainly divided into two compartments: crypt and

villus. In the crypt, the crypt base columnar (CBC) cells, or stem cells, are

interspersed with Paneth cells at the base region of the crypt. These CBC cells

are responsible for the renewal of whole intestine epithelium by continuously

generating the proliferating transient amplifying (TA) cells. These TA cells

migrate ascendantly towards the base of villi and differentiate into several cell

types in villus, mainly enterocytes and goblet cells (Barker, 2014). In order to

investigate the cause of ileum damage, we checked which cell type in the

ileum was lost in Atg5Δ/Δ mice. Immunohistochemistry (IHC) of ATG5 and

ATG7 protein confirmed the loss of ATG5 and ATG7 protein in the ileum and

also duodenum and jejunum in Atg5Δ/Δ and Atg7Δ/Δ mice (Fig. 4.2A, Fig.

S4.3A-S4.3D). However, the intestine of Atg5Δ/Δ mice accumulated more p62

aggregates in comparison to Atg7Δ/Δ mice indicative of a more rapid and

efficient loss of autophagy (Fig. 4.2B, Fig. S4.3E, S4.3F). There was more p62

accumulation in Cag-Cre Atg5Δ/Δ intestine compared to Atg5Δ/Δ intestine too

(Fig. S4.3G-L). Alcian blue staining of the small intestine was decreased in the

ileum of Atg5Δ/Δ mice from day two post TAM and was completely lost on day

three, indicating loss of goblet cells in the ileum but not in the duodenum and

jejunum of Atg5Δ/Δ mice, while wild type and Atg7Δ/Δ mice maintained the goblet

cells throughout the intestine (Fig. 4.2C, Fig. S4.4A, S4.4B). Loss of goblet

cells was also observed in Cag-Cre Atg5Δ/Δ ileum but not duodenum or

Page 95: @2020 Yang Yang ALL RIGHTS RESERVED

80

jejunum (Fig. S4.4C-E). IHC for the stem cell marker OLFM4 revealed loss of

OLFM4 staining starting from day two, which was completely lost by day three

in Atg5Δ/Δ ileum compared to wild type and Atg7Δ/Δ mice (Fig. 4.2D, Fig. S4.4F,

S4.4G). Similar phenotype was observed in Cag-Cre Atg5Δ/Δ ileum but not

duodenum or jejunum (Fig. S4.4H-J). Thus, stem cells are lost in the ileum but

not in the duodenum and jejunum specifically with systemic deletion of Atg5.

IHC for lysozyme showed that staining is maintained in duodenum and

jejunum in wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice (Fig. S4.4K, S4.4L).

However, Atg5Δ/Δ mice displayed a diffuse staining pattern in the ileum at day

three in Atg5Δ/Δ mice (Fig. 4.2E). This diffuse stain pattern is also displayed in

the Cag-Cre Atg5Δ/Δ ileum but not duodenum or jejunum (Fig. S4.4M-S4.4O).

Immunofluorescence (IF) for OLFM4 and p62 showed that there was

increased p62 aggregates colocalized with OLFM4 in Atg5Δ/Δ ileum in

comparison to Atg7Δ/Δ ileum at day two but not at day three because of the

complete loss of stem cells (Fig. 4.2F). There were increased p62 aggregates

in Atg5Δ/Δ duodenum and Atg5Δ/Δ jejunum compared to Atg7Δ/Δ at all time points

but not as much as in the ileum (Fig. S4.4P, S4.4Q). Taken together, the ileum

of Atg5Δ/Δ mice lost intestinal stem cells and goblet cells and had abnormal

Paneth cells, and the autophagy blockage was more severe in the stem cells

of Atg5Δ/Δ mice compared to Atg7Δ/Δ mice. These findings suggest that ileum

stem cells are acutely sensitive to loss of autophagy that may be more severe

upon systemic deletion of Atg5 than Atg7 in mice.

Page 96: @2020 Yang Yang ALL RIGHTS RESERVED

81

Page 97: @2020 Yang Yang ALL RIGHTS RESERVED

82

Figure 4.2. Ileum damage in Atg5Δ/Δ mice is caused by loss of stem cells.

A. Representative ileum Immunohistochemistry (IHC) staining of ATG5 and

ATG7 at indicated time points from wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice. B.

Representative ileum IHC staining of p62 at indicated time points from wild

type, Atg7Δ/Δ and Atg5Δ/Δ mice. Red arrows indicate p62 aggregates. C.

Representative ileum Alcian blue staining at indicated time points from wild

type, Atg7Δ/Δ and Atg5Δ/Δ mice. D. Representative ileum IHC staining of OLFM4

at indicated time points from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. E.

Representative ileum IHC staining of Lysozyme at indicated time points from

wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. F. Representative ileum

Immunofluorescence (IF) costaining of p62 and OLFM4 at indicated time

points from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. Red arrows indicate the p62

aggregates which colocalize with OLFM4. See also Figure S4.3, Figure S4.4.

Page 98: @2020 Yang Yang ALL RIGHTS RESERVED

83

In vitro deletion of Atg5 lead to growth inhibition and can be rescued by

supplementation of Wnt ligands

Since intestinal stem cells are required for the renewal of the other cell types in

the intestinal epithelium, we hypothesized that loss of stem cell was the main

cause of ileum damage and loss of barrier function. The most important

signaling pathway that supports the growth and renewal of the intestinal stem

cells is the canonical WNT signaling pathway (Clevers et al., 2014; Fevr et al.,

2007). WNT signaling is activated by Wnt ligands Wnt3a or Wnt2b in the crypt

region, and β-catenin is the essential signal transducer. To test if conditional

deficiency in ATG5 caused loss of ileum stem cells due to failure of WNT

signaling, we isolated crypts from the ileum of Atg5flox/flox and Atg7flox/flox mice

and plated them in matrigel to culture organoids. Organoids were then treated

with TAM to delete either Atg5 or Atg7 genes in vitro (Fig. 4.3A). Atg5Δ/Δ

organoids showed rapid and complete loss of the ATG5-ATG12 complex, while

Atg7Δ/Δ organoids showed loss of ATG7 protein yet maintained some

ATG5-ATG12 complex (Fig. 4.3B). Both Atg5Δ/Δ and Atg7Δ/Δ organoids

accumulated LC3-I and had less LC3-II post TAM (Fig. 4.3B). These findings

were consistent with ATG5 loss in ileum organoids causing a more rapid and

complete loss of the ATG5-ATG12 complex than loss of ATG7, as was

observed in the ileum in vivo. Thus, loss of ATG5 may thereby cause a more

profound loss of autophagy function.

Page 99: @2020 Yang Yang ALL RIGHTS RESERVED

84

To address how loss of ATG5 and ATG7 altered stem cell function, organoids

were followed over time. After deletion, the viability and budding number of

Atg5Δ/Δ organoids was decreased compared to Atg7Δ/Δ organoids when

passaged for three generations, and this phenotype was rescued by

supplementation with Wnt3a or Wnt2b (Fig. 4.3C, 4.3D, S4.5A-C, H-J). IF of

organoids for OLFM4 and p62 showed significant loss of OLFM4-positive stem

cells with more p62 aggregates in Atg5Δ/Δ compared with Atg7Δ/Δ organoids

(Fig. 4.3E). Real-time PCR for stem cell maker OLFM4, Lgr5 and Wnt target

gene Axin2 and Sox9 revealed decreased transcription level of these genes in

Atg5Δ/Δ compared with Atg5Flox/Flox organoids but can be brought back to similar

or higher level when supplemented with Wnt3a or Wnt2b (Fig. 4.3F, Fig.

S4.5D-G, Fig. S4.5K-N). WB for organoids at passage two and three revealed

that ATG7 protein were restored with low LC3-I accumulation and increased

LC3-II in Atg7Δ/Δ organoids, whereas in Atg5Δ/Δ organoids ATG5 was still lost

with high LC3-I accumulation and no LC3-II, suggesting that the Atg7Δ/Δ

organoids were outcompeted by undeleted Atg7Flox/Flox organoids and had

intact autophagy function (Fig. S4.5O, S4.5P). These data indicated that stem

cell depletion in Atg5Δ/Δ organoids is caused by insufficient Wnt signaling.

Page 100: @2020 Yang Yang ALL RIGHTS RESERVED

85

Figure 4.3. In vitro deletion of Atg5 lead to growth inhibition and can be

rescued by supplementation of Wnt ligands. A. PCR of Atg5 from the

untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids and Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b; And PCR of Atg7 from untreated

Atg7flox/flox organoids, Atg7Δ/Δ organoids and Atg7Δ/Δ organoids supplemented

with Wnt3a or Wnt2b. B. Western blotting for ATG5, ATG7, and LC3 from the

untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, and Atg5Δ/Δ organoids

Page 101: @2020 Yang Yang ALL RIGHTS RESERVED

86

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 1. β-ACTIN is used as the loading control. C. Representative picture

of the untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, and Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 3. D. Alive organoid percentage and average budding numbers of the

untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 3. E. Representative organoid IF costaining of p62 and OLFM4 of the

untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 3. F. Quantitative real-time PCR of OLFM4, Lgr5, Axin2 and Sox9 of

the untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 3. See also Figure S4.5.

Page 102: @2020 Yang Yang ALL RIGHTS RESERVED

87

Atg5 is required to maintain PDGFRα+ mesenchymal cells.

Next, we tested whether ATG5-dependent WNT signaling is required for

maintenance of stem cells in vivo. We observed significant loss of active

β-catenin in the ileum crypt in Atg5Δ/Δ mice by IHC, and decreased

transcription level of Axin2 and Sox9 in Atg5Δ/Δ ileum compared with Atg7Δ/Δ

ileum at Day three post TAM, suggesting failure of Wnt signaling at Day three

but not Day one and Day two (Fig. 4.4A, 4.4B, S4.6A, S4.6B). In Cag-Cre

Atg5Δ/Δ ileum same phenotype was observed (Fig. S4.6C, S4.6D). Wnt ligands

can come from Paneth cells in the epithelium (Farin et al., 2012), however,

Atg5 deletion using a epithelium-specific villin-Cre did not cause loss of ileum

stem cells(Cadwell et al., 2008), suggesting that ATG5-dependent Wnts were

originating outside the epithelium. Recent data demonstrated that PDGFRα+

mesenchymal cells or telocytes surrounding the intestinal epithelium are a

critical source of Wnt ligands essential for intestinal stem cell maintenance

(Greicius et al., 2018; Shoshkes-Carmel et al., 2018; Stzepourginski et al.,

2017). Therefore, we tested whether ATG5 was required for the function and

survival of PDGFRα+ mesenchymal cells, the loss of which in Atg5Δ/Δ ileum

was responsible for ileum stem cell loss in vivo. IF for PDGFRα and CD34

indicated that PDGFRα and CD34 positive cells were present at day one and

two post TAM in the Atg5Δ/Δ and Atg7Δ/Δ ileum (Fig. S4.6E, S4.6F). However,

there was nearly complete loss of PDGFRα and CD34 positive cells on day

three post TAM specifically in the Atg5Δ/Δ ileum but not in the in Atg7Δ/Δ ileum

Page 103: @2020 Yang Yang ALL RIGHTS RESERVED

88

(Fig. 4.4C). In Cag-Cre Atg5Δ/Δ ileum, PDGFRα and CD34 positive cells were

present at day one, and almost complete loss of these cells on day two but not

in the Atg12Δ/Δ ileum (Fig. S4.6G, S4.6H). Thus, deletion of Atg5 caused loss

of telocytes in Atg5Δ/Δ but not Atg7Δ/Δ or Atg12Δ/Δ ileum. We also observed

increased p62 in telocytes on day one and two in Atg5Δ/Δ compared to Atg7Δ/Δ

ileum, whereas p62 staining was lost on day three in Atg5Δ/Δ ileum due to loss

of telocytes (Fig. S4.7A, 4.4C, S4.7B). Similar phenotype of p62 accumulation

was also displayed in Cag-Cre Atg5Δ/Δ ileum on day one but not as much in

Atg12Δ/Δ ileum (Fig. S4.7C, S4.7D). Greater p62 accumulation in Atg5Δ/Δ ileum

is constant with more rapid and complete loss of autophagy function. We

then tested whether Wnt ligand supplementation could compensate for the

loss of PDGFRα+ mesenchymal cells in the Atg5Δ/Δ ileum. Atg5Δ/Δ mice

plemented with Wnt ligands Wnt3a or Wnt2b showed a twenty four hours

extension of their lifespan (Fig. 4.4D). In addition, there was an increase of

OLFM4 IHC signal in the ileum of Atg5Δ/Δ mice supplemented with Wnt3a and

Wnt2b compared to untreated Atg5Δ/Δ mice, but not to the extent as seen in

wild type mice (Fig. 4.4E). These data indicated that PDGFRα+ mesenchymal

cells require autophagy to survive and are essential in maintaining intestinal

stem cells by providing them with Wnt ligands.

Page 104: @2020 Yang Yang ALL RIGHTS RESERVED

89

Page 105: @2020 Yang Yang ALL RIGHTS RESERVED

90

Figure 4.4. Atg5 is required to maintain PDGFRα+ mesenchymal cells. A.

Representative ileum IHC staining of Active β-catenin at Day 3 post TAM from

wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. B. Representative ileum IF costaining of

CD34 and PDGFRα at Day 3 post TAM from wild type, Atg7Δ/Δ and Atg5Δ/Δ

mice. C. Representative ileum IF costaining of p62 and PDGFRα at Day 2 from

wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. White arrows indicate colocalization of p62

with PDGFRα. D. Kaplan-Meier survival curve of Atg5Δ/Δ mice, Atg5Δ/Δ mice

supplemented with Wnt3a and Atg5Δ/Δ mice supplemented with Wnt2b. *

p<0.05, **, p<0.01 (log-rank Mantel-Cox test) E. Representative ileum IHC of

OLFM4 at Day 3 on the duodenum, jejunum and ileum from Atg5Δ/Δ mice,

Atg5Δ/Δ mice supplemented with Wnt3a and Atg5Δ/Δ mice supplemented with

Wnt2b. See also Figure S4.6, S4.7.

Page 106: @2020 Yang Yang ALL RIGHTS RESERVED

91

Telocyte specific deletion of Atg5 causes similar ileum damage

phenotype.

We then sought to investigate whether the ileum damage phenotype is

specifically caused by loss of PDGFRα+ mesenchymal cells in the whole body

Atg5Δ/Δ mice. To test this, we crossed the PDGFRα-CreERT/+ mice with

Atg7flox/flox and Atg5flox/flox mice to generate the PDGFRα-CreERT/+; Atg7flox/flox

and the PDGFRα-CreERT/+; Atg5flox/flox mice and then administered with TAM to

delete Atg7 or Atg5 specifically in the PDGFRα+ mesenchymal cells (Fig. 4.5A).

Similar to whole body Atg5Δ/Δ mice, almost seventy percent of the

PDGFRα-Cre Atg5Δ/Δ mice died within six days, while wild type and

PDGFRα-Cre Atg7Δ/Δ mice survived (Fig. 4.5B). There’s moderate p62

aggregates in PDGFRα+ mesenchymal cells in both PDGFRα-Cre Atg7Δ/Δ and

Atg5Δ/Δ mice at Day two post TAM, suggesting that autophagy function was still

intact at this time point in PDGFRα+ mesenchymal cells (Fig. S4.8A). We

observed increased p62 aggregates in PDGFRα+ mesenchymal cells from

PDGFRα-Cre Atg5Δ/Δ mice at Day three, following the complete loss of these

cells at Day four, indicating blockage of autophagy caused loss of PDGFRα+

mesenchymal cells in PDGFRα-Cre Atg5Δ/Δ ileum (Fig. 4.5C, 4.5D). Significant

loss of active β-catenin and OLFM4 in the ileum crypt of PDGFRα-Cre Atg5Δ/Δ

mice by IHC at Day four were observed, suggesting loss of Wnt signaling and

stem cells(Fig. 4.5E, 4.5F). Alcian blue staining of the PDGFRα-Cre Atg5Δ/Δ

ileum was largely decreased from day three and was completely lost on day

Page 107: @2020 Yang Yang ALL RIGHTS RESERVED

92

four, indicating loss of goblet cells (Fig. S4.8B). IHC for Lysozyme revealed

that staining is maintained and no diffused pattern detected in PDGFRα-Cre

Atg5Δ/Δ ileum, suggesting that Paneth cells were functioning normally (Fig.

S4.8C). Similar to whole body Atg5Δ/Δ ileum, PDGFRα-Cre Atg5Δ/Δ ileum also

demonstrated epithelial damage exemplified by shortening of villi, cytoplasmic

vacuolation, numerous apoptosis, and sloughing of the surface epithelium at

Day four post TAM but no significant inflammatory cell infiltration, which is not

found in duodenum and jejunum (Fig. 4.5G, S4.8D, S4.8E). For the

PDGFRα-Cre Atg5Δ/Δ mice that survived longer than six days, PDGFRα+

mesenchymal cells were maintained with moderate p62 aggregates, and the

small intestine was intact, which indicated that Atg5 might be undeleted in the

PDGFRα+ mesenchymal cells of these mice (Fig. S4.8F, S4.8G). These data

indicated that the ileum damage phenotype we observed in the whole body

Atg5Δ/Δ mice was specifically caused by loss of PDGFRα+ mesenchymal cells.

Page 108: @2020 Yang Yang ALL RIGHTS RESERVED

93

Page 109: @2020 Yang Yang ALL RIGHTS RESERVED

94

Figure 4.5. Telocyte specific deletion of Atg5 causes similar ileum

damage phenotype. A. Experimental design for generation of

PDGFRα-CreERT/+ Atg5Δ/Δ and Atg7Δ/Δ mice. B Kaplan-Meier survival curve of

TAM-treated wild type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice. *

p<0.05 **** p<0.0001 (log-rank Mantel-Cox test) C and D. Representative

ileum IF costaining of p62 and PDGFRα at Day 3 and Day 4 from wild type,

PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice. White arrows indicate

colocalization of p62 with PDGFRα. E. Representative ileum H&E histology at

indicated time points from wild type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre

Atg5Δ/Δ mice. F. Representative ileum IHC staining of Active β-catenin at Day 4

post TAM from wild type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice.

G. Representative ileum IHC staining of OLFM4 at indicated time points from

wild type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice. See also

Figure S4.8.

Page 110: @2020 Yang Yang ALL RIGHTS RESERVED

95

Deleting Atg5 more gradually rescues intestinal ileum function.

Systemic deletion of Atg5 causes more rapid loss of the ATG5-ATG12

complex and greater p62 accumulation in the ileum that systemic deletion of

Atg7, suggesting a more rapid and complete loss of autophagy. This raised the

possibility that gradual loss of autophagy may allow adaptation and ileum

survival. We then tested whether giving Atg5flox/flox mice TAM once per week for

4 weeks (slow deletion) instead of on 5 consecutive days (fast deletion) to

delete Atg5 more gradually could allow adaptation and rescue the lethal

phenotype of Atg5Δ/Δ mice (Fig. 4.6A). In stark contrast to Atg5Δ/Δ mice

generated by fast deletion that died in four days, Atg5Δ/Δ mice generated by

slow deletion survived three months post TAM, more comparable to Atg7Δ/Δ

mice generated by slow (or fast) deletion (Fig. 4.1B, 4.6B). Importantly, both

Atg5Δ/Δ and Atg7Δ/Δ mice generated by slow deletion died of neurodegeneration

similar to Atg7Δ/Δ mice generated by fast deletion (Karsli-Uzunbas et al., 2014).

Tissues in Atg5Δ/Δ and Atg7Δ/Δ mice by slow deletion maintained intact at Day

three, and started to show loss of Purkinje cells in cerebellum (Fig. S4.9A,

S4.9B). At Week 5, there were more loss of Purkinje cells compared to Week 2

in Atg5Δ/Δ mice, but less severe compared to Atg7Δ/Δ mice at this time point,

along with severe loss of hepatocytes in liver, muscle wasting, loss of white

adipose tissue and massive loss of Purkinje cells (Fig. S4.9C). At Week 12,

Atg5Δ/Δ mice also developed massive loss of Purkinje cells, loss of hepatocytes,

loss of white adipose tissue and muscle wasting which were similar as Atg7Δ/Δ

Page 111: @2020 Yang Yang ALL RIGHTS RESERVED

96

mice at Week 5 (Fig. S4.9D). We observed normal villous architecture without

inflammation or significant apoptosis in Atg5Δ/Δ ileum at day three with slow

deletion, whereas fast deletion in these mice caused severe ileum damage

(Fig. 4.6C, Fig.4.1E), with no damage at duodenum and jejunum (Fig. S4.9E).

Deletion of Atg5 in Atg5Δ/Δ mice and deletion of Atg7 in Atg7Δ/Δ mice were

confirmed (Fig. S4.9F). Loss of ATG5-ATG12 complex was displayed in the

ileum on day three post TAM in Atg5Δ/Δ mice by slow deletion, whereas the

ATG5-ATG12 complex still maintained in Atg7Δ/Δ mice which was similar as

observed in whole body knockout mice generated by acute deletion (Fig. 4.6D,

4.1F). Accumulation of the unprocessed form of LC3 (LC3-I) and decreased

levels of the processed form of LC3 (LC3-II) was observed in the Atg5Δ/Δ ileum

compared to the Atg7Δ/Δ ileum, suggesting higher autophagy blockage in

Atg5Δ/Δ ileum (Fig. 4.6D). Similar results were observed in duodenum and

jejunum (Fig. S4.9G). We also observed loss of ATG5 in duodenum, jejunum

and ileum in Atg5Δ/Δ mice and loss of ATG7 in Atg7Δ/Δ mice, suggesting these

proteins are lost in intestine after TAM (Fig. 4.6E). There were increased p62

aggregates observed in duodenum, jejunum and mostly in ileum part of Atg5Δ/Δ

mice compared to Atg7Δ/Δ mice (Fig. 4.6F, S4.9H). In contrast to Atg5Δ/Δ mice

generated by fast deletion, OLFM4-positive ileum stem cells were retained in

Atg5Δ/Δ mice generated by slow deletion, as were the PDGFRα and CD34

positive telocytes (Fig. 4.6G, 4.6H). There were no decrease in the relative

transcriptional level of Axin2 and less than two fold change of Sox9,

Page 112: @2020 Yang Yang ALL RIGHTS RESERVED

97

suggesting Wnt signaling is still functioning (Fig. S4.9I). Moderate p62

accumulation in PDGFRα+ mesenchymal cells in both Atg5Δ/Δ ileum and

Atg7Δ/Δ ileum were observed, suggesting some PDGFRα+ mesenchymal cells

lost function but most of them maintained intact (Fig. 4.6I). These data

suggested that Atg5Δ/Δ mice by slow deletion overcame the deleterious

phenotype and behave similarly as the Atg7Δ/Δ mice.

Page 113: @2020 Yang Yang ALL RIGHTS RESERVED

98

Page 114: @2020 Yang Yang ALL RIGHTS RESERVED

99

Figure 4.6. Deleting Atg5 more gradually rescues intestinal ileum

function. A. Experimental design for generation of Atg5Δ/Δ and Atg7Δ/Δ mice by

slow deletion. B. Kaplan-Meier survival curve of TAM-treated wild type, Atg7Δ/Δ

and Atg5Δ/Δ mice. * p<0.05 **** p<0.0001 (log-rank Mantel-Cox test) C.

Representative ileum H&E histology from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice

by slow deletion at indicated time points. D. Western blotting for ATG5, ATG7,

LC3 at Day 3 from ileum of wild type, Atg7Δ/Δ and Atg5Δ/Δ mice by slow

deletion. β-ACTIN is used as the loading control. E. Representative duodenum,

jejunum and ileum Immunohistochemistry (IHC) staining of ATG5 and ATG7 at

indicated time points from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice by slow deletion.

F. Representative ileum IHC staining of p62 at indicated time points from wild

type, Atg7Δ/Δ and Atg5Δ/Δ mice by slow deletion. Red arrows indicate p62

aggregates. G. Representative duodenum, jejunum and ileum IHC staining of

OLFM4 at indicated time points from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice by

slow deletion. H. Representative ileum IF costaining of CD34 and PDGFRα at

Day 3 from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice by slow deletion. I.

Representative ileum IF costaining of p62 and PDGFRα at Day 3 on the ileum

from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice by slow deletion. White arrows indicate

colocalization of p62 with PDGFRα. See also Figure S4.9.

Page 115: @2020 Yang Yang ALL RIGHTS RESERVED

100

Autophagy is required to maintain mitochondria function, TCA cycle and

nucleotide metabolism in PDGFRα+ mesenchymal cells

We then sought to test why autophagy is required for the maintenance of

PDGFRα+ mesenchymal cells. It was discovered that autophagy deficiency will

trigger p53 to induce downstream cell apoptosis in lung cancer, breast cancer

and pancreatic cancer, and deletion of p53 will attenuate the tumor growth

(Guo et al., 2013; Huo et al., 2013; Rosenfeldt et al., 2013; Strohecker et al.,

2013; Yang and Kimmelman, 2014; Yang et al., 2014; Yang et al., 2011). Thus,

we crossed the Trp53flox/flox (p53 here after) with our Ubc-CreERT2/+; Atg5flox/flox

mouse to generate Ubc-CreERT2/+; Atg5flox/flox; Trp53flox/flox mice. TAM was

injected to these mice to generate Atg5Δ/Δp53Δ/Δ mice and compared with

Atg5Δ/Δ mice (Fig. S4.10A). Whole body codeletion of p53 together with Atg5

did not rescue the survival of Atg5Δ/Δ mice, suggesting that Atg5 deletion

induced death cannot be rescued by p53 deficiency (Fig. S4.10B).

Autophagy is also required for maintenance of cellular metabolism and

mitochondria function (Guo et al., 2013; Guo et al., 2016; Karsli-Uzunbas et

al., 2014). We then sought to investigate whether autophagy is required to

maintain normal cellular metabolism and mitochondria function in telocytes.

Single cell RNA sequencing data from the PDGFRα+ mesenchymal cells

revealed a higher expression level of several essential autophagy genes in the

crypt and villus telocytes compared to fibroblasts, especially the genes

involved in the autophagosome assembly including Gabarap and Gabarap2,

Page 116: @2020 Yang Yang ALL RIGHTS RESERVED

101

Map1lc3a (LC3-I), Sqstm1 (p62), suggesting higher autophagy activity in

telocytes (Fig. 4.7A). We then performed Matrix-assisted laser desorption

ionization coupled to imaging mass spectrometry (MALDI-IMS) to identify the

level of metabolites on wild type and Atg5Δ/Δ ileum. Fourteen metabolites

showed significant difference between wild type and Atg5Δ/Δ PDGFRα+

mesenchymal cells. These metabolites including TCA cycle intermediates

aconitate and citrate/isocitrate, which is accumulating in Atg5Δ/Δ PDGFRα+

mesenchymal cells compared to wild type, suggesting malfunction of TCA

cycle in Atg5Δ/Δ PDGFRα+ mesenchymal cells (Fig. 4.7B). Atg5Δ/Δ PDGFRα+

mesenchymal cells had more glucose-1 phosphate but less

glycerol-phosphate, suggesting more usage of glycogen but less lipids (Fig.

4.7B). Another group of metabolites were TCA cycle derived amino acids:

aspartate, glutamate and glutamine. Atg5Δ/Δ PDGFRα+ mesenchymal cells

displayed decreased aspartate and glutamate but accumulation of glutamine,

suggesting dysfunction of electron transport chain(ETC) in mitochondria as

previously published (Guo et al., 2016; Jain et al., 2019) (Fig. 4.7B). The other

metabolites were related to nucleotide synthesis and degradation. Atg5Δ/Δ

PDGFRα+ mesenchymal cells showed increased ribose phosphate, AMP,

inosine, hypoxanthine, xanthine and uric acid and greatly decreased cytidine

compared to wild type, indicating increased purine synthesis, purine and

pyramidine degradation (Fig. 4.7B). These data suggested that autophagy

deficient PDGFRα+ mesenchymal cells altered their metabolism with blockage

Page 117: @2020 Yang Yang ALL RIGHTS RESERVED

102

of TCA cycle and increased nucleotide degradation; both related to

mitochondria dysfunction caused by autophagy deficiency (Guo et al., 2016)

Thus, we tested the mitochondria numbers in PDGFRα+ mesenchymal cells.

There’s moderate TOM20 in PDGFRα+ mesenchymal cell at Day one, which

was increased in these cells on day two in Atg5Δ/Δ compared to Atg7Δ/Δ and

wild type ileum, suggesting increased abnormal mitochondria numbers as we

discovered before in autophagy deficient tumors (Guo et al., 2013; Joshi et

al., 2015; Karsli-Uzunbas et al., 2014), indicating the altered metabolism in

Atg5Δ/Δ PDGFRα+ mesenchymal cells was likely to be caused by mitochondria

dysfunction. (Fig. 4.7C, S4.10C).

Page 118: @2020 Yang Yang ALL RIGHTS RESERVED

103

Page 119: @2020 Yang Yang ALL RIGHTS RESERVED

104

Fig. 4.7. Autophagy is required to maintain mitochondria function, TCA

cycle and nucleotide metabolism in PDGFRα+ mesenchymal cells. A.

single cell RNA sequencing data of essential autophagy genes. Y axis, in units

of fraction of cellular mRNA (averaged over all the sequenced cells from the

indicated cluster). B. MALDI-IMS data from wild type and Atg5Δ/Δ ileum at

Day 1 and Day 2 post TAM. The scale bar on the left of each image set

represents the intensity range of ions detected for each metabolite. The white

pixels on the telocyte image panel represent the PDGFRα+ mesenchymal

cells. C. Representative ileum IF costaining of TOMM20 and PDGFRα at

Day 2 from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice. See also Figure S4.10.

Page 120: @2020 Yang Yang ALL RIGHTS RESERVED

105

Fig. 4.8. Mechanism by which autophagy is required for telocytes

survival to maintain intestinal homeostasis.

See text for explanation.

Page 121: @2020 Yang Yang ALL RIGHTS RESERVED

106

Discussion

Previous work has suggested that mice with Atg5, Atg16L1 or Atg7 deficiency

in the intestine epithelium layer (under the control of Villin-Cre) had a granule

abnormality in Paneth cells but the general intestine structure is maintained

and mice can survive (Cadwell et al., 2008; Wittkopf et al., 2012). These

findings indicate that autophagy in the epithelial compartment is not essential

for ileum function, in contrast to the loss of ileum function with the whole body

conditional Atg5 deletion shown here. It remains possible that adaptation to

the loss of autophagy occurs in these models as we observed with slow

deletion of Atg5. Similarly, Atg5 -/- mice with transgenic Atg5 expression in

neurons to rescue neonatal lethality showed intestinal morphological changes

with shorter and wider villi and deeper crypts that did not affect mouse survival

(Yoshii et al., 2016). Adaptation to loss of autophagy may also occur in this

setting. Autophagy in the intestinal epithelium layer is also required to

maintain enough number of intestinal stem cells and prevent irradiation

induced damage and necroptosis (Asano et al., 2017; Matsuzawa-Ishimoto et

al., 2017). All these phenotypes caused by intestine specific autophagy

deficiency were not lethal to the mice. However, in our model we observed a

lethal phenotype caused by acute loss of Atg5 in the whole body with a

lifespan of only 4 days. Even though the major damage site is only in the ileum

part of intestine, the damage is severe with complete loss of stem cells villus

cells besides the granules abnormality in Paneth cells. The key reason for the

Page 122: @2020 Yang Yang ALL RIGHTS RESERVED

107

difference is that in previous study the essential autophagy gene was deleted

only in the epithelium layer of intestine by using the Villin-Cre recombinase but

we deleted the target gene in the whole body. Therefore, we are blocking

autophagy function in more cell populations, especially in the PDGFRα+

mesenchymal cells which are not affected in the Villin-Cre models. Thus, in

Villin-Cre models, the intestinal stem cells still receive decent amount of Wnt

ligands to maintain their renewal and proliferation and mice can survive unless

damage was induced by other factors like irradiation(Asano et al., 2017).

However, in our case, we blocked autophagy function in these PDGFRα+

mesenchymal cells and we found that rapid loss of autophagy caused

depletion of these cells. Therefore, the intestinal stem cells cannot get enough

Wnt ligands to turn on Wnt signaling properly (Fig. 4.8). Thus, the intestinal

homeostasis cannot be maintained and cause the mice to die of hypoglycemia

and loss of intestinal barrier function.

Autophagy is important in maintaining cellular metabolism and the pool size of

TCA cycle intermediates, amino acids and nucleotides (Guo et al., 2016;

Karsli-Uzunbas et al., 2014; Poillet-Perez et al., 2018). In our MALDI-IMS

experiment, the fourteen metabolites which showed significant differences all

belonged to these groups, and aspartate is probably a key metabolite among

all of them. Aspartate is a TCA cycle derived non-essential amino acid, and its

synthesis needs proper activation of ETC in mitochondria, which is required for

Page 123: @2020 Yang Yang ALL RIGHTS RESERVED

108

cell proliferation and growth (Birsoy et al., 2015; Sullivan et al., 2015). Here in

our data, accumulation of glutamine and TOMM20 in Atg5Δ/Δ PDGFRα+

mesenchymal cells suggesting dysfunction of ETC and mitochondria, which

can explain the decreased aspartate in these cells (Jain et al., 2019).

Aspartate also serves as an indispensable component for purine synthesis by

donating its amino group to IMP to make AMP and also directly contributes in

forming the pyramidine ring. However, in Atg5Δ/Δ PDGFRα+ mesenchymal cells,

aspartate cannot be synthesized properly and support nucleotide synthesis

due to loss of ETC function caused by autophagy deficiency; At the same time,

short of energy supply caused nucleotide degradation marked by increased

xanthine and uric acid. These caused decreased nucleotide pool similar as in

the tumor derived cell lines, which is very likely to be the cause of death for

Atg5Δ/Δ PDGFRα+ mesenchymal cells (Guo et al., 2016).

Inflammatory bowel disease (IBD), including Crohn’s disease (CD) and

ulcerative colitis (UC), are complex diseases which start with chronic relapsing

and remitting intestinal inflammation which finally leads to tissue fibrosis. The

driver of IBD is usually a combination of genetic, environmental and microbiota

factors in the intestine or colon (Rieder and Fiocchi, 2008). Several genetic

variants have been identified by GWAS to be related to autophagic defect and

linked to IBD onset. The most outstanding one is Atg16L1, which is an

essential autophagy gene directly involved in autophagosome formation. In

Page 124: @2020 Yang Yang ALL RIGHTS RESERVED

109

patients with CD the essential autophagy gene Atg16L1 is usually a risk allele

with a polymorphism of T300A, and intestine of the patient displays

abnormality in Paneth cells granule secretion as seen in the Villin-Cre Atg16L1

KO mice (Cadwell et al., 2008).This Atg16L1 T300A polymorphism also

causes morphological defect in goblet cells and increased production of

cytokine IL-1β (Lassen et al., 2014). Interestingly, Atg16L1 polymorphism in

intestine causes failure of sensing protective signals from the microbiome,

which is a critical gene-environment etiology for development of IBD (Chu et

al., 2016). Another IBD susceptible gene related to autophagy is NOD2, which

is a pattern recognition receptor that is involved in the homeostasis of intestinal

immunity and works together with Atg16L1. Under normal condition, NOD2 will

be activated by invading bacteria and recruits ATG16L1 to the plasma

membrane at the bacteria invading site and triggers autophagosome formation

to wrap the bacteria for clearance; Mutant NOD2 fails to recruit ATG16L1 to

the plasma membrane and bacteria clearance by autophagy will be impaired

(Cooney et al., 2010; Homer et al., 2010; Travassos et al., 2010). Another

gene identified is ULK1, which can directly phosphorylate wild type Atg16L1 in

response to infection and promotes xenophagy, but will destabilize the mutant

Atg16L1 with CD polymorphism (Alsaadi et al., 2019).

It has been shown that in both types of IBD, the PDGFRα+ mesenchymal cells

were significantly decreased as the disease progressed to advanced stage

Page 125: @2020 Yang Yang ALL RIGHTS RESERVED

110

(Manetti et al., 2015; Milia et al., 2013). In our study, we unraveled an

important role of autophagy in maintaining the survival of PDGFRα+

mesenchymal cells, which is essential for maintaining intestinal homeostasis.

Therefore, the decreased number of PDGFRα+ mesenchymal cells in IBD

patient is very likely caused by gradual mutation or loss of essential autophagy

genes in these cells as the disease gets more advanced. Thus, our study

presents a possible mechanism for development of IBD, and upregulation of

autophagy function in those PDGFRα+ mesenchymal cells or supplementing

Wnt ligands in early IBD patients could potentially serve as a therapy to deal

with this disease.

Page 126: @2020 Yang Yang ALL RIGHTS RESERVED

111

Supplemental Figures

Page 127: @2020 Yang Yang ALL RIGHTS RESERVED

112

Figure S4.1. Related to Figure 4.1. A. Kaplan-Meier survival curve of

TAM-treated wild type, Cag-Cre Atg12Δ/Δ and Cag-Cre Atg5Δ/Δ mice. ****

p<0.0001 (log-rank Mantel-Cox test). B. Representative duodenum

Hematoxyin&Eosin stained (H&E) histology at indicated time points from wild

type, Atg7Δ/Δ and Atg5Δ/Δ mice. C. Representative jejunum H&E histology at

indicated time points from wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice. D.

Representative ileum H&E histology at indicated time points from wild type,

Cag-Cre Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice. E. Representative

duodenum H&E histology at indicated time points from wild type, Cag-Cre

Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice. F. Representative jejunum H&E

histology at indicated time points from wild type, Cag-Cre Atg12Δ/Δ mice and

Cag-Cre Atg5Δ/Δ mice.

Page 128: @2020 Yang Yang ALL RIGHTS RESERVED

113

Page 129: @2020 Yang Yang ALL RIGHTS RESERVED

114

Figure S4.2. Related to Figure 4.1. A. Blood glucose concentration

measured by mg/dL from wild type and Atg5Δ/Δ mice supplemented

with/without glucose solution by IP injection at indicated time points. n.s, not

significant (log-rank Mantel-Cox test). B. Kaplan Meyer survival curve of wild

type and Atg5Δ/Δ mice supplemented with/without glucose solution by IP

injection at indicated time points. n.s, not significant (log-rank Mantel-Cox test).

C. Representative PCR of Atg5 from the Atg5Δ/Δ mice at indicated time points.

D. Representative PCR of Atg7 from the Atg7Δ/Δ mice at indicated time points.

E. Western blotting for ATG5, ATG7, LC3 at indicated time points from the

duodenum of wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice by slow deletion.

β-ACTIN is used as the loading control. F. Western blotting for ATG5, ATG7,

LC3 at indicated time points from the jejunum of wild type, Atg7Δ/Δ mice and

Atg5Δ/Δ mice by slow deletion. β-ACTIN is used as the loading control.

Page 130: @2020 Yang Yang ALL RIGHTS RESERVED

115

Page 131: @2020 Yang Yang ALL RIGHTS RESERVED

116

Figure S4.3. Related to Figure 4.2. A-B. Representative duodenum and

jejunum IHC staining of ATG5 at indicated time points from wild type, Atg7Δ/Δ

mice and Atg5Δ/Δ mice. C-D. Representative duodenum and jejunum IHC

staining of ATG7 at indicated time points from wild type, Atg7Δ/Δ mice and

Atg5Δ/Δ mice. E-F. Representative duodenum and jejunum IHC staining of p62

at indicated time points from wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice. G-H.

Representative ileum p62 staining at indicated time points from wild type,

Cag-Cre Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice. I-J. Representative

duodenum p62 staining at indicated time points from wild type, Cag-Cre

Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice. K-L. Representative jejunum p62

staining at indicated time points from wild type, Cag-Cre Atg12Δ/Δ mice and

Cag-Cre Atg5Δ/Δ mice.

Page 132: @2020 Yang Yang ALL RIGHTS RESERVED

117

Page 133: @2020 Yang Yang ALL RIGHTS RESERVED

118

Figure S4.4. Related to Figure 4.2. A-B. Representative duodenum and

jejunum Alcian staining at indicated time points from wild type, Atg7Δ/Δ mice

and Atg5Δ/Δ mice. C. Representative ileum Alcian staining at indicated time

points from wild type, Cag-Cre Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice. D-E.

Representative duodenum and jejunum Alcian staining at indicated time points

from Cag-Cre Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice. F-G. Representative

duodenum and jejunum IHC staining of OLFM4 at indicated time points from

wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice. H. Representative ileum IHC staining

of OLFM4 at indicated time points from wild type, Cag-Cre Atg12Δ/Δ mice and

Cag-Cre Atg5Δ/Δ mice. I-J. Representative duodenum and jejunum IHC staining

of OLFM4 at indicated time points from wild type, Cag-Cre Atg12Δ/Δ mice and

Cag-Cre Atg5Δ/Δ mice. K-L. Representative duodenum and jejunum IHC

staining of Lysozyme at indicated time points from wild type, Atg7Δ/Δ mice and

Atg5Δ/Δ mice. M. Representative ileum IHC staining of Lysozyme at indicated

time points from wild type, Cag-Cre Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice.

N-O. Representative ileum IHC staining of Lysozyme at indicated time points

from wild type, Cag-Cre Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ mice. P-Q.

Representative duodenum and jejunum IF co staining of p62 and OLFM4 at

indicated time points from wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice.

Page 134: @2020 Yang Yang ALL RIGHTS RESERVED

119

Page 135: @2020 Yang Yang ALL RIGHTS RESERVED

120

Figure. S4.5. Related to Figure 4.3. A. Representative picture of the

untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, and Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 1. B-C. Alive organoid percentage and average budding numbers of

the untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 1. D-G. Quantitative real-time PCR of OLFM4, Lgr5, Axin2 and Sox9

of the untreated Atg5flox/flox organoids, Atg5Δ/Δ organoids, Atg5Δ/Δ organoids

supplemented with Wnt3a or Wnt2b and untreated Atg7flox/flox organoids,

Atg7Δ/Δ organoids, Atg7Δ/Δ organoids supplemented with Wnt3a or Wnt2b at

passage 1. H. Representative picture of the untreated Atg5flox/flox organoids,

Atg5Δ/Δ organoids, and Atg5Δ/Δ organoids supplemented with Wnt3a or Wnt2b

and untreated Atg7flox/flox organoids, Atg7Δ/Δ organoids, Atg7Δ/Δ organoids

supplemented with Wnt3a or Wnt2b at passage 2. I-J. Alive organoid

percentage and average budding numbers of the untreated Atg5flox/flox

organoids, Atg5Δ/Δ organoids, Atg5Δ/Δ organoids supplemented with Wnt3a or

Wnt2b and untreated Atg7flox/flox organoids, Atg7Δ/Δ organoids, Atg7Δ/Δ

organoids supplemented with Wnt3a or Wnt2b at passage 2. K-N. Quantitative

real-time PCR of OLFM4, Lgr5, Axin2 and Sox9 of the untreated Atg5flox/flox

organoids, Atg5Δ/Δ organoids, Atg5Δ/Δ organoids supplemented with Wnt3a or

Page 136: @2020 Yang Yang ALL RIGHTS RESERVED

121

Wnt2b and untreated Atg7flox/flox organoids, Atg7Δ/Δ organoids, Atg7Δ/Δ

organoids supplemented with Wnt3a or Wnt2b at passage 2. O-P. Western

blotting for ATG5, ATG7, and LC3 from the untreated Atg5flox/flox organoids,

Atg5Δ/Δ organoids, and Atg5Δ/Δ organoids supplemented with Wnt3a or Wnt2b

and untreated Atg7flox/flox organoids, Atg7Δ/Δ organoids, Atg7Δ/Δ organoids

supplemented with Wnt3a or Wnt2b at passage 2 and passage 3. β-ACTIN is

used as the loading control.

Page 137: @2020 Yang Yang ALL RIGHTS RESERVED

122

Page 138: @2020 Yang Yang ALL RIGHTS RESERVED

123

Figure S4.6. Related to Figure 4.4. A-B. Representative ileum IHC staining of

Active β-catenin at Day 1 and Day 2 post TAM from wild type, Atg7Δ/Δ mice and

Atg5Δ/Δ mice. C-D. Representative ileum IHC staining of Active β-catenin at

Day 1 and Day 2 post TAM from wild type, Cag-Cre Atg12Δ/Δ mice and

Cag-Cre Atg5Δ/Δ mice. E-F. Representative ileum IF costaining of CD34 and

PDGFRα at Day 1 and Day 2 post TAM from wild type, Atg7Δ/Δ mice and

Atg5Δ/Δ mice. G-H. Representative ileum IF costaining of CD34 and PDGFRα

at Day 1 and Day 2 post TAM from wild type, Cag-Cre Atg12Δ/Δ mice and

Cag-Cre Atg5Δ/Δ mice.

Page 139: @2020 Yang Yang ALL RIGHTS RESERVED

124

Page 140: @2020 Yang Yang ALL RIGHTS RESERVED

125

Figure S4.7. Related to Figure 4.4. A-B. Representative ileum IF costaining

of p62 and PDGFRα at Day 1 and Day 3 post TAM from wild type, Atg7Δ/Δ mice

and Atg5Δ/Δ mice. White arrows indicate colocalization of p62 with PDGFRα.

C-D. Representative ileum IF costaining of p62 and PDGFRα at Day 1 and

Day 2 post TAM from wild type, Cag-Cre Atg12Δ/Δ mice and Cag-Cre Atg5Δ/Δ

mice. White arrows indicate colocalization of p62 with PDGFRα.

Page 141: @2020 Yang Yang ALL RIGHTS RESERVED

126

Page 142: @2020 Yang Yang ALL RIGHTS RESERVED

127

Figure S4.8. Related to Figure 4.5. A. Representative ileum IF costaining of

p62 and PDGFRα at Day 2 from wild type, PDGFRα-Cre Atg7Δ/Δ and

PDGFRα-Cre Atg5Δ/Δ mice. White arrows indicate colocalization of p62 with

PDGFRα. B. Representative ileum Alcian blue staining at indicated time points

from wild type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice. C.

Representative ileum IHC staining of Lysozyme at Day 4 post TAM from wild

type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice. D-E.

Representative duodenum and jejunum H&E histology at indicated time points

from wild type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice. F.

Representative ileum IF costaining of p62 and PDGFRα at Day 8 from wild

type, PDGFRα-Cre Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice. White arrows

indicate colocalization of p62 with PDGFRα. G. Representative duodenum,

jejunum and ileum H&E histology at Day 8 from wild type, PDGFRα-Cre

Atg7Δ/Δ and PDGFRα-Cre Atg5Δ/Δ mice.

Page 143: @2020 Yang Yang ALL RIGHTS RESERVED

128

Page 144: @2020 Yang Yang ALL RIGHTS RESERVED

129

Figure S4.9. Related to Figure 4.6. A-C. Representative liver, muscle,

cerebrum, cerebellum, pancreas, White adipose tissue(WAT) and lung H&E

histology at Day 3, Week 2 and Week 5 from wild type, Atg7Δ/Δ mice and

Atg5Δ/Δ mice by slow deletion. D. Representative liver, muscle, cerebrum,

cerebellum, pancreas, WAT and lung H&E histology at Week twelve from wild

type and Atg5Δ/Δ mice by slow deletion. E. Representative duodenum and

jejunum H&E histology from wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice by slow

deletion at indicated time points. F. Representative PCR of Atg5 from the

Atg5Δ/Δ mice and Atg7 from the Atg7Δ/Δ mice at Day 3 post TAM. G. Western

blotting for ATG5, ATG7, LC3 at Day 3 post TAM from the duodenum and

jejunum of wild type, Atg7Δ/Δ mice and Atg5Δ/Δ mice by slow deletion. β-ACTIN

is used as the loading control. H. Representative duodenum and jejunum IHC

staining of p62 at indicated time points from wild type, Atg7Δ/Δ mice and Atg5Δ/Δ

mice by slow deletion.

Page 145: @2020 Yang Yang ALL RIGHTS RESERVED

130

Figure S4.10. Related to Figure 4.7. A. Experimental design for generation of

Atg5Δ/Δ and Atg5Δ/Δp53Δ/Δ mice. B. Kaplan-Meier survival curve of TAM-treated

wild type, Atg5Δ/Δ and Atg5Δ/Δp53Δ/Δ mice. n.s. not significant (log-rank

Mantel-Cox test) C. Representative ileum IF costaining of TOMM20 and

PDGFRα at Day 1 from wild type, Atg7Δ/Δ and Atg5Δ/Δ mice.

Page 146: @2020 Yang Yang ALL RIGHTS RESERVED

131

CHAPTER 5

REFERENCES

Page 147: @2020 Yang Yang ALL RIGHTS RESERVED

132

Abada, A., and Elazar, Z. (2014). Getting ready for building: signaling and

autophagosome biogenesis. EMBO Rep 15, 839-852.

Alsaadi, R.M., Losier, T.T., Tian, W., Jackson, A., Guo, Z., Rubinsztein, D.C.,

and Russell, R.C. (2019). ULK1-mediated phosphorylation of ATG16L1

promotes xenophagy, but destabilizes the ATG16L1 Crohn's mutant. EMBO

Rep 20, e46885.

Asano, J., Sato, T., Ichinose, S., Kajita, M., Onai, N., Shimizu, S., and Ohteki, T.

(2017). Intrinsic Autophagy Is Required for the Maintenance of Intestinal Stem

Cells and for Irradiation-Induced Intestinal Regeneration. Cell reports 20,

1050-1060.

Barker, N. (2014). Adult intestinal stem cells: critical drivers of epithelial

homeostasis and regeneration. Nat Rev Mol Cell Biol 15, 19-33.

Bhatt, V., Khayati, K., Hu, Z.S., Lee, A., Kamran, W., Su, X., and Guo, J.Y.

(2019). Autophagy modulates lipid metabolism to maintain metabolic flexibility

for Lkb1-deficient Kras-driven lung tumorigenesis. Genes & development 33,

150-165.

Birsoy, K., Wang, T., Chen, W.W., Freinkman, E., Abu-Remaileh, M., and

Sabatini, D.M. (2015). An Essential Role of the Mitochondrial Electron

Transport Chain in Cell Proliferation Is to Enable Aspartate Synthesis. Cell 162,

540-551.

Bode, A.M., and Dong, Z. (2004). Post-translational modification of p53 in

tumorigenesis. Nature reviews Cancer 4, 793-805.

Bravo-San Pedro, J.M., Kroemer, G., and Galluzzi, L. (2017). Autophagy and

Mitophagy in Cardiovascular Disease. Circ Res 120, 1812-1824.

Cadwell, K., Liu, J.Y., Brown, S.L., Miyoshi, H., Loh, J., Lennerz, J.K., Kishi, C.,

Kc, W., Carrero, J.A., Hunt, S., et al. (2008). A key role for autophagy and the

autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature

456, 259-263.

Chan, K., Lu, R., Chang, J.C., and Kan, Y.W. (1996). NRF2, a member of the

NFE2 family of transcription factors, is not essential for murine erythropoiesis,

growth, and development. Proceedings of the National Academy of Sciences of

the United States of America 93, 13943-13948.

Cherra, S.J., 3rd, Kulich, S.M., Uechi, G., Balasubramani, M., Mountzouris, J.,

Day, B.W., and Chu, C.T. (2010). Regulation of the autophagy protein LC3 by

phosphorylation. The Journal of cell biology 190, 533-539.

Chu, C.T., Ji, J., Dagda, R.K., Jiang, J.F., Tyurina, Y.Y., Kapralov, A.A., Tyurin,

V.A., Yanamala, N., Shrivastava, I.H., Mohammadyani, D., et al. (2013).

Cardiolipin externalization to the outer mitochondrial membrane acts as an

elimination signal for mitophagy in neuronal cells. Nature cell biology 15,

1197-1205.

Chu, H., Khosravi, A., Kusumawardhani, I.P., Kwon, A.H., Vasconcelos, A.C.,

Cunha, L.D., Mayer, A.E., Shen, Y., Wu, W.L., Kambal, A., et al. (2016).

Gene-microbiota interactions contribute to the pathogenesis of inflammatory

bowel disease. Science 352, 1116-1120.

Page 148: @2020 Yang Yang ALL RIGHTS RESERVED

133

Condon, K.J., and Sabatini, D.M. (2019). Nutrient regulation of mTORC1 at a

glance. J Cell Sci 132.

Cooney, R., Baker, J., Brain, O., Danis, B., Pichulik, T., Allan, P., Ferguson,

D.J., Campbell, B.J., Jewell, D., and Simmons, A. (2010). NOD2 stimulation

induces autophagy in dendritic cells influencing bacterial handling and antigen

presentation. Nature medicine 16, 90-97.

Copetti, T., Bertoli, C., Dalla, E., Demarchi, F., and Schneider, C. (2009).

p65/RelA modulates BECN1 transcription and autophagy. Mol Cell Biol 29,

2594-2608.

Crighton, D., Wilkinson, S., O'Prey, J., Syed, N., Smith, P., Harrison, P.R.,

Gasco, M., Garrone, O., Crook, T., and Ryan, K.M. (2006). DRAM, a

p53-induced modulator of autophagy, is critical for apoptosis. Cell 126,

121-134.

Criollo, A., Senovilla, L., Authier, H., Maiuri, M.C., Morselli, E., Vitale, I., Kepp,

O., Tasdemir, E., Galluzzi, L., Shen, S., et al. (2010). The IKK complex

contributes to the induction of autophagy. EMBO J 29, 619-631.

Devenish, R.J., and Lai, S.C. (2015). Autophagy and burkholderia. Immunol

Cell Biol 93, 18-24.

Di Bartolomeo, S., Corazzari, M., Nazio, F., Oliverio, S., Lisi, G., Antonioli, M.,

Pagliarini, V., Matteoni, S., Fuoco, C., Giunta, L., et al. (2010). The dynamic

interaction of AMBRA1 with the dynein motor complex regulates mammalian

autophagy. The Journal of cell biology 191, 155-168.

Di Malta, C., Siciliano, D., Calcagni, A., Monfregola, J., Punzi, S., Pastore, N.,

Eastes, A.N., Davis, O., De Cegli, R., Zampelli, A., et al. (2017). Transcriptional

activation of RagD GTPase controls mTORC1 and promotes cancer growth.

Science 356, 1188-1192.

Diao, J., Liu, R., Rong, Y., Zhao, M., Zhang, J., Lai, Y., Zhou, Q., Wilz, L.M., Li,

J., Vivona, S., et al. (2015). ATG14 promotes membrane tethering and fusion of

autophagosomes to endolysosomes. Nature 520, 563-566.

Dikic, I. (2017). Proteasomal and Autophagic Degradation Systems. Annu Rev

Biochem 86, 193-224.

Dikic, I., and Elazar, Z. (2018). Mechanism and medical implications of

mammalian autophagy. Nat Rev Mol Cell Biol 19, 349-364.

Djavaheri-Mergny, M., Amelotti, M., Mathieu, J., Besancon, F., Bauvy, C.,

Souquere, S., Pierron, G., and Codogno, P. (2006). NF-kappaB activation

represses tumor necrosis factor-alpha-induced autophagy. The Journal of

biological chemistry 281, 30373-30382.

Donehower, L.A., Harvey, M., Vogel, H., McArthur, M.J., Montgomery, C.A., Jr.,

Park, S.H., Thompson, T., Ford, R.J., and Bradley, A. (1995). Effects of genetic

background on tumorigenesis in p53-deficient mice. Mol Carcinog 14, 16-22.

Dooley, H.C., Razi, M., Polson, H.E., Girardin, S.E., Wilson, M.I., and Tooze,

S.A. (2014). WIPI2 links LC3 conjugation with PI3P, autophagosome formation,

and pathogen clearance by recruiting Atg12-5-16L1. Molecular cell 55,

238-252.

Page 149: @2020 Yang Yang ALL RIGHTS RESERVED

134

Dotto, G.P. (2009). Crosstalk of Notch with p53 and p63 in cancer growth

control. Nature reviews Cancer 9, 587-595.

Farin, H.F., Van Es, J.H., and Clevers, H. (2012). Redundant sources of Wnt

regulate intestinal stem cells and promote formation of Paneth cells.

Gastroenterology 143, 1518-1529 e1517.

Feng, Z., Hu, W., de Stanchina, E., Teresky, A.K., Jin, S., Lowe, S., and Levine,

A.J. (2007). The regulation of AMPK beta1, TSC2, and PTEN expression by

p53: stress, cell and tissue specificity, and the role of these gene products in

modulating the IGF-1-AKT-mTOR pathways. Cancer research 67, 3043-3053.

Fischer, M. (2017). Census and evaluation of p53 target genes. Oncogene 36,

3943-3956.

Fullgrabe, J., Klionsky, D.J., and Joseph, B. (2014). The return of the nucleus:

transcriptional and epigenetic control of autophagy. Nat Rev Mol Cell Biol 15,

65-74.

Gan-Or, Z., Dion, P.A., and Rouleau, G.A. (2015). Genetic perspective on the

role of the autophagy-lysosome pathway in Parkinson disease. Autophagy 11,

1443-1457.

Gomes, L.C., and Dikic, I. (2014). Autophagy in antimicrobial immunity.

Molecular cell 54, 224-233.

Greicius, G., Kabiri, Z., Sigmundsson, K., Liang, C., Bunte, R., Singh, M.K., and

Virshup, D.M. (2018). PDGFRα+ pericryptal stromal cells are the critical source

of Wnts and RSPO3 for murine intestinal stem cells in vivo. 115,

E3173-E3181.

Guo, J.Y., Karsli-Uzunbas, G., Mathew, R., Aisner, S.C., Kamphorst, J.J.,

Strohecker, A.M., Chen, G., Price, S., Lu, W., Teng, X., et al. (2013). Autophagy

suppresses progression of K-ras-induced lung tumors to oncocytomas and

maintains lipid homeostasis. Genes & development 27, 1447-1461.

Guo, J.Y., Teng, X., Laddha, S.V., Ma, S., Van Nostrand, S.C., Yang, Y., Khor,

S., Chan, C.S., Rabinowitz, J.D., and White, E. (2016). Autophagy provides

metabolic substrates to maintain energy charge and nucleotide pools in

Ras-driven lung cancer cells. Genes & development 30, 1704-1717.

Gupta, J., and Nebreda, A.R. (2014). Analysis of Intestinal Permeability in

Mice. Bio-protocol 4, e1289.

Halpern, K.B., Massalha, H., Zwick, R.K., Moor, A.E., Castillo-Azofeifa, D.,

Rozenberg, M., Farack, L., Egozi, A., Miller, D.R., Averbukh, I., et al. (2019).

Lgr5+ telocytes are a signaling hub at the intestinal villus tip. bioRxiv, 850909.

Hamasaki, M., Furuta, N., Matsuda, A., Nezu, A., Yamamoto, A., Fujita, N.,

Oomori, H., Noda, T., Haraguchi, T., Hiraoka, Y., et al. (2013).

Autophagosomes form at ER-mitochondria contact sites. Nature 495, 389-393.

Hara, T., Nakamura, K., Matsui, M., Yamamoto, A., Nakahara, Y.,

Suzuki-Migishima, R., Yokoyama, M., Mishima, K., Saito, I., Okano, H., et al.

(2006). Suppression of basal autophagy in neural cells causes

neurodegenerative disease in mice. Nature 441, 885-889.

Homer, C.R., Richmond, A.L., Rebert, N.A., Achkar, J.P., and McDonald, C.

Page 150: @2020 Yang Yang ALL RIGHTS RESERVED

135

(2010). ATG16L1 and NOD2 interact in an autophagy-dependent antibacterial

pathway implicated in Crohn's disease pathogenesis. Gastroenterology 139,

1630-1641, 1641 e1631-1632.

Honda, R., Tanaka, H., and Yasuda, H. (1997). Oncoprotein MDM2 is a

ubiquitin ligase E3 for tumor suppressor p53. FEBS letters 420, 25-27.

Hosokawa, N., Hara, T., Kaizuka, T., Kishi, C., Takamura, A., Miura, Y., Iemura,

S., Natsume, T., Takehana, K., Yamada, N., et al. (2009). Nutrient-dependent

mTORC1 association with the ULK1-Atg13-FIP200 complex required for

autophagy. Molecular biology of the cell 20, 1981-1991.

Huo, Y., Cai, H., Teplova, I., Bowman-Colin, C., Chen, G., Price, S., Barnard,

N., Ganesan, S., Karantza, V., White, E., et al. (2013). Autophagy opposes

p53-mediated tumor barrier to facilitate tumorigenesis in a model of

PALB2-associated hereditary breast cancer. Cancer discovery 3, 894-907.

Ichimura, Y., Waguri, S., Sou, Y.S., Kageyama, S., Hasegawa, J., Ishimura, R.,

Saito, T., Yang, Y., Kouno, T., Fukutomi, T., et al. (2013). Phosphorylation of

p62 activates the Keap1-Nrf2 pathway during selective autophagy. Molecular

cell 51, 618-631.

Itakura, E., Kishi-Itakura, C., and Mizushima, N. (2012). The hairpin-type

tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with

endosomes/lysosomes. Cell 151, 1256-1269.

Jain, A., Lamark, T., Sjottem, E., Larsen, K.B., Awuh, J.A., Overvatn, A.,

McMahon, M., Hayes, J.D., and Johansen, T. (2010). p62/SQSTM1 is a target

gene for transcription factor NRF2 and creates a positive feedback loop by

inducing antioxidant response element-driven gene transcription. The Journal

of biological chemistry 285, 22576-22591.

Jain, I.H., Zazzeron, L., Goldberger, O., Marutani, E., Wojtkiewicz, G.R., Ast,

T., Wang, H., Schleifer, G., Stepanova, A., Brepoels, K., et al. (2019). Leigh

Syndrome Mouse Model Can Be Rescued by Interventions that Normalize

Brain Hyperoxia, but Not HIF Activation. Cell metabolism 30, 824-832 e823.

Johansen, T., and Lamark, T. (2011). Selective autophagy mediated by

autophagic adapter proteins. Autophagy 7, 279-296.

Joshi, S., Tolkunov, D., Aviv, H., Hakimi, A.A., Yao, M., Hsieh, J.J., Ganesan,

S., Chan, C.S., and White, E. (2015). The Genomic Landscape of Renal

Oncocytoma Identifies a Metabolic Barrier to Tumorigenesis. Cell reports 13,

1895-1908.

Jung, C.H., Ro, S.H., Cao, J., Otto, N.M., and Kim, D.H. (2010). mTOR

regulation of autophagy. FEBS letters 584, 1287-1295.

Karanasios, E., Stapleton, E., Manifava, M., Kaizuka, T., Mizushima, N.,

Walker, S.A., and Ktistakis, N.T. (2013). Dynamic association of the ULK1

complex with omegasomes during autophagy induction. J Cell Sci 126,

5224-5238.

Karsli-Uzunbas, G., Guo, J.Y., Price, S., Teng, X., Laddha, S.V., Khor, S.,

Kalaany, N.Y., Jacks, T., Chan, C.S., Rabinowitz, J.D., et al. (2014). Autophagy

is required for glucose homeostasis and lung tumor maintenance. Cancer

Page 151: @2020 Yang Yang ALL RIGHTS RESERVED

136

discovery 4, 914-927.

Kaufmann, A., Beier, V., Franquelim, H.G., and Wollert, T. (2014). Molecular

mechanism of autophagic membrane-scaffold assembly and disassembly. Cell

156, 469-481.

Kaur, J., and Debnath, J. (2015). Autophagy at the crossroads of catabolism

and anabolism. Nat Rev Mol Cell Biol 16, 461-472.

Kensler, T.W., Wakabayashi, N., and Biswal, S. (2007). Cell survival responses

to environmental stresses via the Keap1-Nrf2-ARE pathway. Annual review of

pharmacology and toxicology 47, 89-116.

Kenzelmann Broz, D., Spano Mello, S., Bieging, K.T., Jiang, D., Dusek, R.L.,

Brady, C.A., Sidow, A., and Attardi, L.D. (2013). Global genomic profiling

reveals an extensive p53-regulated autophagy program contributing to key p53

responses. Genes & development 27, 1016-1031.

Khoury, K., and Domling, A. (2012). P53 mdm2 inhibitors. Curr Pharm Des 18,

4668-4678.

Kim, J., Kundu, M., Viollet, B., and Guan, K.L. (2011). AMPK and mTOR

regulate autophagy through direct phosphorylation of Ulk1. Nature cell biology

13, 132-141.

Kim, J.E., You, D.J., Lee, C., Ahn, C., Seong, J.Y., and Hwang, J.I. (2010).

Suppression of NF-kappaB signaling by KEAP1 regulation of IKKbeta activity

through autophagic degradation and inhibition of phosphorylation. Cell Signal

22, 1645-1654.

Koentjoro, B., Park, J.S., and Sue, C.M. (2017). Nix restores mitophagy and

mitochondrial function to protect against PINK1/Parkin-related Parkinson's

disease. Sci Rep 7, 44373.

Komatsu, M., Waguri, S., Chiba, T., Murata, S., Iwata, J., Tanida, I., Ueno, T.,

Koike, M., Uchiyama, Y., Kominami, E., et al. (2006). Loss of autophagy in the

central nervous system causes neurodegeneration in mice. Nature 441,

880-884.

Komatsu, M., Waguri, S., Ueno, T., Iwata, J., Murata, S., Tanida, I., Ezaki, J.,

Mizushima, N., Ohsumi, Y., Uchiyama, Y., et al. (2005). Impairment of

starvation-induced and constitutive autophagy in Atg7-deficient mice. The

Journal of cell biology 169, 425-434.

Kruiswijk, F., Labuschagne, C.F., and Vousden, K.H. (2015). p53 in survival,

death and metabolic health: a lifeguard with a licence to kill. Nat Rev Mol Cell

Biol 16, 393-405.

Kubbutat, M.H., Jones, S.N., and Vousden, K.H. (1997). Regulation of p53

stability by Mdm2. Nature 387, 299-303.

Kuma, A., Hatano, M., Matsui, M., Yamamoto, A., Nakaya, H., Yoshimori, T.,

Ohsumi, Y., Tokuhisa, T., and Mizushima, N. (2004). The role of autophagy

during the early neonatal starvation period. Nature 432, 1032-1036.

Lassen, K.G., Kuballa, P., Conway, K.L., Patel, K.K., Becker, C.E., Peloquin,

J.M., Villablanca, E.J., Norman, J.M., Liu, T.C., Heath, R.J., et al. (2014).

Atg16L1 T300A variant decreases selective autophagy resulting in altered

Page 152: @2020 Yang Yang ALL RIGHTS RESERVED

137

cytokine signaling and decreased antibacterial defense. Proceedings of the

National Academy of Sciences of the United States of America 111,

7741-7746.

Lau, A., Wang, X.J., Zhao, F., Villeneuve, N.F., Wu, T., Jiang, T., Sun, Z.,

White, E., and Zhang, D.D. (2010). A noncanonical mechanism of Nrf2

activation by autophagy deficiency: direct interaction between Keap1 and p62.

Mol Cell Biol 30, 3275-3285.

Lazarou, M., Sliter, D.A., Kane, L.A., Sarraf, S.A., Wang, C., Burman, J.L.,

Sideris, D.P., Fogel, A.I., and Youle, R.J. (2015). The ubiquitin kinase PINK1

recruits autophagy receptors to induce mitophagy. Nature 524, 309-314.

Lee, I.H., Cao, L., Mostoslavsky, R., Lombard, D.B., Liu, J., Bruns, N.E.,

Tsokos, M., Alt, F.W., and Finkel, T. (2008). A role for the NAD-dependent

deacetylase Sirt1 in the regulation of autophagy. Proceedings of the National

Academy of Sciences of the United States of America 105, 3374-3379.

Levonen, A.L., Hill, B.G., Kansanen, E., Zhang, J., and Darley-Usmar, V.M.

(2014). Redox regulation of antioxidants, autophagy, and the response to

stress: implications for electrophile therapeutics. Free radical biology &

medicine 71, 196-207.

Liu, K., and Czaja, M.J. (2013). Regulation of lipid stores and metabolism by

lipophagy. Cell death and differentiation 20, 3-11.

Mah, L.Y., O'Prey, J., Baudot, A.D., Hoekstra, A., and Ryan, K.M. (2012).

DRAM-1 encodes multiple isoforms that regulate autophagy. Autophagy 8,

18-28.

Malhotra, R., Warne, J.P., Salas, E., Xu, A.W., and Debnath, J. (2015). Loss of

Atg12, but not Atg5, in pro-opiomelanocortin neurons exacerbates diet-induced

obesity. Autophagy 11, 145-154.

Mancias, J.D., Wang, X., Gygi, S.P., Harper, J.W., and Kimmelman, A.C.

(2014). Quantitative proteomics identifies NCOA4 as the cargo receptor

mediating ferritinophagy. Nature 509, 105-109.

Manetti, M., Rosa, I., Messerini, L., and Ibba-Manneschi, L. (2015). Telocytes

are reduced during fibrotic remodelling of the colonic wall in ulcerative colitis. J

Cell Mol Med 19, 62-73.

Manjithaya, R., Nazarko, T.Y., Farre, J.C., and Subramani, S. (2010).

Molecular mechanism and physiological role of pexophagy. FEBS letters 584,

1367-1373.

Marine, J.C., and Lozano, G. (2010). Mdm2-mediated ubiquitylation: p53 and

beyond. Cell death and differentiation 17, 93-102.

Marino, S., Vooijs, M., van Der Gulden, H., Jonkers, J., and Berns, A. (2000).

Induction of medulloblastomas in p53-null mutant mice by somatic inactivation

of Rb in the external granular layer cells of the cerebellum. Genes &

development 14, 994-1004.

Mathew, R., Karp, C.M., Beaudoin, B., Vuong, N., Chen, G., Chen, H.Y., Bray,

K., Reddy, A., Bhanot, G., Gelinas, C., et al. (2009). Autophagy suppresses

tumorigenesis through elimination of p62. Cell 137, 1062-1075.

Page 153: @2020 Yang Yang ALL RIGHTS RESERVED

138

Mathew, R., Khor, S., Hackett, S.R., Rabinowitz, J.D., Perlman, D.H., and

White, E. (2014). Functional role of autophagy-mediated proteome remodeling

in cell survival signaling and innate immunity. Molecular cell 55, 916-930.

Matsumine, H., Saito, M., Shimoda-Matsubayashi, S., Tanaka, H., Ishikawa,

A., Nakagawa-Hattori, Y., Yokochi, M., Kobayashi, T., Igarashi, S., Takano, H.,

et al. (1997). Localization of a gene for an autosomal recessive form of juvenile

Parkinsonism to chromosome 6q25.2-27. American journal of human genetics

60, 588-596.

Matsumoto, G., Shimogori, T., Hattori, N., and Nukina, N. (2015). TBK1

controls autophagosomal engulfment of polyubiquitinated mitochondria

through p62/SQSTM1 phosphorylation. Hum Mol Genet 24, 4429-4442.

Matsuzawa-Ishimoto, Y., Shono, Y., Gomez, L.E., Hubbard-Lucey, V.M.,

Cammer, M., Neil, J., Dewan, M.Z., Lieberman, S.R., Lazrak, A., Marinis, J.M.,

et al. (2017). Autophagy protein ATG16L1 prevents necroptosis in the intestinal

epithelium. J Exp Med 214, 3687-3705.

Milia, A.F., Ruffo, M., Manetti, M., Rosa, I., Conte, D., Fazi, M., Messerini, L.,

and Ibba-Manneschi, L. (2013). Telocytes in Crohn's disease. J Cell Mol Med

17, 1525-1536.

Mizushima, N. (2010). The role of the Atg1/ULK1 complex in autophagy

regulation. Current opinion in cell biology 22, 132-139.

Mizushima, N., and Komatsu, M. (2011). Autophagy: renovation of cells and

tissues. Cell 147, 728-741.

Mizushima, N., and Levine, B. (2010). Autophagy in mammalian development

and differentiation. Nature cell biology 12, 823-830.

Mizushima, N., Yamamoto, A., Matsui, M., Yoshimori, T., and Ohsumi, Y.

(2004). In vivo analysis of autophagy in response to nutrient starvation using

transgenic mice expressing a fluorescent autophagosome marker. Molecular

biology of the cell 15, 1101-1111.

Montes de Oca Luna, R., Wagner, D.S., and Lozano, G. (1995). Rescue of

early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature

378, 203-206.

Moors, T., Paciotti, S., Chiasserini, D., Calabresi, P., Parnetti, L., Beccari, T.,

and van de Berg, W.D. (2016). Lysosomal Dysfunction and alpha-Synuclein

Aggregation in Parkinson's Disease: Diagnostic Links. Movement disorders :

official journal of the Movement Disorder Society 31, 791-801.

Mortensen, M., Ferguson, D.J., Edelmann, M., Kessler, B., Morten, K.J.,

Komatsu, M., and Simon, A.K. (2010). Loss of autophagy in erythroid cells

leads to defective removal of mitochondria and severe anemia in vivo.

Proceedings of the National Academy of Sciences of the United States of

America 107, 832-837.

Narendra, D.P., Jin, S.M., Tanaka, A., Suen, D.F., Gautier, C.A., Shen, J.,

Cookson, M.R., and Youle, R.J. (2010). PINK1 is selectively stabilized on

impaired mitochondria to activate Parkin. PLoS biology 8, e1000298.

Neumann, Y., Bruns, S.A., Rohde, M., Prajsnar, T.K., Foster, S.J., and

Page 154: @2020 Yang Yang ALL RIGHTS RESERVED

139

Schmitz, I. (2016). Intracellular Staphylococcus aureus eludes selective

autophagy by activating a host cell kinase. Autophagy 12, 2069-2084.

Nguyen, L., Walburger, A., Houben, E., Koul, A., Muller, S., Morbitzer, M.,

Klebl, B., Ferrari, G., and Pieters, J. (2005). Role of protein kinase G in growth

and glutamine metabolism of Mycobacterium bovis BCG. J Bacteriol 187,

5852-5856.

Niida, M., Tanaka, M., and Kamitani, T. (2010). Downregulation of active IKK

beta by Ro52-mediated autophagy. Mol Immunol 47, 2378-2387.

Nishimura, T., Tamura, N., Kono, N., Shimanaka, Y., Arai, H., Yamamoto, H.,

and Mizushima, N. (2017). Autophagosome formation is initiated at

phosphatidylinositol synthase-enriched ER subdomains. EMBO J 36,

1719-1735.

Niture, S.K., and Jaiswal, A.K. (2012). Nrf2 protein up-regulates antiapoptotic

protein Bcl-2 and prevents cellular apoptosis. The Journal of biological

chemistry 287, 9873-9886.

Noad, J., von der Malsburg, A., Pathe, C., Michel, M.A., Komander, D., and

Randow, F. (2017). LUBAC-synthesized linear ubiquitin chains restrict

cytosol-invading bacteria by activating autophagy and NF-kappaB. Nat

Microbiol 2, 17063.

Novak, I., Kirkin, V., McEwan, D.G., Zhang, J., Wild, P., Rozenknop, A., Rogov,

V., Lohr, F., Popovic, D., Occhipinti, A., et al. (2010). Nix is a selective

autophagy receptor for mitochondrial clearance. EMBO Rep 11, 45-51.

Okamoto, K. (2014). Organellophagy: eliminating cellular building blocks via

selective autophagy. The Journal of cell biology 205, 435-445.

Olsvik, H.L., Lamark, T., Takagi, K., Larsen, K.B., Evjen, G., Overvatn, A.,

Mizushima, T., and Johansen, T. (2015). FYCO1 Contains a C-terminally

Extended, LC3A/B-preferring LC3-interacting Region (LIR) Motif Required for

Efficient Maturation of Autophagosomes during Basal Autophagy. The Journal

of biological chemistry 290, 29361-29374.

Pankiv, S., Clausen, T.H., Lamark, T., Brech, A., Bruun, J.A., Outzen, H.,

Overvatn, A., Bjorkoy, G., and Johansen, T. (2007). p62/SQSTM1 binds

directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates

by autophagy. The Journal of biological chemistry 282, 24131-24145.

Pengo, N., Agrotis, A., Prak, K., Jones, J., and Ketteler, R. (2017). A reversible

phospho-switch mediated by ULK1 regulates the activity of autophagy protease

ATG4B. Nature communications 8, 294.

Pohl, C., and Dikic, I. (2019). Cellular quality control by the

ubiquitin-proteasome system and autophagy. Science 366, 818-822.

Poillet-Perez, L., and White, E. (2019). Role of tumor and host autophagy in

cancer metabolism. Genes & development 33, 610-619.

Poillet-Perez, L., Xie, X., Zhan, L., Yang, Y., Sharp, D.W., Hu, Z.S., Su, X.,

Maganti, A., Jiang, C., Lu, W., et al. (2018). Autophagy maintains tumour

growth through circulating arginine. Nature 563, 569-573.

Rabinowitz, J.D., and White, E. (2010). Autophagy and metabolism. Science

Page 155: @2020 Yang Yang ALL RIGHTS RESERVED

140

330, 1344-1348.

Real, E., Rodrigues, L., Cabal, G.G., Enguita, F.J., Mancio-Silva, L.,

Mello-Vieira, J., Beatty, W., Vera, I.M., Zuzarte-Luis, V., Figueira, T.N., et al.

(2018). Plasmodium UIS3 sequesters host LC3 to avoid elimination by

autophagy in hepatocytes. Nat Microbiol 3, 17-25.

Richter, B., Sliter, D.A., Herhaus, L., Stolz, A., Wang, C., Beli, P., Zaffagnini, G.,

Wild, P., Martens, S., Wagner, S.A., et al. (2016). Phosphorylation of OPTN by

TBK1 enhances its binding to Ub chains and promotes selective autophagy of

damaged mitochondria. Proceedings of the National Academy of Sciences of

the United States of America 113, 4039-4044.

Rieder, F., and Fiocchi, C. (2008). Intestinal fibrosis in inflammatory bowel

disease - Current knowledge and future perspectives. J Crohns Colitis 2,

279-290.

Rioux, J.D., Xavier, R.J., Taylor, K.D., Silverberg, M.S., Goyette, P., Huett, A.,

Green, T., Kuballa, P., Barmada, M.M., Datta, L.W., et al. (2007). Genome-wide

association study identifies new susceptibility loci for Crohn disease and

implicates autophagy in disease pathogenesis. Nature genetics 39, 596-604.

Rockel, J.S., and Kapoor, M. (2016). Autophagy: controlling cell fate in

rheumatic diseases. Nat Rev Rheumatol 12, 517-531.

Rosenfeldt, M.T., O'Prey, J., Morton, J.P., Nixon, C., MacKay, G., Mrowinska,

A., Au, A., Rai, T.S., Zheng, L., Ridgway, R., et al. (2013). p53 status

determines the role of autophagy in pancreatic tumour development. Nature

504, 296-300.

Rubinsztein, D.C., Codogno, P., and Levine, B. (2012). Autophagy modulation

as a potential therapeutic target for diverse diseases. Nat Rev Drug Discov 11,

709-730.

Ruzankina, Y., Pinzon-Guzman, C., Asare, A., Ong, T., Pontano, L., Cotsarelis,

G., Zediak, V.P., Velez, M., Bhandoola, A., and Brown, E.J. (2007). Deletion of

the developmentally essential gene ATR in adult mice leads to age-related

phenotypes and stem cell loss. Cell Stem Cell 1, 113-126.

Saito, T., Ichimura, Y., Taguchi, K., Suzuki, T., Mizushima, T., Takagi, K.,

Hirose, Y., Nagahashi, M., Iso, T., Fukutomi, T., et al. (2016). p62/Sqstm1

promotes malignancy of HCV-positive hepatocellular carcinoma through

Nrf2-dependent metabolic reprogramming. Nature communications 7, 12030.

Sakamaki, J.I., Wilkinson, S., Hahn, M., Tasdemir, N., O'Prey, J., Clark, W.,

Hedley, A., Nixon, C., Long, J.S., New, M., et al. (2017). Bromodomain Protein

BRD4 Is a Transcriptional Repressor of Autophagy and Lysosomal Function.

Molecular cell 66, 517-532 e519.

Sato, T., Vries, R.G., Snippert, H.J., van de Wetering, M., Barker, N., Stange,

D.E., van Es, J.H., Abo, A., Kujala, P., Peters, P.J., et al. (2009). Single Lgr5

stem cells build crypt-villus structures in vitro without a mesenchymal niche.

Nature 459, 262-265.

Satoo, K., Noda, N.N., Kumeta, H., Fujioka, Y., Mizushima, N., Ohsumi, Y., and

Inagaki, F. (2009). The structure of Atg4B-LC3 complex reveals the mechanism

Page 156: @2020 Yang Yang ALL RIGHTS RESERVED

141

of LC3 processing and delipidation during autophagy. EMBO J 28, 1341-1350.

Saxton, R.A., and Sabatini, D.M. (2017). mTOR Signaling in Growth,

Metabolism, and Disease. Cell 169, 361-371.

Scherz-Shouval, R., Shvets, E., Fass, E., Shorer, H., Gil, L., and Elazar, Z.

(2007). Reactive oxygen species are essential for autophagy and specifically

regulate the activity of Atg4. EMBO J 26, 1749-1760.

Schlottmann, S., Buback, F., Stahl, B., Meierhenrich, R., Walter, P., Georgieff,

M., and Senftleben, U. (2008). Prolonged classical NF-kappaB activation

prevents autophagy upon E. coli stimulation in vitro: a potential resolving

mechanism of inflammation. Mediators Inflamm 2008, 725854.

Schneider, J.L., and Cuervo, A.M. (2014). Autophagy and human disease:

emerging themes. Curr Opin Genet Dev 26, 16-23.

Seibenhener, M.L., Babu, J.R., Geetha, T., Wong, H.C., Krishna, N.R., and

Wooten, M.W. (2004). Sequestosome 1/p62 is a polyubiquitin chain binding

protein involved in ubiquitin proteasome degradation. Mol Cell Biol 24,

8055-8068.

Sentelle, R.D., Senkal, C.E., Jiang, W., Ponnusamy, S., Gencer, S., Selvam,

S.P., Ramshesh, V.K., Peterson, Y.K., Lemasters, J.J., Szulc, Z.M., et al.

(2012). Ceramide targets autophagosomes to mitochondria and induces lethal

mitophagy. Nat Chem Biol 8, 831-838.

Settembre, C., Di Malta, C., Polito, V.A., Garcia Arencibia, M., Vetrini, F., Erdin,

S., Erdin, S.U., Huynh, T., Medina, D., Colella, P., et al. (2011). TFEB links

autophagy to lysosomal biogenesis. Science 332, 1429-1433.

Shoshkes-Carmel, M., Wang, Y.J., Wangensteen, K.J., Toth, B., Kondo, A.,

Massasa, E.E., Itzkovitz, S., and Kaestner, K.H. (2018). Subepithelial telocytes

are an important source of Wnts that supports intestinal crypts. Nature 557,

242-246.

Singh, R., Kaushik, S., Wang, Y., Xiang, Y., Novak, I., Komatsu, M., Tanaka, K.,

Cuervo, A.M., and Czaja, M.J. (2009). Autophagy regulates lipid metabolism.

Nature 458, 1131-1135.

Stolz, A., Ernst, A., and Dikic, I. (2014). Cargo recognition and trafficking in

selective autophagy. Nature cell biology 16, 495-501.

Strohecker, A.M., Guo, J.Y., Karsli-Uzunbas, G., Price, S.M., Chen, G.J.,

Mathew, R., McMahon, M., and White, E. (2013). Autophagy sustains

mitochondrial glutamine metabolism and growth of BrafV600E-driven lung

tumors. Cancer discovery 3, 1272-1285.

Stzepourginski, I., Nigro, G., Jacob, J.M., Dulauroy, S., Sansonetti, P.J., Eberl,

G., and Peduto, L. (2017). CD34+ mesenchymal cells are a major component

of the intestinal stem cells niche at homeostasis and after injury. Proceedings of

the National Academy of Sciences of the United States of America 114,

E506-E513.

Su, H., Yang, F., Wang, Q., Shen, Q., Huang, J., Peng, C., Zhang, Y., Wan, W.,

Wong, C.C.L., Sun, Q., et al. (2017). VPS34 Acetylation Controls Its Lipid

Kinase Activity and the Initiation of Canonical and Non-canonical Autophagy.

Page 157: @2020 Yang Yang ALL RIGHTS RESERVED

142

Molecular cell 67, 907-921 e907.

Sullivan, L.B., Gui, D.Y., Hosios, A.M., Bush, L.N., Freinkman, E., and Vander

Heiden, M.G. (2015). Supporting Aspartate Biosynthesis Is an Essential

Function of Respiration in Proliferating Cells. Cell 162, 552-563.

Sullivan, L.B., Luengo, A., Danai, L.V., Bush, L.N., Diehl, F.F., Hosios, A.M.,

Lau, A.N., Elmiligy, S., Malstrom, S., Lewis, C.A., et al. (2018). Aspartate is an

endogenous metabolic limitation for tumour growth. Nature cell biology 20,

782-788.

Terman, A., and Brunk, U.T. (2005). Autophagy in cardiac myocyte

homeostasis, aging, and pathology. Cardiovasc Res 68, 355-365.

Toledo, F., and Wahl, G.M. (2006). Regulating the p53 pathway: in vitro

hypotheses, in vivo veritas. Nature reviews Cancer 6, 909-923.

Towers, C.G., Fitzwalter, B.E., Regan, D., Goodspeed, A., Morgan, M.J., Liu,

C.W., Gustafson, D.L., and Thorburn, A. (2019). Cancer Cells Upregulate

NRF2 Signaling to Adapt to Autophagy Inhibition. Dev Cell 50, 690-703 e696.

Travassos, L.H., Carneiro, L.A., Ramjeet, M., Hussey, S., Kim, Y.G.,

Magalhaes, J.G., Yuan, L., Soares, F., Chea, E., Le Bourhis, L., et al. (2010).

Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma

membrane at the site of bacterial entry. Nat Immunol 11, 55-62.

Tripathi, D.N., Chowdhury, R., Trudel, L.J., Tee, A.R., Slack, R.S., Walker, C.L.,

and Wogan, G.N. (2013). Reactive nitrogen species regulate autophagy

through ATM-AMPK-TSC2-mediated suppression of mTORC1. Proceedings of

the National Academy of Sciences of the United States of America 110,

E2950-2957.

Tsukamoto, S., Kuma, A., Murakami, M., Kishi, C., Yamamoto, A., and

Mizushima, N. (2008). Autophagy is essential for preimplantation development

of mouse embryos. Science 321, 117-120.

Valenta, T., Degirmenci, B., Moor, A.E., Herr, P., Zimmerli, D., Moor, M.B.,

Hausmann, G., Cantu, C., Aguet, M., and Basler, K. (2016). Wnt Ligands

Secreted by Subepithelial Mesenchymal Cells Are Essential for the Survival of

Intestinal Stem Cells and Gut Homeostasis. Cell reports 15, 911-918.

van Wijk, S.J.L., Fricke, F., Herhaus, L., Gupta, J., Hotte, K., Pampaloni, F.,

Grumati, P., Kaulich, M., Sou, Y.S., Komatsu, M., et al. (2017). Linear

ubiquitination of cytosolic Salmonella Typhimurium activates NF-kappaB and

restricts bacterial proliferation. Nat Microbiol 2, 17066.

Vassilev, L.T., Vu, B.T., Graves, B., Carvajal, D., Podlaski, F., Filipovic, Z.,

Kong, N., Kammlott, U., Lukacs, C., Klein, C., et al. (2004). In vivo activation of

the p53 pathway by small-molecule antagonists of MDM2. Science 303,

844-848.

Walden, H., and Muqit, M.M. (2017). Ubiquitin and Parkinson's disease through

the looking glass of genetics. Biochem J 474, 1439-1451.

Webb, A.E., and Brunet, A. (2014). FOXO transcription factors: key regulators

of cellular quality control. Trends in biochemical sciences 39, 159-169.

Wei, J., Long, L., Yang, K., Guy, C., Shrestha, S., Chen, Z., Wu, C., Vogel, P.,

Page 158: @2020 Yang Yang ALL RIGHTS RESERVED

143

Neale, G., Green, D.R., et al. (2016). Autophagy enforces functional integrity of

regulatory T cells by coupling environmental cues and metabolic homeostasis.

Nat Immunol 17, 277-285.

Wild, P., Farhan, H., McEwan, D.G., Wagner, S., Rogov, V.V., Brady, N.R.,

Richter, B., Korac, J., Waidmann, O., Choudhary, C., et al. (2011).

Phosphorylation of the autophagy receptor optineurin restricts Salmonella

growth. Science 333, 228-233.

Wilkinson, D.S., Jariwala, J.S., Anderson, E., Mitra, K., Meisenhelder, J.,

Chang, J.T., Ideker, T., Hunter, T., Nizet, V., Dillin, A., et al. (2015).

Phosphorylation of LC3 by the Hippo kinases STK3/STK4 is essential for

autophagy. Molecular cell 57, 55-68.

Wittkopf, N., Gunther, C., Martini, E., Waldner, M., Amann, K.U., Neurath, M.F.,

and Becker, C. (2012). Lack of Intestinal Epithelial Atg7 Affects Paneth Cell

Granule Formation but Does Not Compromise Immune Homeostasis in the

Gut. Clin Dev Immunol.

Yang, A., and Kimmelman, A.C. (2014). Inhibition of autophagy attenuates

pancreatic cancer growth independent of TP53/TRP53 status. Autophagy 10,

1683-1684.

Yang, A., Rajeshkumar, N.V., Wang, X., Yabuuchi, S., Alexander, B.M., Chu,

G.C., Von Hoff, D.D., Maitra, A., and Kimmelman, A.C. (2014). Autophagy is

critical for pancreatic tumor growth and progression in tumors with p53

alterations. Cancer discovery 4, 905-913.

Yang, S., Wang, X., Contino, G., Liesa, M., Sahin, E., Ying, H., Bause, A., Li, Y.,

Stommel, J.M., Dell'antonio, G., et al. (2011). Pancreatic cancers require

autophagy for tumor growth. Genes & development 25, 717-729.

Yee, K.S., Wilkinson, S., James, J., Ryan, K.M., and Vousden, K.H. (2009).

PUMA- and Bax-induced autophagy contributes to apoptosis. Cell death and

differentiation 16, 1135-1145.

Yoshii, S.R., Kuma, A., Akashi, T., Hara, T., Yamamoto, A., Kurikawa, Y.,

Itakura, E., Tsukamoto, S., Shitara, H., Eishi, Y., et al. (2016). Systemic

Analysis of Atg5-Null Mice Rescued from Neonatal Lethality by Transgenic

ATG5 Expression in Neurons. Dev Cell 39, 116-130.

Zhang, X.D., Wang, Y., Wang, Y., Zhang, X., Han, R., Wu, J.C., Liang, Z.Q.,

Gu, Z.L., Han, F., Fukunaga, K., et al. (2009). p53 mediates mitochondria

dysfunction-triggered autophagy activation and cell death in rat striatum.

Autophagy 5, 339-350.

Zhao, J., Brault, J.J., Schild, A., Cao, P., Sandri, M., Schiaffino, S., Lecker,

S.H., and Goldberg, A.L. (2007). FoxO3 coordinately activates protein

degradation by the autophagic/lysosomal and proteasomal pathways in

atrophying muscle cells. Cell metabolism 6, 472-483.