Classification and E Classification and Effect of Corr ect of Correctors on Sit ors on Sitosterolemia- olemiaAssociated Mutants in ABCG8emiaAssociated Mutants in ABCG8 Brittney Poole Univ

Category
CAA

 

 

University of Kentucky

UUKKnnoowwlleeddggee

 

Theses and Dissertations--Medical Sciences

Medical Sciences

2022

Classification and Effect of Correctors on Sitosterolemia- Associated Mutants in ABCG8

Brittney Poole

University of Kentucky, brittney.poole@uky.com

Author ORCID Identifier:

http://orcid.org/0000-0002-0955-8815

Digital Object Identifierhttps://doi.org/10.13023/etd.2022.339

RRiigghhtt cclliicckk ttoo ooppeenn aa ffeeeeddbbaacckk ffoorrmm iinn aa nneeww ttaabb ttoo lleett uuss kknnooww hhooww tthhiiss ddooccuummeenntt bbeenneefifittss yyoouu..

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Recommended Citation

Poole, Brittney, "Classification and Effect of Correctors on Sitosterolemia-Associated Mutants in ABCG8" (2022). Theses and Dissertations--Medical Sciences. 23.

https://uknowledge.uky.edu/medsci_etds/23

 

This Master's Thesis is brought to you for free and open access by the Medical Sciences at UKnowledge. It has been accepted for inclusion in Theses and Dissertations--Medical Sciences by an authorized administrator of  UKnowledge. For more information, please contact UKnowledge@lsv.uky.edu.


 

STUDENT AGREEMENT:

I represent that my thesis or dissertation and abstract are my original work. Proper attribution has been given to all outside sources. I understand that I am solely responsible for obtaining  any needed copyright permissions. I have obtained needed written permission statement(s)   from the owner(s) of each third-party copyrighted matter to be included in my work, allowing   electronic distribution (if such use is not permitted by the fair use doctrine) which will be

submitted to UKnowledge as Additional File.

I hereby grant to The University of Kentucky and its agents the irrevocable, non-exclusive, and royalty-free license to archive and make accessible my work in whole or in part in all forms of media, now or hereafter known. I agree that the document mentioned above may be made

available immediately for worldwide access unless an embargo applies.

I retain all other ownership rights to the copyright of my work. I also retain the right to use in future works (such as articles or books) all or part of my work. I understand that I am free to register the copyright to my work.

REVIEW, APPROVAL AND ACCEPTANCE

The document mentioned above has been reviewed and accepted by the student’s advisor, on behalf of the advisory committee, and by the Director of Graduate Studies (DGS), on behalf of

the program; we verify that this is the final, approved version of the student’s thesis including all  changes required by the advisory committee. The undersigned agree to abide by the statements above.

Brittney Poole, Student

DrGregory AGrafMajor Professor

Dr. Melinda Wilson, Director of Graduate Studies


 

  
 

 

 
 

 

 

 

 

 


 

CLASSIFCATION AND EFFECT OF CORRECTORS ON SITOSTEROLEMIA-

ASSOCIATED MUTANTS IN ABCG8

 

 

 

 

 

 

 

THESIS

 

A thesis submitted in partial fulfillment of the

requirements for the degree of Master of Science in the

College of Medicine

at the University of Kentucky

 

 

 

By

Brittney Poole

Lexington, Kentucky

DirectorDrGregory AGrafProfessor of Pharmaceutical Sciences

Lexington, Kentucky

 

 

 

 

 

 

 

Copyright © Brittney Poole 2022

http://orcid.org/0000-0002-0955-8815


ABSTRACT OF THESIS

 

 

 

CLASSIFICATION AND EFFECT OF CORRECTORS ON SITOSTEROLEMIA- ASSOCIATED CYTOSOLIC MUTANTS IN ABCG8

 

 

Objective: To classify mutants of ABCG8 identified in subjects with clinically confirmed Sitosterolemia, a rare form of Familial Hypercholesterolemia distinguished by the

accumulation of phytosterols in plasma and tissues and determine the effects of correctors

and/or regulators of proteostasis on maturation of the ABCG5/ABCGsterol transporter.

MethodsDisease-causing missense mutants within the cytosolic domain of ABCG8 were generated through site-directed mutagenesis. Normal and mutant proteins were expressed  in human hepatocytes. Cellular proteins were prepared, and maturation was assessed by

SDS-PAGE and immunoblotting. Formation of the higher molecular weight, mature form of glycoproteins was used as a bioassay for trafficking the G5G8 complex beyond the

Endoplasmic ReticulumThe impact of correctors and regulators of proteostasis on Class II mutant maturation was also determined.

Results: Approximately 44% of cytosolic, Sitosterolemia-associated mutants in ABCG8 are maturation incompetent. Of those which matured beyond the ER, 60% were not able to traffic to the cell membrane. Of the mutants that did not mature, none were able to be rescued by small molecular chaperones (correctors).

Conclusion: HuH-7 cells are an efficiently transfected cell line that provides a system to manipulate ABCG5 and ABCG8 to make conclusions about protein maturation and

trafficking to the cell surface. These experiments gave insight into the complexity of

diseases caused by genetic mutations and the underlying mechanism of loss-of-function mutationsFurther experimentation would be required to determine the fate of the CFTR correctors and/or regulators of proteostasis in the application in cases of Sitosterolemia.

 

 

KEYWORDS: Sitosterolemia, ABC transporters, lipids, correctors, proteostasis

 

Brittney Poole

(Name of Student)

 

07/27/22

Date


 

 

 

 

 

 

 

 

 

 

 

 

 

CLASSIFICATION AND EFFECT OF CORRECTORS ON SITOSTEROLEMIA- ASSOCIATED CYTOSOLIC MUTANTS IN ABCG8

 

 

By

Brittney Poole

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Gregory AGraf

 

Director of Thesis

Melinda Wilson

Director of Graduate Studies

07/27/22

Date


 

 

 

 

 

 

 

ACKNOWLEDGMENTS

 

The following thesis, while an individual work, benefited from the insights and direction of several people. First, my Thesis Chair, Dr. Gregory A. Graf, who provided a space to have a “first day in the lab.” Dr. Graf was not only supportive during the research process,  but also  challenged  me  to  understand the  literature  and  develop  my  own scientific thinking. Next, I wish to thank the Thesis Committee: Dr. Scott Gordon and Dr. Ryan Temel. Each individual provided insights that guided and challenged my thinking. Next, I would like to thank all the lab members of the Graf and Helsleylab for their support and expertise in the lab.  I would also like to acknowledge the use of BioRender to create

the figures in this thesis.

 

In  addition to  the  assistance mentioned  above,  I received  equally  important assistance from family and friends. To my family, Brett, Sonja, and Courtney Poole

provided on-going support while navigating living in a new state alone.


 

 

 

TABLE OF CONTENTS

 

 

 

CLASSIFCATION AND EFFECT OF CORRECTORS ON SITOSTEROLEMIA-

ASSOCIATED MUTANTS IN ABCG8 ..................................................................... i

ABSTRACT OF THESIS ......................................................................................... ii

ACKNOWLEDGMENTS........................................................................................ iii

TABLE OF CONTENTS.......................................................................................... iv

LIST OF TABLES................................................................................................... vi

LIST OF FIGURES ............................................................................................... vii

CHAPTER 1.  Introduction.................................................................................... 1

1.1 Background........................................................................................................ 1

1.2 Cholesterol vs. Phytosterols ................................................................................ 4

1.3 Sterol Absorption and Excretion........................................................................... 5

1.4 Sitosterolemia ................................................................................................... 8

1.5 ABCG5/G8 Physiology...................................................................................... 10

1.6 ABC Transporters ............................................................................................ 11

1.7 Proteostasis Regulation and Roscovitine............................................................... 15

1.8 Ivacaftor and ABCC7 Potentiation ..................................................................... 19

1.9 Statement of Hypothesis.................................................................................... 22

CHAPTER 2.  Materials and Methods ............................................................... 23

2.1 Materials and Methods ...................................................................................... 23

2.1.1      Reagents.................................................................................................................... 23

2.1.2      Cell Culture............................................................................................................... 23

2.1.3      GFP Assay ............................................................................................................... 24

2.1.4      Western Botting Analysis ....................................................................................... 24

2.1.5      In-vitro Bioassay...................................................................................................... 25

2.1.6      Densitometric Analysis............................................................................................ 25

2.1.7      ImmunofluorescenceMicroscopy ......................................................................... 26

2.1.8      Statistical Analysis................................................................................................... 26

2.2 Experiment I- Generation of Sitosterolemia Associated ABCG8 Cytosolic Mutants... 27

2.3 Experiment IIOptimization of Transient Transfection of Human Cell Lines............ 30

2.4 Experiment III- Mutant Maturation Assay ........................................................... 31

2.5 Experiment IV- Native ABCG5/G8 Complex Compound Screening........................ 33

2.6 Experiment V- Roscovitine Toxicity .................................................................. 34

2.7 Experiment VI- Corrector Testing of Class II Mutants........................................... 34

2.8 Experiment VIIImmunofluorescence of Maturation CompetentMutants ............... 35


 


 


 

LIST OF FIGURES

 

 

 

FIGURE 1.2-1 CHOLESTEROL AND PHYTOSTEROL STRUCTURE  ............................................................................... 5

FIGURE 1.4-1 DIAGRAM DEMONSTRATING THE ABSORPTIONEXCRETION, AND SECRETION PATHWAY OF

PHYTOSTEROLS AND CURRENT THERAPEUTICS FOR SITOSTEROLEMIA...................................................... 9

FIGURE 1.6-1 STRUCTURE OF ABC TRANSPORTER FAMILIES45............................................................................... 13

FIGURE 2.2-1 EXAMPLE OF SITE-DIRECTED MUTAGENSIS RESTRICTION ENZYMDIGEST AND SEQUENCE

VERIFICATION (GENERATED USING SNAPGENE SOFTWARE VERSION 4.3.11)  ........................ 29

FIGURE 2.4-1 GRAPHICAL REPRESENTATION OF IN VITRO MATURATION ASSAY........................................... 32

FIGURE 3.1-1 ABCG8 MUTATION  DIAGRAM .................................................................................................................. 37

FIGURE 3.1-2. ABCG8 SEQUENCE CONSERVATION AMONG SPECIES................................................................... 38

FIGURE 3.1-3. ABCG8 SEQUENCE CONSERVATION AMONG ABCG FAMILY MEMBERS   ....................... 38

FIGURE 3.2-1 COMPARISON OF DIFFERENT TRANSFECTION REAGENTS IN THE HUH-7 CELL LINE   .. 40

FIGURE 3.2-2 TRANSFECTION  OPTIMIZATION IN HUH-7 CELLS.............................................................................. 41

FIGURE 3.3-1. PROTEIN MATURATION BIOASSAY DEMONSTRATING ABCG8 CYTOSOLIC MUTANTS CO-

TRANSFECTED WITH ABCG5-MYC  ........................................................................................................................ 42

FIGURE 3.3-2. PROTEIN STABILITY WESTERN BLOTS OF MUTANTS IN ABCG8  ............................... 43

FIGURE 3.3-3RESTRICTION ENZYME DIGEST ON CLASS II MUTANTS  ............................................................ 44

FIGURE 3.3-4. GEL ELECTROPHORESIS OF CDNA FROM HUH-7 LYSATES  ..................................................... 45

FIGURE 3.4-1 PROTEIN MATURATION BIOASSAY WITH TREATMENT WITH PUBLISHED CONCENTRATIONS OF

CFTR CORRECTORS68   .................................................................................................................................................... 46

FIGURE 3.5-1. ROSCOVITINE SCREENING ON NATIVE ABCG5/G8 COMPLEX IN HUH-7 CELLS AT 100 UM.......................................................................................................................................................... 47

FIGURE 3.5-2. ROSCOVITINE SCREENING ON NATIVE ABCG5/G8 COMPLEX IN HEPG2 CELLS AT 100 UM.......................................................................................................................................................... 48

FIGURE 3.6-1 DOSE-RESPONSE DATA ON ROSCOVITINE IN HUH-7 CELLS  .................................................... 49

FIGURE 3.7-1. IMMUNOFLUORESCENCE AND IMAGES FROM THE CYCLOHEXIMIDE TIME COURSE EXPERIMENT.

...................................................................................................................................................... 50

FIGURE 3.7-2. TIME-COURSE WITH 50 AND 100 UG/ML CHX ........................................................... 51

FIGURE 3.7-3 IMMUNOFLUORESCENCE IMAGES OF ABCG8 MUTANTS.............................................................. 52


 

 

 

 

CHAPTER 1. INTRODUCTION

 

1.1 Background

Sitosterolemia is a rare form of familial hypercholesterolemia (FH) caused by two mutations in either the ATP-Binding Cassette protein (ABC) G5 or G8 gene, which is in

close proximity of 375 bp between the initiation codons and share a common promotor

 

that function in ahead-to-head orientation 1,2,3 . Sitosterolemia results from a lack of

function for ABCGor Gin the absence of theirrespective binding partnerwhich is

 

distinguished from other forms of FH. Sitosterolemia is an autosomal recessive inherited disease that affects about 1 in 200,000 individuals; however, it is unclear how often this

disease is misdiagnosed. Clinical laboratory assays fail to distinguish cholesterol from

 

phytosterols.  Consequently, plasma from a patient with Sitosterolemia would present  with what appears to be elevated total plasma cholesterol in clinical lab testing. Gas or

liquid chromatography is required to distinguish phytosterols from cholesterol, a

 

technique and instrumentation often unavailable in clinical laboratories.

 

Individuals diagnosed with FH and Sitosterolemia similarly present with

 

xanthomas and premature coronary artery disease. The two diseases are distinguished by

dominant vs. recessive genetics, clinical presentation, and sterol composition in the

 

plasma. FH patients present with elevated LDL cholesterol, while Sitosterolemia patients

present with increased plasma phytosteroldecreased excretion of phytosterols and

cholesterol, and hemolytic and blood disorders 4,5 . The underlying genetic causes of FH

and Sitosterolemia differ. In cases of FH, the mutations affect either LDL-R, its ligand

 

Apolipoprotein B (ApoB100), or the machinery required for LDL/LDL-R internalization,


 

LDL Receptor Related Protein-Associated Protein 1 (LRPAP1) or proprotein convertase

subtilisin/kexin type 9 (PCKS9) while Sitosterolemia results from genetic mutations in

 

ABCGand/or ABCG8.

 

Many ABC transporters are associated with diseases such as; Cystic Fibrosis

 

(ABCC7), Progressive familial intrahepatic cholestasis type 3 (ABCB4), among others (Table 1.1)6. Investigation and FDA-approved drugs (Roscovitine; CFTR modulators)

partially restore function to ABCC7 and ABCB4 mutants based on their underlying

 

molecular defect. The rationale behind testing these modulators on cytosolic mutants of ABCG8 is that if these correctors and regulators of proteostasis are effective in multiple

ABC transporters, then due to the evolutionarily conserved nature of the ABC

 

transporters (Fig. 1.2- 1), these may also be effective for mutants of ABCG87.


 

Table 1.1 ABC Transporters and their associated disease46.

 

ABCTransporter

Associated Disease

ABCA1

Tangier’s Disease/Familial Hypoapoproteinemia

ABCA4

Stargardt’s Disease

ABCB2/3

Immune Deficiency

ABCB4

PFIC3

ABCB7

Anemia

ABCB11

PFIC2

ABCC2

Dubin-Johnson Syndrome

ABCC6

Pseudoxanthoma Elasticum

ABCC7

Cystic Fibrosis

ABCD1

X-linked Adrenoleukodystrophy (ALD)

Table 1.1   shows  different ABC transporters and their  associated diseases, demonstrating a wide range of diseases caused due to mutations in ABC transporters.

 

We hypothesize that small molecule correctors will enhance the

 

maturation of native G5/G8 complex and Class II, maturation deficient mutants of

ABCG8.


 

1.2 Cholesterol vs. Phytosterols

Sterols  are  an  essential  cellular  component  of  eukaryotic membranes,  with cholesterol in animal cells and phytosterols in plant cells. Cholesterol creates rigidity and curvature to the plasma membrane of animal cells. Cholesterol is acquired through diet or is generated by de novo synthesis. Cholesterol biosynthesis is a tightly regulated process that demonstrates negative feedback inhibition. When cholesterol is in excess, it can be toxic to the cell, while cells depleted of cholesterol cannot undergo normal physiological responses,  for  example, receptor  signaling8.  The transcription  factor  sterol regulatoryelement-binding protein 2 (SREBP2) regulates gene expression of the enzymes responsible for  cholesterol  biosynthesis and  uptake  to maintain  homeostasis9. While  endogenous cholesterol  synthesis  is  tightly  regulated,  dietary   cholesterol  ranges due  to   dietary

preferences and maybe a pro-atherogenic factor10.

 

Phytosterols are not endogenously synthesized de novo and are strictly enter from the diet. Phytosterols are structurally similar to cholesterol, but differ from cholesterol by  the side chain on the D ring of the sterol backbone (Fig. 1). For this reason, chromatography  and/or mass spectrophotometry is required to distinguish phytosterols from cholesterol in the  plasma.  Phytosterols  have  an  observed  toxicity  in  the  body,  as  a  study  of ABCG5/ABCG8 KOmice fed a high-phytosterol showed signs of premature death, cardiac lesions, liver damage, and hepatosplenomegaly11. With a functional ABCG5/8 transporter effectively opposing absorption and excreting of phytosterols into the feces, mice and  humans are not affected by high-phytosterol containing diets11. In atypical lipid panel used to measure cholesterol, cholesterol oxidase attacks the 3ß -hydroxyl group on the A ring of

cholesterol. This structural feature is shared amongsterols (animal/phytosterols) (Fig. 1) 12.


 

GC-Mass Spectrometry would be required to separate phytosterols from cholesterol in a

plasma sample.

 

It has been proposed that phytosterols help reduce LDL cholesterol by competing with cholesterol for intestinal absorption resulting in a modest reduction (30-50%) of cholesterol absorption and a 10% decrease in total plasma cholesterol13. The mechanism by which phytosterols compete with cholesterol for intestinal absorption is by reducing the solubilization into the mixed micelle. However, other studies suggest phytosterols promote

cholesterol secretion13,14,15 .

 

Figure 1.2- 1 Cholesterol and Phytosterol structure.

 

 

Figure 1.2- 1  Chemical structure of cholesterol and 4 of the most common phytosterols showing the differences in side chains as well as the common 3ß-Hydroxyl group. The red circle indicates common  -Hydroxyl amongst sterols and ring structures are labelled in  cholesterol.

 

1.3 Sterol Absorption and Excretion

As previously mentionedcholesterol can enter the body in one of two ways.

 

Cholesterol-esters and phytosterols that enter the body exogenously are metabolized by

enzymes (pancreatic lipases) and emulsified with bile salts and phospholipids that enter


 

the intestinal lumen via the gallbladder, ABCB11, and ABCB4, respectively, along with

 

dietary cholesterol to form mixed micelles. Mixed micelles act as the "cholesterol

 

acceptor" for dietary sterols16. Free cholesterol and phytosterols are absorbed from the

mixed micelle into the small intestine (duodenum/jejunum) by NPC1L1 17,18 . NPC1L1

 

(Niemann-Pick C1 Like 1) functions at the apical surface of enterocytes and mediates the internalization of cholesterol and phytosterol to promote absorption19. In the enterocyte,

cholesterol is esterified by the enzyme ACAT-2 (Acyl-CoA: Cholesterol

 

Acyltransferase), which preferentially esterifies cholesterol relative tophytosterols20.

 

Cholesterol is incorporated into chylomicrons, secreted into the lymphatic system, and

 

enters the plasma compartment via the left subclavian vein. Cholesterol and plant sterols that are not esterified by ACAT are exported by ABCG5/G8 back to the intestinal lumen.

In the plasma compartment, chylomicrons and triglycerides are metabolized by

 

Lipoprotein lipases (LPL), generating a smaller, cholesterol-rich chylomicron remnant, which can enter the liver via LDL-R or other related receptors. This pathway delivers

dietary cholesterol andre-absorbed biliary cholesterol to the liver.

 

Another mechanism by which cholesterol can enter the liver is reverse cholesterol transport (RCT). RCT is a pathway in which cholesterol accumulated in peripheral tissue is transported through the plasma to the liver for excretion as neutral sterols or bile acids

in the feces21. Peripheral tissues, such as macrophages, export excess cholesterol by

 

ABCA1 to a pre-HDL particle composed of a lipidated Apo-A1. This process is

 

dependent on the Lecithin Cholesterol Acyltransferase (LCAT) enzyme. LCAT esterifies

cholesterol to fatty acids to generate cholesterol esters that get deposited in the core of

HDL particles22. As opposed to ACAT, LCAT is thought to freely esterify phytosterols to


 

generate mature, spherical HDL. ABCG1, SR-B1, CD36, and other possible sterol transporting enzymes can deliver esterified sterols in the form of HDL to the liver

through Scavenger Receptor Class B I (SR-BI)23.

 

Once cholesterol enters the hepatocyte, it can be excreted in one of two ways. The first pathway is metabolism by cytochrome P450 enzymes (ex. CYP7A1/CYP27A1) into

bile salts. Bile salts are secreted at the canalicular membrane of hepatocytes by the

 

transporter BSEP (Bile salt export protein, ABCB11)23. The other mechanism by which   cholesterol can be excreted from the liver is as free cholesterol via ABCG5/8,which also functions at the hepatocyte canalicular membrane. However, 95% of bile acids and 80%

of cholesterol secreted will be re-absorbed in the intestine by apical sodium bile acid

 

transporter (ASBT) and Niemann-Pick C1-Like 1 (NPC1L1)25,26  . Alternatively, sterols

can return to the plasma in the form of VLDL particles along with Apo-B100,

 

triglycerides, and LDL particles are secreted from the liver a shared mechanism with    chylomicrons and ultimately give rise to the LDL particle. Both chylomicron remnants

and LDL are pro-atherogenicparticles.

 

The Transintestinal Cholesterol Excretion (TICE) pathway is an alternative

 

pathway to hepatobiliary excretion of cholesterol from the bodyTICE is the process of

cholesterol transport from the bloodstream, across the enterocyte, and directly into the

 

intestinal lumen27. Cholesterol is taken up by LDL-R on the basolateral membrane of

enterocytes (with possibilities for another pathway)28 . Cholesterol is then translocated

 

across the enterocyte and secreted via ABCG5/8 at the apical membrane for excretion in

the feces or possible reabsorption by NPC1L128. This alternate pathway is thought to

account for 30-50% of RCT29.


 

1.4 Sitosterolemia

In 1974, two sisters presented with tuberous xanthomas and plasma plant sterols that accounted for roughly 30% of their total plasma sterolsIn atypical human subject,

plant sterols are typically only detected in trace amounts due to poor absorption, only

about 5%4. Both parents of the sisters that unaffected, leading to the conclusion that this rare lipid disease was inherited in an autosomal recessive manner4. It wasn’t until 2000

that ABCGand ABCG8 were discovered to be the two genes mutated in clinically

 

confirmed patients with Sitosterolemia30. In 2003, it was confirmed that ABCG5 and

 

ABCG8 function as obligate heterodimers, with both proteins being required to reach the

cell surface and transport sterols across the membrane31. In addition to the previously

 

reported phenotypes of Sitosterolemic patients, subjects with clinically confirmed

Sitosterolemia could also present with hematological abnormalities, including

 

macrothrombocytopenia, hemolytic anemia, and splenomegaly5.

 

Currently, the primary treatment(s) for Sitosterolemic patients are dietary

 

modifications (low sterol diet), Ezetimibe, bile acid-binding resins, ileal bypass surgery, or LDL apheresis. However, these treatments decrease plasma phytosterols and improve

symptoms but fail to return plasma sterols to the range of a normal human subject32.

 

Ezetimibe is an NPC1L1 inhibitor, reduces cholesterol, and phytosterol absorption by

 

~50%, and is currently the primary pharmacotherapy used to treat cases of

Sitosterolemia33. While Ezetimibe has shown efficacy in reducing phytosterol

 

accumulation, the drug does not target ABCG5/G8. This is important because ABCG5/8

promotes sterol, primarily phytosterol, efflux at the enterocyte and hepatocyte apical

 

surface, whereas NPC1L1 primarily functions in sterol absorption in the enterocyte.


 

NPC1L1 is highly expressed in both the liver and intestine and oppose biliary sterol

secretion in humans, while there is low expression in mice liver19. Even if phytosterol

 

uptake into absorption is opposed, there is still a lack of hepatic secretion and an inability

to eliminate phytosterols from plasma and tissues once accumulated (Fig. 1.4- 1).

 

Figure 1.4- 1 Diagram demonstrating the absorption, excretion, and secretion pathway of

phytosterols and current therapeutics for Sitosterolemia.

 

Figure 1.4- 1 Pathway for excretion and secretion of phytosterols in the liver and intestine. Diagram also exhibits the mechanism of action for ezetimibe, the most common current treatment management of Sitosterolemia.

 

In a 10-year follow-up study of two sisters who had a homozygous nonsense

 

mutation (R446X inABCG5), treatment with Ezetimibe only moderately decreased

 

phytosterol levels while diet changes had little to no change on phytosterol levels34.

 

Starting phytosterol (sitosterol and campesterol) levels were greater than 300 µmol/L and

decreased to ~100 µmol/L, however,the normal range for a healthy individual is 10-20

 

µmol/L34. While Ezetimibe is a form of Sitosterolemia management, these patients still

have far higher cholesterol and phytosterol absorption, which can increase the risk of


 

cardiovascular events later in life. This demonstrates a need for a more specific

 

pharmacologic agent that targets the underlying molecular defect of ABCG5 and ABCG8

disease-causing mutants as opposed to disease management through Ezetimibe.

 

 

 

1.5 ABCG5/G8 Physiology

ABCG5 and ABCG8 (ABCG5/8) are two ABC-half transporters that function as a

heterodimer at the apical membrane of hepatocytes (liver) and enterocytes (small

 

intestine)30. ABCG5 and ABCG8 at the transcriptional level have been shown to be

 

regulated by two nuclear receptors, Liver X Receptors (LXR) and Farnesoid X Receptors (FXR). LXR“ and β belong to the family of nuclear receptors that are master regulators

of genes involved in cholesterol elimination pathways and form heterodimers with

 

Retinoid X Receptors (RXR)35These transcriptional factors areactivated by cholesterol metabolitesoxysterols36It was determined that gene expression of ABCG5 and ABCG8was increased when mice were fed a high cholesterol diet in wildtype mice but in LXR-/- ,

indicating the two genes are also regulated by the LXR/RXR transcription factor37.

 

ABCGand ABCG8 can also be upregulated by the FXR pathwaya nuclear receptor

 

activated in the presence of bile acids37. ABCGand ABCG8 are also positively regulated by the transcription factors hepatocyte nuclear factor 4(HNF4“), Forkhead box protein

O1 (FOXO1), and Liver receptor homolog 1 (LRH1), but the relationships to sterol

 

homeostasis are not as clear38,39 .

 

Beyond the nucleus, ABCG5 and ABCG8 are independently translated in the

 

rough ER. It is unclear how ABCG5 and ABCG8 emerge from the ribosome and find

 

theirrespective binding partnerWithin the ERABCGand ABCGare recognized by


 

molecular chaperones, such as calnexin, that facilitate folding of the ABCG5/ABCG8

complex in an N-linked glycan-dependent mechanism. Chaperones recognize the

 

terminal glucose on theN-linked glycana tag that sorts proteins for their destination. Proteins that are unfolded are recognized by a glucosyltransferase in the ER and the

protein is either is re-glucosylated or exits the chaperone folding “cycle." If in fact

 

ABCG5/8 have dimerized, they are sorted to the apical membrane through the cell's

secretory pathways to function as a heterodimer40.

 

In summary, ABCG5/G8 function as an obligate heterodimer at the apical surface of hepatocytes and enterocytesIn the liverABCG5/8 promotes secretion of phytosterols and cholesterol into bile31. In the intestine, ABCG5/G8 opposes phytosterol absorption by NPC1L1. In mice, expression in one organ, either liver or intestine, is sufficient to protect the animal from Sitosterolemia and the downstream effects of phytosterol accumulation41.

However, it is unclear whether this remains true regarding RCT.

 

 

 

1.6 ABC Transporters

ABC transporters are a family of integral proteins that function to translocate

 

substrates across membranes, a mechanism powered by ATP hydrolysis7. The ABC

 

transporter superfamily consists of 48 proteins and can be further divided into

subfamilies, A-G42The key characteristics of ABC transporters are two nucleotide-

 

binding domains (NBD), two transmembrane domains (TMD) that have 12 membrane-

spanning alpha-helices, and two ATP-Binding Cassette’s (ABC), however, there are

exceptions among the ABC transporters (Fig. 2)42,43 . Among the 48 transporters in

humans, there are half transporters (ABCG5/8) and full transporters (for example,


 

ABCB1).  TheNBD of ABC transporters is a domain that is highly conserved and

 

includes the Walker A motif, Walker B motif, Walker C motif (signature motif), Q-loop,

and H-loop (switch motif)6,7 .

 

While structurally similar across ABC transporters, the TMD sequence differs

 

significantly across and within families, likely a reflection of different substrate binding    pockets6TheNBD binds and hydrolyzes ATP to power the pump6ATP binds to both of theNBDs which forms a tight dimer and two ATPase active sites44A helix in the TMD

fits into a groove on theNBD, resulting in the opening and closing of theNBD

simultaneously to transmembrane helices movement44.


 

 

 

Figure 1.6- 1 Structure of ABC transporter families45.

 

 

Figure 1.6- 1 Structure of the ABC transporter families showing the Transmembrane domain and Nucleotide Binding domain, as well    as the structural differences between each of the families, most notably differences in whether theNBD resides at theN or C-terminus.

 

ABC Transporters import (prokaryotes) or export (eukaryotes) substrates via ATP

hydrolysis cycling7.  Substrate translocation begins with the substrate binding to the

 

TMD and initiating a conformational change from an open to closed conformation. After

the substrate is bound to the TMDATP can bind to theNBDwhich induces the ATP

 

"power stroke” and promotes the closed formation of the TMD. ATP is then hydrolyzed

into ADP and inorganic phosphate (Pi), effectively translocating the substrate. The

 

release of ADP and Pi indicates the end of the ATP hydrolysis cycle. The protein is  restored to its original open conformation and is ready to transport another substrate

across the membrane6.


 

The G family of ABC transporters all function as dimers and transport sterols47.

 

Sterols, and a number of other lipids, are amphipathic and insoluble in water. Sterols

 

require carriers and transporters to move through the body and plasma membrane.

 

ABCG1, 2, and 4 are functionally active as homodimers whileABCG5/G8 is functionally

active as an obligate heterodimer. ABCG1/4 have been proposed as candidates for

 

heterodimerization due to their sequence similarity, although the evidence is lacking48.

ABCG5/8 specifically functions to secrete sterols from hepatocytes into bile and from

 

enterocytes back to the intestinal lumen for excretion. Both hepatocytes and enterocytes

are polarized cell types each expressing an apical and basolateral membrane that serve

 

different functions for the cell in addition to different transporter makeup. NBS2 is

 

thought to be the driver of conformational change with respect to ATP hydrolysis and

sterol transport while NBS1 binds ATP but does not hydrolyze it49. NBS1 and NBS2 are

located within ABCG5 and ABCG849.

 

Many ABC transporters that are disease-causing, such as ABCC7/Cystic Fibrosis

or ABCB4/PFIC3, have systems to classify mutations50,51 . Shown in Table 1.2 is the

 

proposed classification system for Sitosterolemia-associated G5/G8 mutations based on

the systems mentioned above already in place52. Class I mutants are

 

nonsense/frameshift/deletion mutations, whereas classes II-V aremissense mutations.

 

Class II mutants are defective in maturing beyond the Endoplasmic Reticulum, class III    mutants are ones in which the protein is properly folded but lack functional activity, class IV mutants are unstable, and class V mutants are proposed to be defective in trafficking

at the apical surface.


 

Table 1.2. Proposed Sitosterolemia Classification System52

 

CLASS

DESCRIPTION

ABCG5 MUTANTS

ABCG8 MUTANTS

I

Nonsense,

Frameshift, Deletion

57 known or

predicted

58 known or predicted

II

Maturation

R389H, R419P, N437K

R189H, P231T, R236Q

III

Activity

 

 

IV

Stability

 

 

V

Trafficking

 

 

Unclassified

Inconclusive Results

E146Q

R543S

Table 1.2. Table of proposed Sitosterolemia classification system in addition to previously published classifications of ABCG5 and G8 mutants.

 

 

 

1.7 Proteostasis Regulation and Roscovitine

Proteostasis (protein homeostasisrefers to the process of maintaining a protein's

quaternary structure and location, typically through transcriptional/translational

modifications53. Proteostasis influences protein synthesis, folding, trafficking,

 

disaggregation, and degradation53. The ER is responsible for the synthesis, folding,

 

maturationand trafficking of transmembrane proteins as well as secretory proteins. In

disease states where proteins are misfolded, proteostasis pathways detect and rapidly

 

degrade themisfolded proteins via the Endoplasmic Reticulum-Associated Degradation

(ERAD) pathway. Disruptions in protein folding trigger of ER Stress and activate the

 

Unfolded Protein Response (UPR). Conditions that disrupt protein homeostasis and

 

activate UPR are changes in temperature, calcium concentration disturbance, mutations,

redox disruptions, and changes in cellular pH54.


 

The unfolded protein response (UPRis activated when the cellsprotein folding

machinery becomes overwhelmed and misfolded proteins accumulate in the ER55.

 

Disruption of protein homeostasis occurs in conditions as previously mentioned,

 

including mutant proteins, alterations in gene expression, or permanently when there are

mutations that affect protein maturation56. The ERAD (ER-Associated Degradation)

 

pathway is an essential step in newly synthesized protein quality control57.  Chaperones  recognize proteins such as calnexin/calreticulin and heat-shock proteins, which facilitate

folding. Transmembrane proteins are glycosylated with an N-linked glycan and the

 

terminal glucose acts a tag that the protein is unfolded and thus recognized by

 

calnexin/calreticulinProteins that are unfolded are recognized by aglucosyltransferase in the ER and the protein is either re-glucosylated or exits the chaperon folding "cycle."

When prolongedthemisfolded protein is targeted by the retro-translocon and/or E3

ligases58Themisfolded protein is then retrotranslocated to the cytoplasmubiquitinated

by E3 ligases, recognized by the 19S cap of the 26S proteasome, and degraded58.

 

Roscovitine is a small molecule pharmacologic that arrests cell cycle progression by inhibiting cyclin-dependent kinases (CDK) 1, 2, 5, 7, and 959. Roscovitine was first    studied in ABC transporters in its rescue of an ABCC7 (CFTR) mutant, F508del60. The

group suggested that the mechanism of action of Roscovitine F508del correction was

 

likely independent of inhibition of CDK's andmore generally in inhibition of kinases as

well as directly inhibiting activity of the proteasome60This led to the proposal of

 

Roscovtine's rescue ofF508del through modulation of proteostasisspecifically the

ERQC (ER quality control) and ERAD system independently60. The research done by

this group influenced another group (Falguières) to test Roscovitine on three ABCB4


 

mutants in an attempt to achieve similar outcomes61. The three mutants tested were class  II (maturation deficient) mutations61. Because ATP transporters are well conserved across ABC transporter families, there are potential compounds successfully used in other ABC

transporter families that could be repurposed to treat Sitosterolemia-associated ABCG8

 

mutations.

 

Mutations in class II that get retained in the ER would make a compound like

 

Roscovitine a viable target for "rescuing" defective ABCG5/G8 transporters, as it has

 

shown the ability to modulate proteostasis60. It has been well established that ABCG5/G8

are poor folding proteins, as only about 40-50% of the protein matures beyond the ER,

which strengthens the argument for potential stabilization31.  Roscovitine has significant

cellular toxicity. Therefore, 11 analogs were synthesized in an attempt to lower CDK

 

inhibition and cytotoxic activity (Table 1.3)61. Some of these analogs partially rescue  mutants of ABCB4, which strengthens the hypothesis that Roscovitine rescue works    independently of CDK inhibition in ABCCand ABCB4. The efficacy of Roscovitine across multiple ABC transporters (ABCB4/ABCC7) and their structural similarities to

ABCG5/G8 is the scientific premise of this project.


 

Table 1.3. Roscovitine and Analogs Structure.

 

 

Compound Name

 

Compound Structure

 

Compound Name

 

Compound Structure

 

Roscovitine

 

HN

N

N

HO

N

N

N

H

 

MRT2-237

 

HN

 

HO

N

H

N

O

 

Aftin-4

 

N

HO

N

N              N

H

 

MRT2-239

 

F

F

HN

N                      

N

                         N

N              N

HO

              H                                   

O                                              

 

M3

 

 

 

 

HO

O

 

HN

N

N

N

N

N

H

 

MRT2-241

 

F

HN

HO

N

N             N

H

O

 

MRT2- 163

 

HN

HO

N

N

N

H

O

 

MRT2-243

 

 

 

 

HO

O

 

F

HN

       N

N                  

N               N

       N

H                                   

 

MRT2- 164

 

HN

N

HO

N

O

 

MRT2-245

 

HN

HO

N

N              N

H

O

 

MRT2-235

 

HN

HO

N

N

N

H

O

 

MRT2-249

 

F

F

HN

HO

N

H

N

N

O

      

 

Table 1.3. Table of Roscovitine and 11 analogs synthesized by ManRos Therapeutics61.


 

1.8 Ivacaftor and ABCC7 Potentiation

Cystic Fibrosis (CF) is a disease in which the Cystic Fibrosis Transmembrane

 

Regulator (CFTR),ABCC7fails to flux chloride ions leading to an accumulation of

 

chloride ions in the epithelial cells and in turn a change in mucus viscosity in the

lungs62,63 . Ivacaftor is an FDA-approved Cystic Fibrosis treatment that potentiates

 

(increased channel open probability) CFTR and partially restores chloride ion transport.

ABCC7 differs from most ABC transporters as it is an ion channel that utilizes ATP

 

hydrolysis, whereas ABCG5/8 is a protein transporter that uses energy from ATP to

 

efflux sterols64. Ivacaftor potentiator function in CFTR has therapeutic benefits in

patients with CFTR gating (class III) defective mutations50,65 . The mechanism of action

of Ivacaftor is binding to the PKA-phosphorylated ABCC7 in an ATP-independent

fashion66. Ivacaftor functions mainly for mutations that disrupt gating due to disruption to the ATP-dependent binding site66. It has not been reported that regulation of ABCG5 and

ABCG8 is PKA-dependent and ABCG5/G8 does not operate as an ion channel.

 

Therefore, there is not a strong foundation for why Ivacaftor would be able to stabilize

mutants of ABCG8.

 

However, in maturation, defective mutants of CFTR are “rescued” by small

 

molecule correctors of the Ivacaftor-family (Table 1.4), which may facilitate folding of

ABCG5/G8. Elexacaftor, Tezacaftor, and Lumacaftor are compounds that were

 

formulated after Ivacaftor for CF mutants that were not gating defective. They are small molecular "correctorsas they correct themisfolding of proteins that fail to exit the ER. The correctors' mechanism of action is directly binding to CFTR, but how they function

as a protein folding chaperone to escape degradation remains unclear. Rescued CFTR


 

mutants localize to the cell surface67. This suggests that escaping ERAD is sufficient for activity. In more recent studies, looking at the cryo-EM structure of CFTR in complex

with Lumacaftor or Tezacaftor demonstrates that the drugs bind to CFTR in the

 

hydrophobic pocket of TMD- 1, which ultimately stabilized the domain and prevented

 

degradation68. Lumacaftor and Tezacaftor bind within the hydrophobic pocket at a Lysine

and Arginineresidue before the start of the TMD- 168ABCGis structurally similar in

that it too has a Lysine and Arginine residue prior to the start of the first TM domain49.

One could hypothesize that because of this structural similarity there is potential for

 

Lumacaftor or Tezacaftor binding to ABCG5 at the apical membrane.

 

There is amuch larger family of CFTR modulators (including correctors) that are

being investigated in vitro that have demonstrated stabilization in mutants of multiple

 

ABC transporter systems. Most notably, correction of ABCG2 mutants involved in Gout

by Cor-4a and due to sequence homology may too be effective in stabilizing

 

Sitosterolemia-associated mutants69. Stabilization with these and other compounds could

give reason to believe that these compounds could be used in Class II mutants across a

 

wide range of ABC transporter-related diseases.


 

Table 1.4. Ivacaftor and Correctors Structure

 

Compound Name

Compound Structure

Ivacaftor

 

 

O

O

N

H

 

 

 

 

N H

Lumacaftor

 

 

N

 

 

 

 

H N

 

 

 

O

 

 

 

F

O

F

O

Elexacaftor

 

 

 

 

 

O

S

O

 

F

 

O

N

N

N

N

O

Tezacaftor

F           O

F      

O

 

H N

OH

O

N

HO

HO

         

Table 1.4: Chemical structures of Ivacaftor and CFTR correctors. Compounds were purchased from Medchemexpress or Selleckchem.


 

1.9 Statement of Hypothesis

This thesis aims to characterize Sitosterolemia-associated cytosolic

 

mutants in ABCG8 based on their underlying molecular defects. The impact of correctors

on maturation deficient mutants (class II) were tested for efficacy in correcting protein

 

maturation in other ABC transporter families. For maturation competent mutants, we

 

analyzed via immunofluorescence microscopy the degree to which mutants traffic to the

plasma membrane. Maturation of ABCG8 Sitosterolemia-associated mutants and the

 

effect of correctorswill be studied in vitroin a human hepatocyte carcinoma cell line.

 

We hypothesize that when transiently expressed in human hepatocytes, some

 

ABCG8 missense mutations compromise protein folding, transporter complex formation, and trafficking beyond the Golgi. The addition of a compound with proteostatic regulator or CFTR corrector activity used to treat diseases caused by other ABC transporters may

correct folding defects in ABCG8 caused by class II missensemutations.


 

CHAPTER 2. MATERIALS AND METHODS

 

2.1 Materials and Methods

2.1.1    Reagents

 

Roscovitine and its 11 analogs were generously sent to us by Laurent Meijer

 

(ManRos Therapeutics). The CFTR modulators were ordered through Medchemexpress

or SelleckChem (Elexacaftor). DrMahmood Hussian (NYU Langone),provided the

 

HuH-7 cells to test in our experiments. Antibodies used in our Western blotting and

 

Immunofluorescent protocol include 1B10A5 Mouse anti-ABCG8 (Novus), in-house

 

mouse anti-human ABCG8 hybridoma notated as KWE5, Rabbit anti-c-Myc (Upstate

 

cell Signaling Solutions 06-549), Mouse anti- βactin (Sigma A5441- 100UL), Rabbit anti- Calnexin (Enzo Lifesciences ADI-SPA-860-F), and E-Cadherin (Cell Signaling). For the

LDH Assay, the Cytoscan LDH Cytotoxicity assay kit (G-Biosciences 786-324) was

 

utilized to measure cytotoxicity.

 

2.1.2    Cell Culture

 

HepG2 and HuH-7 cells were grown and cultured in Dulbecco’s Modified EagleMedium (DMEM)-High Glucose supplemented with 10% FBS, 1% Pen-Strep, and 1%

GlutaMax. HEK293 cells were grown and cultured in DMEM- High Glucose

 

(Dulbecco’s Modified Eagle Medium) supplemented with 5% FBS, 1% Pen-Strep, and

1% GlutaMax. All cell lines were incubated at 37°C and 5% CO2 .


 

2.1.3    GFP Assay

 

GFP Lysates were made by washing the cells with 2x PBS and incubating the cells

 

in  GFP  Lysis  Buffer  (25  mM  Tris  (pH=7.8),  2mM trans- 1,2-diaminocyclohexane- N’N’N’N’-tetraacetic acid, 1% Triton X- 100, 10% Glycerol) for 30 minutes at 37°C and then 30 minutes at room temperature. The lysates were then imaged on a plate reader for fluorescence  (excitation 485  nm/emission  515  nm) and  absorbance  of (280 nm)  and

expressed as fluorescence 515/280 (arbitrary units).

 

 

 

2.1.4    Western Botting Analysis

 

Cell   lysates  were  analyzed   for  protein   concentration  with  a   Bicinchoninic

 

acid (BCA) assay incubated at 37°C for 30 minutes and measured at an absorbance of 562 nm. Protein concentration was determined by a bovine serum albumin (BSA) standardcurve in each assay. Samples were prepared in 5X Laemelli Buffer (Tris Base (250 mM, pH 6.8), SDS (2.5%), glycerol (50%), and bromophenol blue (0.125%)) and equal protein (15-25 ug) were loaded onto a 4-8% bis-acryland ran at 100 V for approximately 2-3 hours to ensure the differences in immature/mature protein glycosylation were present. The gelswere then transferred to a nitrocellulose membrane at 100 V at 4°C for 1 hour or using

BioRad’s semi-dry Turboblot system.


 

2.1.5    In-vitro Bioassay

 

 

 

Monitoring the shift in electrophoretic mobility on an SDS-PAGE gel is indicative

 

of modifications to the N-linked glycan residue(s) attached in the third extracellular loop of  either  ABCG5  or   8.  We  used  this  change  in  apparent  molecular  weight  in immunoblotting  for ABCG5  or  G8  as  a marker  for  maturation. We  quantified  these measures by densitometric analysis and expressed the data as a percent (mature signal over total signal), an index (mature signal over immature signal), or total mature signal to

loading control.

 

 

 

2.1.6    Densitometric Analysis

 

Gel files were analyzed for densitometry using Adobe Photoshop. Once the gel

 

image was cropped, aligned, and at desired contrast, the gel image was uploaded into Photoshop and aduplicate image was created. The duplicate image was inverted and using the measurement tool, captured the integrated density for each band in the blot (mature band, immature band, and loading control) along with a background measurement. For each signal, the measurement size kept constant. The background was subtracted from each

value and expressed as described for each experiment.


 

2.1.7    Immunofluorescence Microscopy

 

HuH-7 cells were seeded in a 10 cm dish with 6 UV-sterilized coverslips at ~1x106

 

cells. On day one, coverslips were transferred to a 6 well dish, and transfected with wildtype hG5-myc and either wildtype hG8 or mutant hG8. On day two, cells were fixed with Methanol by an adaptation of Ann Hubbards “Cassio” protocol. Cells were incubated with a primary antibody and a secondary conjugated fluorophore with either a 488 or 568 Alexa-Fluor tag. Coverslips were mounted with Molecular Probes ProLong Gold Antifade Mountant with DAPI mounting medium from ThermoFisher. This assay was done with or without treatment  with  50  ug/ml  cycloheximide  to  deplete  the  ER  of  new  protein

translation.

 

 

Cells were imaged with the Zeiss Axiovert 200M at the 100X objective as either

 

still images or using confocal imaging using the Apotome camera. Each image was taken

with about 10- 12 slices, each at .3 um thick. The images were then processedusing

 

Axiovision as a 3D rendered image or z-stack slices.

 

 

 

2.1.8    Statistical Analysis

 

All cell culture experiments were analyzed by a one-way ANOVA with either a

 

Tukey or Dunnett's post-hoc test (indicated within each experiment). Experiments with statistical analysis  were  repeated in  triplicate  giving  an  n=3,  for  most  experiments. Statistical  significance was  expressed  as  follows;   *  p<.05,  **  p<.01,   ***  p<.001,

****p<.0001.


 

2.2 Experiment I- Generation of Sitosterolemia Associated ABCG8 Cytosolic Mutants

Purpose:  To  generate ABCG8  Cytosolic  Sitosterolemia-associated  mutations to

 

characterize each mutation based on its molecular defect.  Mutation Database: A mutationdatabase was generated to determine which mutations were "clinically" associated with Sitosterolemia. The database consisted of mutations that were clinically published fromhuman  subjects,  predicted  through the   global  minor  allele  frequency   (GMAF),  or likelihood of pathogenicity on the America College of Medical Genetics and Genomics (ACMG)  scale.  Two criteria  were  taken into  consideration when  determining  which mutants would be  analyzed. One,  they  had  to  be  Sitosterolemia-Associated,  either a published mutation in a clinically confirmed Sitosterolemia patient or high on the ACMG scale. Two, we only analyzed missense mutations. While several nonsense and frameshift mutations are associated with Sitosterolemia, we limited our studies tomissense mutations. Truncated proteins, while have been described to be inactive proteins, were not in our analysis because one, they are already characterized as a class I mutant and two, there is

little to no chance to “rescue” or stabilize a truncated protein with the modulators.

 

Experimental   Design:  ABCG8   mutants   were    generated  via    Site-Directed Mutagenesis. The workflow of each mutant is as follows: primer design, PCR reaction, DpnI  Digest,  Bacterial  Transformation,   SalI Digest,  Gel  Electrophoresis,   and DNA Sequencing. In the primer design, oligos were designed using IDT and GeneArt software’s to generate an oligo that was approximately 35-39 nucleotides long. The forward andreverse primers were designed as direct complements of each other, with the point mutation of interest directly in the middle of the oligo.  The oligos were then reconstituted in TE

Buffer at 100 µM.


 

The PCR reaction was designed according to the Pfu polymerase manufacturer's  protocol and the annealing step temperature was based on the oligo melting temperature  (Annealing temp= Tm - 5°C). DpnI is a restriction enzyme that cleaves methylated DNA

produced by host cell machinery. Therefore, prior to the PCR reaction, the parent

 

ABCG8 plasmid was methylated to ensure DpnI digest provided the proper negative

 

control. Control reactions contained no Pfu polymerase and primers from one mutant.

 

Changes to the Pfu manufacture protocol were: 18 total cycles and a 10minute extension

time. Following PCR, the DNA was cleaned up using Qiagen Spin Miniprep 2.0 kit to

 

remove buffers or salts. The DNA then underwent DpnI digest based on NewEngland

 

Biolabs protocol and another round of DNA clean-up. After the second DNA clean-up,

the DNA was transformed into DH5competent cells and grown on Agar + 50 ug/ml

 

Ampicillin plates. Bacterial colonies were then grown in LB Broth with 50 µg/ml of

 

Ampicillin overnight at 37°C and 200 rpm. The colonies were prepped using the Qiagen Mini Prep kit and quantified using a Nanodrop spectrophotometer. If the colonies had a  positive yield above 160 ng/µl, they were SalI digested based on NewEngland Biolabs

protocol and ran on a 1% agarose gel, then fluorescently imaged using SYBR-Safe.

 

After confirmation that SalI cut the mutant plasmids at the equivalent cut sites to the parent ABCG8 plasmid, the plasmid was subjected to Sanger sequencing (Eurofins,

Chicago, IL). The DNA sequence was then analyzed in SnapGene 4.3.11 to confirm

 

desired base pair change. Once the desired base change was confirmed, the construct was

sent for full plasmid sequencing of the coding region to check for any additional

 

undesired mutations.


 

    
 

A)

  
 

 

 


 

Figure 2.2- 1 Example of Site-directed Mutagensis Restriction Enzyme Digest and sequence verification (Generated using Snapgene software version 4.3.11).

 

 

Fig. 2.2- 1. A. Native ABCG8 and ABCG8 mutants were digested with the SalI restriction enzyme. The samples were then ran on a 1% Agarose gel and fluorescently imaged via SYBRsafe. B. Samples were Sanger sequenced (by Eurofins) and the output was

importing to Snapgene 4.3.11. Parent hG8 native DNA compared to mutant DNA, showing a single base pair change resulting in a mutation of Leucine to Proline at amino acid position 228.

 

Table 2.5. Sitosterolemia-Associated Mutants generated by Site-Directed Mutagenesis in the Cytosolic Domain  of ABCG8

 

Mutant

Mutant

R184H

T400K

L195Q

N409I

L228P

N409D

P231T

P415H

R263Q

 

 

Table 2.5: Table of the nine cytosolic mutants in ABCG8 generated via site-directed mutagenesis


 

2.3 Experiment IIOptimization of Transient Transfection of Human Cell Lines

Purpose: ABCG5/8   are  located  on  the  apical  membrane  of hepatocytes  and

 

enterocytes, both showing difficulties in transient transfection of human genes. Different

 

cell lines were optimized to find the optimal ratio of DNA to transfection agent to express our protein of interest (ABCG5 and ABCG8). In addition to difficulties with transfection,optimization was needed to find an antibody that detected the human ABCG5/8 protein via

Western Blotting.

 

 

 

Experimental Design:  Each cell  line  was  seeded on  day  zero,  at  varying  sub-

 

confluent densities in a 6 or 12-well dish. On day one, cells were transiently transfected using the ThermoFisher Scientific Lipofectamine 3000 kit according to their protocol. Transfections were performed initially at various ratios of DNA to Lipofectamine and then performed at a standard 1.5 µg DNA:2 µl Lipofectamine, 2 µg DNA: 2 µl Lipofectamine, and  3 µg  DNA:  2 µl  Lipofectamine.  The  Lipofectamine  complexes  were  combined according to the manufacturer's protocol and incubated for 30 minutes at room temperature. Prior to adding the complexes to the cells, 200-300 µl of serum-free media was added to the cells. The complexes were added dropwise to the cells and incubated at 37°C for 1-3 hours before adding 1 ml of complete media. On day two, GFP lysates were prepared and

analyzed as previously described.

 

 

 

After standardizing the concentration of DNA and Lipofectamine that would be

 

utilized in subsequent experiments, each cell line was transfected at 2 µg DNA: 2 µl

 

Lipofectamine on day one in the following conditions: GFP, GFP+G5, GFP+G8, G5+G8.

On day two, the media was replaced and 1% Triton Lysates were made on day three.


 

Lysates were quantified by a BCA assay and Immunoblotted for protein detection of

human ABCG8.

 

 

2.4 Experiment III- Mutant Maturation Assay

Purpose: Clinically confirmed ABCG8 mutations have different molecular

 

mechanisms that cause a lack of function of ABCG5/8 to efflux sterols. In the mutant    maturation assay, we are referring to the maturation of the protein from the synthesis in

the ER to beyond the Golgi. This experiment was performed to see to what extent

 

ABCG8 mutants matured beyond the ER (Fig. 2.4- 1).


 

 

 

Figure 2.4- 1 Graphical Representation of in vitro maturation assay.

 

 

 

Figure 2.4- 1. Graphical representation of in vitro assay to monitor ABCG8 maturation beyond the ER. Observing the change in

in(el)c(e)ompet(ctroph)e(o)n(r)t(e)m(tic)utat(mo)io(bi)n(li)s(t)l(f)ass(A)I(B)G8 or ABCG5 can be used as amarker for maturation, indicative of maturation competent or

 

Experimental  Design: After  mutants  were  generated  and  sequence  verified,  allmutants were prepped with the Qiagen Midi Prep Plus kit. Two independent cell culture experiments were  conducted. In  all  experiments, HuH-7  cells  were seeded  at  a  sub-confluent density in a 6 or 12-well dish on day zero. In the first experiment, cells were transfected on day one with V, V+myctagged-G5, V+G8, myctagged-G5+G8,and native myc tagged-G5+mutant G8. Using a C-terminal myc tagged ABCG5 construct was to be able to monitor both G5 and G8 maturation, as during experimentation, we did not have avalidated human ABCG5 antibody. In the second experiment, cells were transfected on day one with V, V+G8, myc tagged-G5+G8, and V+mutant G8 alongside a GFP loadingcontrol to monitor ABCG8 mutant stability in the absence of ABCG5. For all experiments,

culture media was replaced the day following transfections, and 48 hours post-transfection,


 

 

Triton (1%) lysates were prepared. Lysates were analyzed through a BCA assay and

analyzed as previously described.

 

Subsequent experiments include SalI digest (as previously described) and RNA extraction. The RNA extraction in vitro assay resembled the V+mutant hG8 stability assay, the only difference being that rather than lysing the cells with 1% triton, they were lysed with TRIzol (Life Technologies) and proceeded to be processed using the RNeasy kit from Qiagen. The RNA was transcribed into cDNA using the iScript Reverse Transcriptase kit. The cDNA was then amplified with forward and reverse primers utilized in the site-directed

mutagenesis PCR reactions and ran on a 2% agarose gel containing .01% SYBRsafe.

 

 

 

2.5 Experiment IV- Native ABCG5/G8 Complex Compound Screening

Purpose: Based on previous experiments done in ABCC7 and ABCB4 transporters,Roscovitine along with its 11 analogs were tested to see if there was potential in increasing

the maturation of the native ABCG5/8 complex.

 

Experimental Design: HuH-7 cells were seeded in two  12-well dishes at a sub- confluent density on day zero. On day one, cells were transfected with control V, V/G5, V/G8, G5/G8, and the remaining wells with G5/G8. On day two, cells were supplemented with low serum media (DMEM + .2% BSA) and either 100 uM of Roscovitine and analogs or .1% DMSO vehicle (Vauthier, 2019). On day three, 1% tritonlysates were prepared and analyzed with a BCA assay. Lysates were Immunoblotted as previously described for

ABCG8 and Calnexin.


 

2.6 Experiment V- Roscovitine Toxicity

Purpose: Based on previously published data on Roscovitine and its known

 

toxicity, we wanted to observe further both the toxicity and impact on protein

 

concentration when HuH-7 cells were treated with Roscovitine in a dose-dependent

manner.

 

 

Experimental Design: On day zero, Huh-7 cells were seeded at a sub-confluent

 

density in 6-well dishes. The cells received treatment media on day two. Treatment media composed of 0.2% BSA with added 1% Pen-Strep and 1% GlutaMAX. The five treatment conditions were: .1% DMSO, 100 uM Roscovitine, 20 uM Roscovitine, 5 uM Roscovitine, and 1 uM Roscovitine. On day 3, media was collected and centrifuged to remove cellular debris and triton lysates were prepared. An LDH Assay was performed on the media tomeasure toxicity and a BCA Assay was performed on the lysates to measure protein

concentration.

 

 

2.7 Experiment VI- Corrector Testing of Class II Mutants

Purpose: After establishing a classification system for clinically found

 

Sitosterolemia-associated cytosolic mutants in ABCG8, FDA-approved correctors

 

(Luma-, Teza-, Elexacaftor) used in the treatment of Cystic Fibrosis were tested in vitro   to observe if heterodimerization and trafficking beyond the ER could be restored in Class

II mutants.


 

Experimental Design: Huh-7 cells were seeded in a 6-well dishes at sub-confluent

 

density on day zero. On day one cells were transfected with control GFP, GFP/G5-

 

GFP/G8- G5/G8 and the remaining wells with wildtype G5/mutant G8. On day two, cells were supplemented with low serum media (DMEM + .2% BSA) and six combinations of corrector treatments (Vehicle, Luma-, Teza-, Elexa-, Luma + Elexa, and Teza + Elexa) in

previously used concentrations68On day three, 1% tritonlysates were prepared and

 

analyzed for maturation.

 

 

 

2.8 Experiment VII- Immunofluorescence of Maturation Competent Mutants

Purpose:  Mutants  that are  maturation  competent  or  demonstrate  a  decrease  in

 

maturation  were analyzed  via  immunofluorescent  microscopy to  observe  if they  are capable of trafficking to the cell surface in the presence of cycloheximide, a known protein

synthesis inhibitor.

 

 

 

Experimental Design: Cells were seeded on coverslips, co-transfected with ABCG5-

 

myc and either mutant and wildtype ABCG8. Cells were fixed and treated cells with the KWE5 (mouse anti-human ABCG8) and then a conjugated secondary fluorophore (goat anti-mouse  488  or goat  anti-rabbit  568). Coverslips  were  mounted  with  a  mounting medium that contained DAPI to label the nuclei and imaged under blue, red, and green

fluorescence.


 

CHAPTER 3.  RESULTS

 

We investigated cytosolicmissense mutations in ABCGto determine if ABCG5/G8 dimerization and trafficking beyond the ER were compromised. After understanding the    molecular defects of the mutantswe observed the effects of proteostasis regulators on the native ABCG5/G8 complex to observe whether or not the transporter dimerization could  be enhanced. Further, we observed the effects of FDA-approved correctors (Luma-, Teza- , and Elexacaftor) in class II mutants of ABCG8. The results for each of the following are

listed below.

 

 

3.1 Experiment I- Generation of Sitosterolemia-Associated ABCG8 Cytosolic Mutants

We generated 9 cytosolic, Sitosterolemia-associated mutants in ABCG8 via site- directed mutagenesis (Fig. 3.1- 1). The mutations we generated were clinically found in   patients with a biochemical diagnosis of Sitosterolemia. The purpose of generating these

missense mutations was to observe the effects of the mutations in vitro in human

hepatocytesas seen in Experiment IIIUsing site-directed mutagenesis allowed us to make a single base pair change to introduce our desired mutation without altering the

remainder of the protein.

 

The native ABCG8 construct was used as a negative control throughout the site-

directed mutagenesis to ensure that the only bacterial growth post-transformation was

 

PCR product and not the original plasmid (DpnI digest). The native ABCG8 construct  was also used as a positive control in the SalI restriction enzyme digest to confirm that

our mutant constructs did not have significant structural changes in the DNA. This

 

process was described in the methods section.


 

519a.a

3aa

491a.a●

554a.a●

705 a.a

555 a.a

H

H

-        NH

3

Figure 3.1- 1 ABCG8 Mutation Diagram

 

61 9

 

 

 

 

 

 

 

 

Fig. 3.1- 1 Diagram of the ABCG8 transporter and Sitosterolemia mutations placed in the corresponding location on the transporter. Amino acid 619 represents the glycosylation site for ABCG8.

 

 

AdditionallyBioinformatic analysis of these nine mutations are shown in Figure

3.1-2 and 3.1-3. The protein analysis shown is both comparisons of ABCG8 across

 

species and between the ABCG Family of transporters.


 

Figure 3.1-2. ABCG8 Sequence conservation among species.

 

180 - 240  A · A .                         R184H                    L195Q

L228P  P231T

>H .   sapi ens                          SQAQRDKRVEDVIAELRLRQCADTRVGNMYVRGLSGGERRRVS

IGVQLLWNPGILI LDEP

>M.   mulatta                         SQAQRDKRVEDVIAELRLRQCADTRVGNTYVRGVSGGERRRVS

IGVQLLWNPGILI LDEP

>C .   l uPUS   familiari SQAQRDQRVDDVIAELRLRQCANTRVGNAYVRGVSGGERRRVS

IGVQLLWNPGILI LDEP

>M .   muscul US                       SQAQRDKRVEDVIAELRLRQCANTRVGNTYVRGVSGGERRRVS

IGVQLLWNPGILI LDEP

>D .   reri o                              SQKQRDQRVDDVIAELRLRQCAHTRVGNEYVRGVSGGERRRVS

IAVQLLWNPGILI LDEP

 

 


 

258 - 269  A . A ·

>H   sapi ens

>M .   mul a t ta

>c   l upus   familiaris

>M .   muscul uS

>D .  reri o


 

R263Q

LAKGNRLVL IS

LAKGNRLVL IS

LAKGNRLVLVS

LAKGNRLVL IS

LARGNRLVLLS


 

 

390 - 420  A A ·                              T400K         N409D/工  P415H

 

>H .

sapiens

AVQQFTTLIRRQ ISNDFRDLPTLLIH

>M.

mul atta

AVQQFTTLIRRQ ISNDFRDLPTLLIH

>C .

l upus   familiaris

PVQQFTMLI RRQIFNDFRDLPT LLI R

>M ·

muscul us

MIEQFSTLI RRQ ISNDFRDLPTLLIH

>D .

reri o

KVHQFTTLI RRQVFNDYRDLVT LVVH

 

 

Fig. 3.1-2. Sequence alignment using FASTA files from NCBI (Source) and Snapgene 4.3.11. Species aligned in order top to bottom are human, Rhesus monkey, Dog, Mouse, Zebrafish. Mutants in the analysis are in bold, yellow highlighted residues are conserved    while green are not conserved.

 

Figure 3.1-3. ABCG8 Sequence conservation among ABCG family members.

 

180 - 240

A A          R184             L195Q                                                                      L228P  P231T

hABCG1

KDEGRREMVKEI LTAGLL SCANTRTGS - - - - - LSGGQRKRLAIALELVNNPPVMFFDEP

hABCG2

TNHEKNERINRVI QEGLDKVADSKVGTQFIRGVSGGERKRTS IGMELITDPS ILFLDEP

hABCG4

KQEVKKELVTE ILTAGLMSCSHTRTAL - - - - - LSGGQRKRLAIALELVNNPPVMFFDEP

hABCG5

NPGSFQKKVEAVMAESLSHVADRLIGNYSLGGISTGERRRVSIAAQLLQDPKVMLFDEP

hABCG8

SQAQRDKRVEDVIAERLRQCADTRVGNMYVRGLSGGERRRVSIGVQLLWNPGILI LDEP

258 - 269  A A .       R263Q

hABCG1             LAQGGRSI ICT

>hABCG2             MSKQGRTI IFSI

hABCG4             LAQGGRTI ICT

>hABCG5               LARRNRIVVLT

>hABCG8            LAKGNRLVL IS L

 


 

390 - 420

hABCG1

hABCG2

hABCG4

hABCG5

hABCG8


 

A . A .       T400K         N409D/I  P415H

CLTQFCI LFKRTFLSIMRDSVLTHLR

FCHQLRWVSKRSFKNLLGNPQASIAQ

TLTQFCI LFKRTFLSILRDTVLTHLR

VFSKLGVLLRRVTRNLVRNKAVITR

AVQFTTLIRRQISNDFRDLPTLLIH


 

 

Fig. 3.3- 1. Sequence alignment using FASTA files from NCBI (Source) and Snapgene 4.3.11. ABCG8 Family members include 1, 2, 4, 5, and 8.  Mutants in the analysis are in bold, yellow highlighted residues are conserved while green arenot conserved.


 

3.2 Experiment IIOptimization of Transient Transfection of Human Cell Lines

For transient transfection in vitro, there are different transfection reagents

 

commercially available, both liposome and non-liposome. In a series of transfection

 

optimization experiments, we observed differences in transfection efficiency between

 

three different reagents; Lipofectamine 3000, FuGene6, and Endofectin Max in our HuH- 7 system. HuH-7 cells are a human hepatocarcinoma cell line that have been reported to

have optimal transfection efficiency as well as potential applications with polarization.

 

Ratios were determined by previously published studies done in HuH-7 cells for

 

FuGene6 and Endofectin70. GFP signal was quantified by fluorescence reading of 485 nm excitation and 515 nm emission and normalized to the A280  (arb. units). The densitometric

analysis was done in Adobe Photoshop and quantified by a ratio of Mature to Immature

 

signal and expressed as GFP signal vs. Maturation Index for the ABCG5 blot.


 

Figure 3.2- 1 Comparison of different transfection reagents in the HuH-7 cell line.

 

Fig. 3.2- 1. A. Huh-7 cells transfected with ~2 ug of DNA (1 ug hG5-myc, 1 ug hG8, 100 ng GFP) and 4 ul of transfection reagent

(Lipofectamine 3000, FuGene6, and Endofectin Max) in triplicates. Samples were analyzed by GFP fluorescent detection expressed as 515/A280 and a same day control lysate was background subtracted. Data was analyzed by a One-way ANOVA and significant

differences were determined by Tukey Post-Hoc, * p<.05, ** p<.01, *** p<.001, ****p<.0001. B. Western blot of lysates prepped to confirm transfection efficiency was synonymous with signal intensity, with C) regression analysis on the % Maturation, Maturation    Index, and Normalization to Loading Control.

 

 

 

From initial transfection optimization experiments, it was determined that at the

 

concentrations of DNA and volume of transfection reagents we testedthe HepGcells

 

had the lowest transfection efficiency while Huh-7 and HEK293 cells had comparable

 

transfection efficiencies. Because our desired experimental cell line are hepatocytes and  due to HEK293’s poor adherence and inability to tolerate compounds, these findings led

us to continue the remainder of our in vitro experiments in the Huh-7 system, with

 

transfection optimization shown below (Fig. 3.2-2). No statistical differences were

 

detected across various volumes of Lipofectamine or plasmid DNA. Previous

 

optimizations in other cell lines determined 2 ug plasmid DNA to 2 ul Lipofectamine was

the optimal ratio and therefore we selected a 1:1 ratio to stay consistent with conditions

 

used in other cell lines in the lab.


 

 

 

Figure 3.2-2 Transfection Optimization in HuH-7 cells.

Fig. 3.2-2 Transfection efficiency in the Huh-7 Cells transfected with the same concentration of DNA (2 ug of GFP) and varying

T(v)ukey(olum)s p(s o)o(f)sLt-ih(p)o(o)c(f)test(ecta)mine. Samples were ran in triplicate on a 12 well dish. Data was analyzed with a One-way ANOVA and a

 

 

 

3.3 Experiment III- Mutant Maturation Assay

 

 

The findings from the mutant trafficking assay determined that

 

approximately 44% of ABCG8 were maturation incompetent. We determined this by

 

monitoring the upper molecular weight form of ABCG8, which indicates that the protein

has been glycosylated and matured beyond the ER. Transmembrane proteins become

 

glycosylated during trafficking from the ER to the Golgi and are further modified as the

protein transits the Golgi. The bulky moiety causes a shift in molecular weight, an

 

indicator of protein maturation. We monitored ABCG8 mutant maturation in 3 ways:

 

tracking ABCGmaturationtracking ABCGmaturationand the stability of the

 

ABCG8 mutants in the absence of their partner. The densitometric analysis was

 

completed by using the measurement feature in Adobe Photoshop and using the

 

integrated density as a measurement for signal intensity. In this particular assay, we

 

expressed the signals as a percent (mature signal/total signal) and as an index (immature


 

signal/mature signal) using the ABCG5 blots. Due to the appearance of a high upper

molecular weight band (*), the ABCG8 blot was not included in the densiometric

 

analysis.

 

Figure 3.3- 1.  Protein maturation bioassay demonstrating ABCG8 cytosolic mutants co- transfected with ABCG5-myc.

 

 

Figure 3.3- 1. Huh-7 cells transfected with Controls and native hG5-myc/mutant hG8. A) Western blot depicts in vitro trafficking

assay, with B) Densiometric analysis for % maturation (Mature Signal/Total Signal) and Maturation index (Mature Signal/Immature    Signal). The % maturation was internally normalized to GFP. Data (n=3) was analyzed by a One-way ANOVA with a Dunnett’s Post- hoc test. (*) p<.05, (**) p< .01, p<.001 (***), p<.0001 (****).   C) Normalization to loading control of the one experiment shown

(n=1).

 

From this maturation assay, it was apparent that in the mutants with a statistically significant reduction in maturation in the ABCG5 blots, a signal was not detected in the

ABCG8 blot. Additionally, we investigated the stability of mutants in ABCG8 by

 

expressing the mutants in HuH-7 cells in the absence of ABCG5. The blots (shown

 

below) demonstrated the same pattern of apparent Class II mutants lacking the ABCG8

immature signal.


 

Figure 3.3-2. Protein stability Western blots of mutants in ABCG8.

 

 

Fig. 3.3-2 Huh-7 cells transfected with vector and mutant hG8 in the absence of hG5. Western blot depicts in vitro stability of the monomer compared to native hG8 and native ABCG5/G8.

 

We then asked two questions; did our DNA plasmid preps contain DNA at the   concentration the spectrophotometer was giving out? Furthermore, was the DNA being  transcribed into RNA? We knew the protein was not expressed due to the lack of signal in the ABCG8 blots. The first experiment to test these was a restriction enzyme digest

using SalI to confirm we had DNA and to verify there were no significant structural

 

changes compared to wildtype ABCG8.


 

Figure 3.3-3Restriction enzyme digest on Class II mutants.

 

Figure 3.3-3. Restriction enzyme digest of wildtype ABCG8 and mutant ABCG8. Samples were digested using SalI on a thermocycler block and analyzed on a 1% agarose gel and fluorescently imaged via SYBRsafe.

 

After the restriction enzyme digest and gel electrophoresis, we concluded

that while signal intensity did vary mutant to mutant, there was in fact, DNA in our

 

plasmid prep. We seeded HuH-7 cells at a sub-confluent density on day zero, day one

 

transfected cells with vector and either hG8 or Class II mutant hG8, and treated cells with Trizol on day two. We extracted RNA using the Qiagen RNA extraction kit. After RNA

extraction, cDNA was generated using reverse transcriptase and the amplicons were

 

analyzed on a 2% agarose gel (Fig. 3.3-4).


 

 

 

 

Figure 3.3-4.  Gel electrophoresis of cDNA from HuH-7 lysates

 

Figure 3.3-4. cDNA of wildtype ABCG8 and mutant ABCG8 amplified and analyzed on a 2% agarose gel and fluorescently imaged via SYBRsafe. Samples were lysed from HuH-7 cells, extracted RNA, transcribed cDNA, and amplified using SYBRgreen.

 

3.4 Experiment IV-Corrector Testing of Class II Mutants

 

 

In testing the CFTR correctors on the Class II maturation deficient mutants, we

determined in the conditions we tested, that the CFTR modulators were unable to

 

"correct" maturation deficient mutants in the cytosolic domain of ABCG8. Figure 15 is

an example of the full screen for each mutant (R263Q) and the two dual therapies

 

(Lumacaftor and Elexacaftor or Tezacaftor and Elexacaftor) on all maturation

incompetent or maturation comprised mutants.


 

    
  

 


 

Figure 3.4- 1 Protein maturation bioassay with treatment with published concentrations of CFTR correctors68.

 


 

A)

 

 

 

 

 

 

 

 

 

 

 

C)

 

 

 

 

 

 

 

 

 

 


 

B)

 

 

 

 

 

 

 

 

 

 

 

D)


 

 

Fig. 3.4- 1. Huh-7 cells transfected with native hG5-myc/mutant hG8 with treatment of CFTR correctors at published concentrations. A) Western blot depicts in vitro trafficking assay in HuH-7 cells, with B) Densiometric analysis for % maturation (Mature

Signal/Total Signal), C) Maturation index (Immature/Mature Signal), and D) Normalization to loading control (Mature Siganl/B - actin).

 

3.5 Experiment V- Testing Regulators of Proteostasis on Native ABCG58

Roscovitine and the 11 analogs synthesized were tested on both HuH-7 (Fig. 16)

and HepG2 cells (Fig. 17). The densitometric analysis demonstrates in both cell lines

 

these 12 treatments at 100 uM, specifically on the MRT2-237-245 compounds, an

 

increase in maturation compared to the native complex. Throughout testing these

 

compounds at a 100 uM concentration, patterns of low protein yield in cell lysates as well

as acidification of the media (yellow) media, suggesting cell lysis. We proceeded to run


 

an LDH activity assay to determine a concentration where Roscovitine, the most

 

observed toxic compound, had a reduced level of LDH activity (see results in Experiment

VI).

 

The results of experiment VI led to reducing the regulator of proteostasis screen  from 100 uM to 20 uM in an attempt to retain the cell morphology during culturing. The

results of the 20 uM experiments are ongoing.

 

Figure 3.5- 1. Roscovitine Screening on native ABCG5/G8 complex in HuH-7 cells at 100 uM.

 


 

 

 

 

A)

 

 

 

 

 

 

 

 

 

C)


 

 

 

 

 

Maturation Index

(Mature Signal/Immature Signal)

 

140

120

100

5

4

3

2

1

0

Treatment Group


 

 

 

 

B)

 

 

 

 

 

 

 

 

 

 

D)

Normalization of Mature Signal/Loading Control

 

Mature Signal/Loading Contro

(arb.)

4

3

2

1

0

Treatment Group


 

 

 

 

 

Fig. 3-5.1. HuH-7 cells transfected with native hG5-myc/hG8 with treatment of Roscovitine and 11 analogs at 100 uM. A) Western

blot depicts in vitro trafficking assay in HuH-7 cells, with B) Densiometric analysis for % maturation (Mature Signal/Total Signal), C) Maturation Index (Mature Signal/Immature Signal), and D) Normalization to the loading control (Mature Signal/Calnexin). The

following blot does not have an internal GFP standardization or statistical analysis (n=1).


 

A)

Figure 3.5-2. Roscovitine Screening on native ABCG5/G8 complex in HepG2 cells at100 uM.

 

 

 

 

 

C)

 

 

 

 

 

 

 

 

 

Fig. 3.5-2 A. Western blot depicts in vitro trafficking assay in HepG2 cells when treated with 100 uM Roscovitine and analogs. B.

Densiometric analysis for % maturation (Mature Signal/Total Signal) and C. normalization to the loading control (Mature Signal/Calnexin). n=1.

 

3.6 Experiment IV- Roscovitine Dose-Response

From previously published experiments and based on difficulties retaining a high

protein concentration in lysates from Roscovitine, we wanted to observe the toxicity of

 

Roscovitine both in LDH activity as well as its impact on protein concentrations. Using a LDH activity assay, we measured both LDH activity and toxicity of Roscovitine at four   doses. (Fig. 18). Based on these experiments, we elected to proceed with a concentration

of 20 uM in the HuH-7 system due to the reduced toxicity and LDH activity as well as

 

increased protein concentration.


 

Figure 3.6- 1 Dose-Response data on Roscovitine in HuH-7 cells.

 

Fig. 3.6- 1 A. HuH-7 cells were treated in a dose-dependent manner with Roscovitine, a compound that has shown toxicity in multiple cell lines in our hands as well as published by other research groups. The data was expressed as LDH Activity (A480-A690) vs.

concentration of compound. B. Toxicity of Roscovitine calculated with max LDH activity and negative control of vehicle treated cells. LDH Activity was analyzed by a One Way ANOVA and with a Dunnett’s Post-hoc test (n=3).

 

3.7 Experiment VI- Immunofluorescence Trafficking Assay

As described in the methods section, HuH-7 cells were processed for

 

immunofluorescence microscopy. Initially, we tested the native ABCG5/G8 complex in a cycloheximide time course experiment. We treated cells with 50 ug/ml cycloheximide for

1 hour, 2 hours, 4 hours, 8 hours, and overnight as well as anon-treated well (Fig. 3.7- 1).


 

 

 

Figure 3.7- 1.  Immunofluorescence and images from the cycloheximide time course experiment.

 

 

Fig. 3.7- 1 HuH-7 cells were transfected with native G5G8. Cells were then treated at different time points with 50 ug/ml

cycloheximide. The two images shown are representative images of our negative control, without cycloheximide treatment, and the time point we continued experimentation with, 8 hours. Cells were stained for ABCG8 (KWE5 lot- 1), AlexaFluor-488. Images are  slices taken at 100X with Zeiss Axiovert 200Musing the Apotome camera for confocal microscopy.

 

Additionally, we did a time-course using 50 ug/ml and 100 ug/ml of

cycloheximide to monitor ER depletion and analyzed with SDS-PAGE and

 

Immunoblotting as previously described (Fig. 3.7-2).


 

Figure 3.7-2. Time-course with 50 and 100 ug/ml CHX.

 

Fig. 3.7-2. HuH-7 cells were transfected with native G5G8. Cells were then treated at different time points with 50 ug/ml OR 100

ug/ml cycloheximide. Cell lysates were analyzed by SDS-PAGE and immunoblotted for ABCG5 with a c-myc antibody and calnexin loading control.

 

After these cycloheximide time-course experiments, we concluded that 50 ug/ml

of cycloheximide at 8 hours was the optimal dose and time at the conditions that we

 

tested.  We then transfected HuH-7 cells with the maturation competent ABCG8 mutants

to investigate whether they were capable of localizing to the plasma membrane or not.


 

 

 

Figure 3.7-3 Immunofluorescence images of ABCG8 mutants.

 

 

Fig. 3.7-3. HuH-7 cells were transfected with G8 control, native G5G8, or mutant G8 co-transfected with wildtype G5. Cells were   then treated 50 ug/ml cycloheximide for 8 hours. Cells were stained for ABCG8 (KWE5 lot- 1) and AlexaFluor-488, E-cadherin and AlexaFluor-568, and DAPI. Images are slices taken at 100X with Zeiss Axiovert 200Musing the Apotome camera for confocal

microscopy.

 

The results from the analysis of the Immunofluorescence microscopy was mutants

R184H and T400K exhibited a trafficking pattern that resembled the wildtype complex

 

while N409D, N409I, and P415H appeared to localize to some subcellular compartment


 

that was distinct from the wildtype. Representative images are shown in Figure 3.7-3 of

 

one mutant that had apparent trafficking or trafficking to another subcellular

compartment.


 

CHAPTER 4. DISCUSSION

 

 

 

 

Our protein of interest is endogenously expressed at high levels in the liver

 

(hepatocytes) and small intestine (enterocytes). Both of these cell types have known

 

difficulties for in vitro assays, particularly that they are not easily transfected. Because of the nature of the project, we needed to find a cell line that could be efficiently transfected

and had a translational relationship. Initially, we began our analysis in HepG2 cells.

 

These are another human hepatocyte cell line, and while they can be polarized, their

 

ability to transfect our gene of interest was sub-optimal. We then moved into a well-

 

established cell line successful in transient transfections, HEK293 cells. While these cells

were not our desired model system, they could express our proteins in high abundance

 

and were easily detected via immunoblotting. However, because HEK293 cells are a

 

loosely adherent cell type, they could not withstand compound testing when treated at

 

100 uM of the Roscovitine screening. This finally led us to our working system of HuH-7

cells. HuH-7 cells are a human hepatocarcinoma cell line that has been described as a

 

viable substitute to primary hepatocytes. Their transfection efficiency exceeds HepG2, comparable to the HEK293 cells, and could tolerate compound testing. The cons to the

HuH-7 system was that in our hands, did not appear to polarize.

 

ABCG5 and ABCG8 are known to be the two genes to cause Sitosterolemia30. In

these experimentswe have begun a characterization of clinically published

 

Sitosterolemia-associated mutants in the cytosolic domain of ABCG8. Additionally, we

investigated potential therapies that could not only treat Sitosterolemia patients but also


 

open a door for several other ABC transporter-caused diseases. Experiments in this thesis led to the classification of 9 cytosolic mutants in ABCG8. In our in vitro experiments, we were able to classify the mutants into three classes; Class II, Class V, or unclassified. In

vivo or in vitro (polarized cells) experimentation would be required to classify any

 

unclassified mutants further. After the mutants were classified, Class II mutants were further investigated for the effects after treatment with CFTR correctors to observe if

these mutants could be stabilized and traffic beyond the ER and thus "corrected."

 

Regulators of proteostasis were tested on the native ABCG5/8 complex to observe if there could be enhancement to an already poor-folding protein complex. In our hands,    the Roscovitine analogs demonstrated modest increases in maturation, but the interesting

story is the increased mature signal compared to the native complex. While one could

 

argue that this could be due to changes in transfection efficiency, our GFP signal was

 

utilized as an internal measure for differences in transfection across the wells. Because

the mechanism of action of these analogs are thought to be inhibition of the proteasome,

specifically with the Class II mutants that appear to be rapidly degraded, we can

 

hypothesize that these compounds could provide some benefit to both native ABCG5/G8

as well as Class II maturation deficient mutants of ABCG8.

 

During our western blotting antibody troubleshooting, which will be further

 

discussed in the limitations, we were monitoring G8 maturation through the c-myc tag on ABCG5. We probed the blots for ABCG8 with a KWE5 subclone of IB10A5 and noticed

an ABCG8-specific banding pattern. A consistent pattern of a high molecular weight

 

band (>100 kDa) appeared in the G8 only lane and for the majority resolved in the co-

transfected lane but at a reduced signal. This phenomenon became apparent to us when


 

using the HuH-7 cells, as previously in the HepG2 or HEK293 cells we were having

 

either low transfection efficiency or lower protein concentrations loaded on our SDS-

 

PAGE gels (respectively). Additionally, this higher molecular weight banding pattern

 

skewed the densitometric analysis due to the appearance of unresolved protein. The

 

ABCG5 blots did not exhibit this high molecular band as seen in the ABCG8 blots. This

is particularly interesting to study further as it could reveal a regulation of ABCG8

 

independent of ABCG5.

 

Approximately 44% of the mutants tested are maturation incompetent (Class II);

L195Q, L228P, P231T, andR263Q. In addition to these four mutants, two exhibited a

 

reduced level of maturation compared to the parent, R184H and N409I. None of the

 

mutants that did not mature beyond the ER could be rescued with the CFTR modulators

at this time. Of the remaining cytosolic mutants, we could not determine if any could

 

localize to the plasma membrane with the tools we utilized. However, three of the

 

maturation competent mutants, N409D, N409I, and P415H, appeared to localize to a

 

subcellular vesicular compartment.  The identity of this compartment will require further

studies.


 

 

 

 

Table 4.6. Updated Sitosterolemia Classification system for mutants52.

 

CLASS

DESCRIPTION

ABCG8 MUTANTS

I

Nonsense, Frameshift, Deletion

58 known or predicted

II

Maturation

L195Q, L228P, P231T, R263Q

III

Activity

 

IV

Stability

 

V

Trafficking

N409D, N409I, P415H

Unclassified

Inconclusive Results

R184H, T400K

 

Table 4.6. Updated Sitosterolemia classification system for mutants analyzed in this thesis.

 

 

 

4.1 Limitations

In our experimentation, there were a few limitations to our research. The first

 

major limitation was that there are no commercial antibodies that recognize human

 

ABCG5 and virtually only one antibody that is available commercially to recognize

 

human ABCG8. For this reason, most of the blots were done with co-transfected human

ABCG8 with a C-terminus myc-tagged human ABCG5 construct and immunoblotted

 

with a c-myc antibody. However, because the myc tag is on the C-terminus and the

 

glycosylation sites sit on the third extracellular loop of G5, we have no reason to believe

this tag had any influence on our maturation bioassay. Towards the end of the thesis

 

project, the lab had successfully grown and cultured the 1B10A5 hybridoma, which was

producing a suitable amount of antibody to conclude the remainder of the

 

immunoblotting and immunofluorescence experiments.


 

Another limitation we had for these experiments was in the generation of the

 

cytosolic mutants in ABCG8. Because these are clinically published mutations, we had

 

no control over the surrounding DNA when designing our oligos for the PCR reaction.

 

The only components that were in our control were the annealing temperature, the size of

the oligo, and the placement of the oligos with respect to where the point mutation

 

occurred. For this reason, some mutants were more difficult than others based on the

flanking DNA sequence.

 

 

 

4.2 Future Directions

In the HuH-7 system, we hypothesize that the likely source of this high molecular

banding pattern observed in ABCG8 could result from a post-transcriptional

 

modification, such as Ubiquitination, SUMOylation, or even rapid lysosomal

 

degradation. Ongoing experiments to explore this upper molecular band include cell

 

treatment with Chloroquine, a known lysosomal inhibitor. Ubiquitination and

 

SUMOylation will be explored by Immunoprecipitation experiments to see if either Ub or

SUMO will be pulled down alongside ABCG8.

 

Currently, the two Sitosterolemia treatment approaches still have experimentation

to be completed to determine whether or not they could restore ABCG5 and ABCG8

 

function. At this time we do not have a positive control for CFTR and the CFTR delF508 mutant system or the ABCB4 mutants in hand to use as a positive control for Roscovitine

and the analogs which is a current ongoing experiment. While Roscovitine was first

 

tested in the F508del mutant, the analogs were tested in mutants of ABCB4. Having that

as a positive control to continue to test the Class II ABCG8 mutants with these


 

compounds would be necessary as the previously published data was in HepG2 and

 

HEK293 cells. Testing a handful of the analogs that show promise in enhancing the

 

native ABCG5/8 complex on the Class II mutants would be the next direction in finding a

treatment option for Sitosterolemia.

 

Additionally, the proteostatic regulator impact on the native ABCG5/G8 complex

is intriguing, independent of Sitosterolemia. We can hypothesize that the enhancement

 

seen in the screen of Roscovitine and analogs would lead to an increase in protein in the

tissue in vivo. Additional experiments that would follow these results would include

 

dose-response and time course experiments in vitro, in vivo administration of the

compounds, and pharmacokinetic/pharmacodynamics experiments.

 

The future directions for these mutants are to further classify them beyond

 

maturation and trafficking to the cell surface. First, we want to take a more quantitative

 

approach to distinguish whether mutants can traffic to the plasma membrane in vitro. One

planned assay is to biotinylate G8 to calculate the percentage of G8 on the cell surface.

 

The following steps would be for in vivo experimentation to observe the activity of these

mutants and in vitro polarization experiments would be required to study if these

 

mutants’ traffic to the apical membrane.


 

REFERENCES

 

1.   Hubacek, J. A., Berge, K. E., Cohen, J. C., & Hobbs, H. H. (2001). Mutations in ATP-cassette binding proteins G5 (ABCG5) and G8 (ABCG8) causing

sitosterolemiaHuman mutation, 18(4), 359-360.

https://doi.org/10.1002/humu.1206

2.   Zein, A. A., Kaur, R., Hussein, T., Graf, G. A., & Lee, J. Y. (2019). ABCG5/G8: a structural view to pathophysiology of the hepatobiliary cholesterol

secretion. Biochemical Society transactions, 47(5), 1259- 1268.

https://doi.org/10.1042/BST20190130

3.   Ikeda, S., Mochizuki, A., Sarker A.H., Seki, S. Identification of Functional

Elements in the Bidirectional Promoter of the Mouse Nthland Tsc2

Genes,Biochemical and Biophysical Research Communications,Volume 273,

Issue 3, 2000, Pages 1063- 1068, ISSN 0006-291X,

https://doi.org/10.1006/bbrc.2000.3071

4.   Bhattacharyya AK, Connor WE. Beta-sitosterolemia and xanthomatosis. A newly described lipid storage disease in two sisters. J Clin Invest. 1974 Apr;53(4):1033- 43. doi: 10.1172/JCI107640. PMID: 4360855; PMCID: PMC333088.

5.   Wang, Z., Cao, L., Su, Y., Wang, G., Wang, R., Yu, Z., Bai, X., & Ruan, C. (2014). Specific macrothrombocytopenia/hemolytic anemia associated with

sitosterolemiaAmerican journal of hematology, 89(3), 320-324.

https://doi.org/10.1002/ajh.23619

6.   Higgins, C., Linton, K. The ATP switch model for ABC transporters. Nat Struct Mol Biol 11, 918-926 (2004).https://doi.org/10.1038/nsmb836

7.   Rees, D., Johnson, E. & Lewinson, O. ABC transporters: the power to

changeNat Rev Mol Cell Biol 10, 218-227 (2009).

https://doi.org/10.1038/nrm2646

8.   Musso, G., Gambino, R., & Cassader, M. (2013). Cholesterol metabolism and the pathogenesis of non-alcoholic steatohepatitis. Progress in lipid research, 52(1),    175- 191.https://doi.org/10.1016/j.plipres.2012.11.002

9.   Brown, M. S., & Goldstein, J. L. (1997). The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription

factor. Cell, 89(3), 331-340.https://doi.org/10.1016/s0092-8674(00)80213-5

10. Linton, M. F., Yancey, P. G., Davies, S. S., Jerome, W. G., Linton, E. F., Song, W. L., Doran, A. C., & Vickers, K. C. (2019). The Role of Lipids and

Lipoproteins in Atherosclerosis. In K. R. Feingold (Eds.) et. al., Endotext. MDText.com, Inc.

11. McDaniel, A. L., Alger, H. M., Sawyer, J. K., Kelley, K. L., Kock, N. D., Brown, J. M., Temel, R. E., & Rudel, L. L. (2013). Phytosterol feeding causes toxicity in  ABCG5/G8 knockout mice. The American journal of pathology, 182(4), 1131-

1138.https://doi.org/10.1016/j.ajpath.2012.12.014

12. Izar, M. C., Tegani, D. M., Kasmas, S. H., & Fonseca, F. A. (2011). Phytosterols

and phytosterolemia: gene-diet interactions. Genes & nutrition, 6(1), 17-26.

https://doi.org/10.1007/s12263-010-0182-x


 

13. Cohn, J. S., Kamili, A., Wat, E., Chung, R. W., & Tandy, S. (2010). Reduction in intestinal cholesterol absorption by various food components: mechanisms and

implications. Atherosclerosis. Supplements, 11(1), 45–48.

https://doi.org/10.1016/j.atherosclerosissup.2010.04.004

14. Sugano, M., Morioka, H., & Ikeda, I. (1977). A comparison of

hypocholesterolemic activity of beta-sitosterol and beta-sitostanolin rats. The

Journal of nutrition, 107(11), 2011–2019.https://doi.org/10.1093/jn/107.11.2011

15. Brufau G, Kuipers F, Lin Y, Trautwein EA, Groen AK. A reappraisal of the

mechanism by which plant sterols promote neutral sterol loss in mice. PLoS One. 2011;6(6):e21576. doi: 10.1371/journal.pone.0021576. Epub 2011 Jun 30. PMID: 21738715; PMCID: PMC3128081.

16. Wittenburg H, Carey MC. Biliary cholesterol secretion by the twinned sterol half- transporters ABCG5 and ABCG8. J Clin Invest. 2002 Sep;110(5):605-9. doi:

10.1172/JCI16548. PMID: 12208859; PMCID: PMC151117.

17. Kangmo Lu, Mi-Hye Lee, Shailendra B Patel, Dietary cholesterol absorption;

more than just bile, Trends in Endocrinology & Metabolism, Volume 12, Issue 7, 2001, Pages 314-320, ISSN 1043-2760,https://doi.org/10.1016/S1043-

2760(01)00433-7.

(https://www.sciencedirect.com/science/article/pii/S1043276001004337)

18. Davis, H. R., Jr, Zhu, L. J., Hoos, L. M., Tetzloff, G., Maguire, M., Liu, J., Yao, X., Iyer, S. P., Lam, M. H., Lund, E. G., Detmers, P. A., Graziano, M. P., &

Altmann,S. W. (2004). Niemann-Pick C1 Like 1 (NPC1L1) is the intestinal

phytosterol and cholesterol transporter and a key modulator of whole-body

cholesterol homeostasis. The Journal of biological chemistry, 279(32), 33586– 33592.https://doi.org/10.1074/jbc.M405817200

19. Altmann, S. W., Davis,Harry R.,,Jr, Li-ji, Z., Yao, X., & al, e. (2004). Niemann- Pick C1 Like 1 Protein Is Critical for Intestinal Cholesterol

Absorption. Science, 303(5661), 1201-4.

http://ezproxy.uky.edu/login?url=https://www.proquest.com/scholarly-        journals/niemann-pick-c1-like-1-protein-is-critical/docview/213596845/se- 2?accountid=11836

20. Nguyen TM, Sawyer JK, Kelley KL, Davis MA, Rudel LL. Cholesterol

esterification by ACAT2 is essential for efficient intestinal cholesterol absorption: evidence from thoracic lymph duct cannulation. J Lipid Res. 2012 Jan;53(1):95-    104. doi: 10.1194/jlr.M018820. Epub 2011 Nov 1. PMID: 22045928; PMCID:

PMC3243485.

21. Glomset J. A. (1968). The plasma lecithins:cholesterol acyltransferasereaction. Journal of lipid research, 9(2), 155– 167.

22. J.A. Glomset, in: G.J. Nelson (Ed.), Plasma Lipids and Lipoproteins, Wiley InterscienceNew York, 1972,pp. 745–787.

23. Krieger M. Charting the fate of the “good cholesterol”: identification and    characterization of the high-density lipoprotein receptor SR-BI. Annu. Rev. Biochem. 68, 523–558 (1999).

24. Gerloff, T., Stieger, B., Hagenbuch, B., Madon, J., Landmann, L., Roth, J.,

Hofmann, A. F., & Meier, P. J. (1998). The sister of P-glycoprotein represents the


 

canalicular bile salt export pump of mammalian liver. The Journal of biological chemistry, 273(16), 10046- 10050.

25. Bosner MS, Lange LG, Stenson WF, Ostlund RE. Percent cholesterol absorption in normal women and men quantified with dual stable isotopic tracers and

negative ion mass spectrometryJ Lipid Res. 1999 Feb;40(2):302-8.

26. Dawson PA. Role of the intestinal bile acid transporters in bile acid and drug dispositionHandbExpPharmacol2011;201:169-203.

27. van der Velde, A. E., Vrins, C. L., van den Oever, K., Seemann, I., Oude Elferink, R. P., van Eck, M., Kuipers, F., & Groen, A. K. (2008). Regulation of direct

transintestinal cholesterol excretion in mice. American journal of physiology.

Gastrointestinal and liver physiology, 295(1), G203-G208.

https://doi.org/10.1152/ajpgi.90231.2008

28. Grefhorst, A., Verkade, H. J., & Groen, A. K. (2019). The TICE Pathway:

Mechanisms and Lipid-Lowering TherapiesMethodist DeBakey cardiovascular journal, 15(1), 70-76.https://doi.org/10.14797/mdcj-15-1-70

29. van der Velde, A. E., Vrins, C. L., van den Oever, K., Kunne, C., Oude Elferink, R. P., Kuipers, F., & Groen, A. K. (2007). Direct intestinal cholesterol secretion  contributes significantly to total fecal neutral sterol excretion in

mice. Gastroenterology, 133(3), 967-975.

https://doi.org/10.1053/j.gastro.2007.06.019

30. Berge, K. E., Tian, H., Graf, G. A., Yu, L., Grishin, N. V., Schultz, J.,

Kwiterovich, P., Shan, B., Barnes, R., & Hobbs, H. H. (2000). Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC

transporters. Science (New York, N.Y.), 290(5497), 1771-1775.

https://doi.org/10.1126/science.290.5497.1771

31. Graf, G. A., Yu, L., Li, W. P., Gerard, R., Tuma, P. L., Cohen, J. C., & Hobbs, H. H. (2003). ABCG5 and ABCG8 are obligate heterodimers for protein trafficking   and biliary cholesterol excretion. The Journal of biological chemistry, 278(48),

48275-48282.https://doi.org/10.1074/jbc.M310223200

32. Tsubakio-Yamamoto, K., Nishida, M., Nakagawa-Toyama, Y., Masuda, D.,

Ohama, T., & Yamashita, S. (2010). Current therapy for patients with

sitosterolemia--effect of ezetimibe on plant sterol metabolism. Journal of

atherosclerosis and thrombosis, 17(9), 891-900.https://doi.org/10.5551/jat.4614

33. Yoo E. G. (2016). Sitosterolemia: a review and update of pathophysiology,

clinical spectrumdiagnosisand managementAnnals of pediatric endocrinology metabolism, 21(1), 7- 14.https://doi.org/10.6065/apem.2016.21.1.7

34. Veit, L., Allegri Machado, G., Bürer, C., Speer, O., & Häberle, J. (2019).

Sitosterolemia- 10 years observation in two sisters. JIMD reports, 48(1), 4- 10.

https://doi.org/10.1002/jmd2.12038

35. Hong C, Tontonoz P. 2014. Liver X receptors in lipid metabolism: opportunities

for drug discovery. Nat Rev Drug Discov 13:433-444. doi: 10.1038/nrd4280.

36. Chen W, Chen G, Head DL, Mangelsdorf DJ, Russell DW. Enzymatic reduction of oxysterols impairs LXR signaling in cultured cells and the livers of mice. CellMetab. 2007 Jan;5(1):73-9. doi: 10.1016/j.cmet.2006.11.012. PMID: 17189208;  PMCID: PMC3080013


 

37. Repa, J. J., Berge, K. E., Pomajzl, C., Richardson, J. A., Hobbs, H., &

Mangelsdorf, D. J. (2002). Regulation of ATP-binding cassette sterol transporters ABCG5 and ABCG8 by the liver X receptors alpha and beta. The Journal of

biological chemistry, 277(21), 18793-18800.

https://doi.org/10.1074/jbc.M109927200

38. Freeman, L.A., et al., The orphan nuclear receptor LRH- 1 activates the ABCG5/ABCG8 intergenic promoter. J Lipid Res, 2004.

39. Sumi, K., et al., Cooperative Interaction between Hepatocyte Nuclear Factor

4{alpha} and GATA Transcription Factors Regulates ATP-Binding Cassette

Sterol Transporters ABCG5 and ABCG8. Mol Cell Biol, 2007. 27(12): p. 4248-60.

40. Graf, G. A., Li, W. P., Gerard, R. D., Gelissen, I., White, A., Cohen, J. C., &

Hobbs, H. H. (2002). Coexpression of ATP-binding cassette proteins ABCG5 and ABCG8 permits their transport to the apical surface. The Journal of clinical

investigation, 110(5), 659-669.https://doi.org/10.1172/JCI16000

41. Wang, J., Mitsche, M. A., Lütjohann, D., Cohen, J. C., Xie, X. S., & Hobbs, H. H. (2015). Relative roles of ABCG5/ABCG8 in liver and intestine. Journal of lipid

research, 56(2), 319-330.https://doi.org/10.1194/jlr.M054544

42. Moitra, K., & Dean, M. (2011). Evolution of ABC transporters by gene

duplication and their role in human diseaseBiological chemistry, 392(1-2), 29- 37.https://doi.org/10.1515/BC.2011.006

43. Locher K. P. (2009). Review. Structure and mechanism of ATP-binding cassette  transporters. Philosophical transactions of the Royal Society of London. Series B, Biological sciences, 364(1514), 239-245.https://doi.org/10.1098/rstb.2008.0125

44. Procko, E., O'Mara, M. L., Bennett, W. F., Tieleman, D. P., & Gaudet, R. (2009). The mechanism of ABC transportersgeneral lessons from structural and

functional studies of an antigenic peptide transporterFASEB journalofficial

publication of the Federation of American Societies for Experimental Biology, 23(5), 1287-1302.https://doi.org/10.1096/fj.08-121855

45. Xiong, J., Feng, J., Yuan, D. et al. Tracing the structural evolution of eukaryotic

ATP binding cassette transporter superfamily. Sci Rep 5, 16724 (2015).

https://doi.org/10.1038/srep16724

46. Online Mendelian Inheritance in Man, OMIM® . McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University (Baltimore, MD), {Apr. 6, 2022}. World Wide Web URL:https://omim.org/

47. Velamakanni, S., Wei, S. L., Janvilisri, T., & van Veen, H. W. (2007). ABCG transporters: structure, substrate specificities and physiological roles : a brief

overviewJournal of bioenergetics and biomembranes, 39(5-6), 465-471.

https://doi.org/10.1007/s10863-007-9122-x

48. Cserepes, J., Szentpétery, Z., Seres, L., Ozvegy-Laczka, C., Langmann, T.,

Schmitz, G., Glavinas, H., Klein, I., Homolya, L., Váradi, A., Sarkadi, B., &

Elkind, N. B. (2004). Functional expression and characterization of the human  ABCG1 and ABCG4 proteins: indications for heterodimerization. Biochemical

and biophysical research communications, 320(3), 860-867.

https://doi.org/10.1016/j.bbrc.2004.06.037


 

49. Lee, JY., Kinch, L., Borek, D. et al. Crystal structure of the human sterol

transporter ABCG5/ABCG8. Nature 533, 561-564 (2016).

https://doi.org/10.1038/nature17666

50. Welsh, M. J., & Smith, A. E. (1993). Molecular mechanisms of CFTR chloride

channel dysfunction in cystic fibrosis. Cell, 73(7), 1251-1254.

https://doi.org/10.1016/0092-8674(93)90353-r

51. Delaunay, J. L., Durand-Schneider, A. M., Dossier, C., Falguières, T., Gautherot, J., Davit-Spraul, A., Aït-Slimane, T., Housset, C., Jacquemin, E., & Maurice, M.  (2016). A functional classification of ABCB4 variations causing progressive

familial intrahepatic cholestasis type 3. Hepatology (Baltimore, Md.), 63(5), 1620- 1631.https://doi.org/10.1002/hep.28300

52. Williams K, Segard A, Graf GA. Sitosterolemia: Twenty Years of Discovery of the Function of ABCG5ABCG8. International Journal of Molecular

Sciences, 2021; 22(5): 2641.

53. Balch, W. E., Morimoto, R. I., Dillin, A., & Kelly, J. W. (2008). Adapting

Proteostasis for Disease InterventionScience, 319(5865), 916-919.

http://www.jstor.org/stable/20053369

54. Powers, E. T., Morimoto, R. I., Dillin, A., Kelly, J. W., & Balch, W. E. (2009).

Biological and chemical approaches to diseases of proteostasis deficiency. Annual

review of biochemistry, 78, 959-991.

https://doi.org/10.1146/annurev.biochem.052308.114844

55. Rao, R. V., & Bredesen, D. E. (2004). Misfolded proteins, endoplasmic reticulum

stress and neurodegeneration. Current opinion in cell biology, 16(6), 653-662.

https://doi.org/10.1016/j.ceb.2004.09.012

56. Bernales, S., Papa, F. R., & Walter, P. (2006). Intracellular signaling by the

unfolded protein response. Annual review of cell and developmental biology, 22, 487-508.https://doi.org/10.1146/annurev.cellbio.21.122303.120200

57. McCracken, A. A., & Brodsky, J. L. (1996). Assembly of ER-associated protein degradation in vitro: dependence on cytosol, calnexin, and ATP. The Journal of  cell biology, 132(3), 291-298.https://doi.org/10.1083/jcb.132.3.291

58. Vembar, S., Brodsky, J. One step at a time: endoplasmic reticulum-associated

degradationNat Rev Mol Cell Biol 9, 944-957 (2008).

https://doi.org/10.1038/nrm2546

59. Taylor, S. L., Kinchington, P. R., Brooks, A., & Moffat, J. F. (2004). Roscovitine, a cyclin-dependent kinase inhibitor, prevents replication of varicella-zoster

virus. Journal of virology, 78(6), 2853-2862.

https://doi.org/10.1128/jvi.78.6.2853-2862.2004

60. Norez, C., Vandebrouck, C., Bertrand, J., Noel, S., Durieu, E., Oumata, N.,

Galons, H., Antigny, F., Chatelier, A., Bois, P., Meijer, L., & Becq, F. (2014). Roscovitine is a proteostasis regulator that corrects the trafficking defect of

F508del-CFTR by a CDK-independent mechanism. British journal of

pharmacology, 171(21), 4831-4849.https://doi.org/10.1111/bph.12859

61. Vauthier, V., Ben Saad, A., Elie, J. et al. Structural analogues of roscovitine

rescue the intracellular traffic and the function of ER-retained ABCBvariants in cell modelsSci Rep 9, 6653 (2019).https://doi.org/10.1038/s41598-019-43111-y


 

62. Tang, X. X., Ostedgaard, L. S., Hoegger, M. J., Moninger, T. O., Karp, P. H.,

McMenimen, J. D., Choudhury, B., Varki, A., Stoltz, D. A., & Welsh, M. J.

(2016). Acidic pH increases airway surface liquid viscosity in cystic fibrosis. The

Journal of clinical investigation, 126(3), 879-891.

https://doi.org/10.1172/JCI83922

63. Naehrig, S., Chao, C. M., & Naehrlich, L. (2017). Cystic Fibrosis. Deutsches

Arzteblatt international, 114(33-34), 564-574.

https://doi.org/10.3238/arztebl.2017.0564

64. Bear, C. E., Li, C. H., Kartner, N., Bridges, R. J., Jensen, T. J., Ramjeesingh, M., & Riordan, J. R. (1992). Purification and functional reconstitution of the cystic

fibrosis transmembrane conductance regulator (CFTR). Cell, 68(4), 809-818.

https://doi.org/10.1016/0092-8674(92)90155-6

65. Condren, M. E., & Bradshaw, M. D. (2013). Ivacaftor: anovel gene-based

therapeutic approach for cystic fibrosis. The journal of pediatric pharmacology

and therapeutics JPPTthe official journal of PPAG, 18(1), 8- 13.

https://doi.org/10.5863/1551-6776-18.1.8

66. Eckford, P. D., Li, C., Ramjeesingh, M., & Bear, C. E. (2012). Cystic fibrosis

transmembrane conductance regulator (CFTR) potentiator VX-770 (ivacaftor)

opens the defective channel gate of mutant CFTR in a phosphorylation-dependent but ATP-independent manner. The Journal of biological chemistry, 287(44),

36639-36649.https://doi.org/10.1074/jbc.M112.393637

67. Fohner, A. E., McDonagh, E. M., Clancy, J. P., Whirl Carrillo, M., Altman, R. B., & Klein, T. E. (2017). PharmGKB summary: ivacaftor pathway,

pharmacokinetics/pharmacodynamicsPharmacogenetics and genomics, 27(1), 39-42.https://doi.org/10.1097/FPC.0000000000000246

68. Fiedorczuk, K., & Chen, J. (2022). Mechanism of CFTR correction by type I

folding correctors. Cell, 185(1),158- 168.e11.

https://doi.org/10.1016/j.cell.2021.12.009

69. Woodward, O. M., Tukaye, D. N., Cui, J., Greenwell, P., Constantoulakis, L. M., Parker, B. S., Rao, A., Köttgen, M., Maloney, P. C., & Guggino, W. B. (2013).

Gout-causing Q141K mutation in ABCG2 leads to instability of the nucleotide- binding domain and can be corrected with small molecules. Proceedings of the  National Academy of Sciences of the United States of America, 110(13), 5223-  5228.https://doi.org/10.1073/pnas.1214530110

70. Wang, T., Larcher, L. M., Ma, L., & Veedu, R. N. (2018). Systematic Screening of Commonly Used Commercial Transfection Reagents towards Efficient

Transfection of Single-Stranded Oligonucleotides. Molecules (Basel,

Switzerland), 23(10), 2564.https://doi.org/10.3390/molecules23102564

71. Hwang, T. C., & Kirk, K. L. (2013). The CFTR ion channel: gating, regulation, and anion permeation. Cold Spring Harbor perspectives in medicine, 3(1),

a009498.https://doi.org/10.1101/cshperspect.a009498

72. Soutar, A. K., & Naoumova, R. P. (2007). Mechanisms of disease: genetic causes of familial hypercholesterolemiaNature clinical practice. Cardiovascular

medicine, 4(4), 214-225.https://doi.org/10.1038/ncpcardio0836


 

73. Taylor, N., Manolaridis, I., Jackson, S. M., Kowal, J., Stahlberg, H., & Locher, K.

P. (2017). Structure of the human multidrug transporter

ABCG2. Nature, 546(7659), 504-509.https://doi.org/10.1038/nature22345 74. Higgins, C., Hiles, I., Salmond, G. et al. A family of related ATP-binding

subunits coupled to many distinct biological processes in

bacteriaNature 323, 448-450 (1986).https://doi.org/10.1038/323448a0

75. Cuperus, F. J., Claudel, T., Gautherot, J., Halilbasic, E., & Trauner, M. (2014). The role of canalicular ABC transporters in cholestasisDrug metabolism and  disposition: the biological fate of chemicals, 42(4), 546-560.

https://doi.org/10.1124/dmd.113.056358

76. Nam, S. M., & Jeon, Y. J. (2019). Proteostasis In The Endoplasmic Reticulum:  Road to Cure. Cancers, 11(11), 1793.https://doi.org/10.3390/cancers11111793

77. Lippincott-Schwartz, J., Bonifacino, J. S., Yuan, L. C., & Klausner, R. D. (1988). Degradation from the endoplasmic reticulumdisposing of newly synthesized

proteins. Cell, 54(2), 209-220. https://doi.org/10.1016/0092-8674(88)90553-3

78. Van Goor F, Hadida S, Grootenhuis PD, et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiatorVX-770. Proc Natl Acad Sci USA.

2009;106(44):18825- 18830. doi:10.1073/pnas.0904709106

79. Bianchi TS and Bauer JE (2011) Particulate Organic Carbon Cycling and

Transformation. In: Wolanski E and McLusky DS (eds.) Treatise on Estuarine and Coastal ScienceVol 5,pp. 69- 117. WalthamAcademic Press.

80. Lu, K., Lee, M. H., & Patel, S. B. (2001). Dietary cholesterol absorption; more

than just bile. Trends in endocrinology and metabolism: TEM, 12(7), 314-320.

https://doi.org/10.1016/s1043-2760(01)00433-7

81. Norlin, M., & Wikvall, K. (2007). Enzymes in the conversion of cholesterol into

bile acids. Current molecular medicine,7(2), 199-218.

https://doi.org/10.2174/156652407780059168

82. Stefková, J., Poledne, R., & Hubácek, J. A. (2004). ATP-binding cassette (ABC) transporters in human metabolism and diseases. Physiological research, 53(3),   235-243.

83. Procko, E., Ferrin-O'Connell, I., Ng, S. L., & Gaudet, R. (2006). Distinct

structural and functional properties of the ATPase sites in an asymmetric ABC

transporterMolecular cell, 24(1), 51-62.

https://doi.org/10.1016/j.molcel.2006.07.034

84. Kusuhara, H., & Sugiyama, Y. (2007). ATP-binding cassette, subfamily G

(ABCG family). Pflugers ArchivEuropean journal of physiology, 453(5), 735- 744. https://doi.org/10.1007/s00424-006-0134-x

85. Kullak-Ublick, G. A., Stieger, B., & Meier, P. J. (2004). Enterohepatic bile salt transporters in normal physiology and liver disease. Gastroenterology, 126(1),  322-342.https://doi.org/10.1053/j.gastro.2003.06.005

86. Salen, G., Ahrens, E. H., Jr, & Grundy, S. M. (1970). Metabolism of beta- sitosterol in man. The Journal of clinical investigation, 49(5), 952-

967.https://doi.org/10.1172/JCI106315

87. Yamamoto, H., Yamanashi, Y., Takada, T., Mu, S., Tanaka, Y., Komine, T., &

Suzuki, H. (2019). Hepatic Expression of Niemann-Pick C1-Like 1, a Cholesterol Reabsorber from Bile, Exacerbates Western Diet-Induced Atherosclerosis in LDL


 

Receptor Mutant MiceMolecular pharmacology, 96(1), 47-55.

https://doi.org/10.1124/mol.119.115840

88. Hazard, S. E., & Patel, S. B. (2007). Sterolins ABCG5 and ABCG8: regulators of whole body dietary sterolsPflugers ArchivEuropean journal of

physiology, 453(5), 745-752. https://doi.org/10.1007/s00424-005-0040-7

89. Horton, J. D., Shah, N. A., Warrington, J. A., Anderson, N. N., Park, S. W.,

Brown, M. S., & Goldstein, J. L. (2003). Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP

target genes. Proceedings of the National Academy of Sciences of the United

States of America, 100(21), 12027- 12032.

https://doi.org/10.1073/pnas.1534923100

90. Venkateswaran, A., Laffitte, B. A., Joseph, S. B., Mak, P. A., Wilpitz, D. C.,

Edwards, P. A., & Tontonoz, P. (2000). Control of cellular cholesterol efflux by  the nuclear oxysterol receptor LXR alphaProceedings of the National Academy

of Sciences of the United States of America, 97(22),12097- 12102.

https://doi.org/10.1073/pnas.200367697

91. Yu, L., Gupta, S., Xu, F., Liverman, A. D., Moschetta, A., Mangelsdorf, D. J.,

Repa, J. J., Hobbs, H. H., & Cohen, J. C. (2005). Expression of ABCG5 and

ABCG8 is required for regulation of biliary cholesterol secretion. The Journal of biological chemistry, 280(10), 8742-8747.

92. Chiang J. Y. (2013). Bile acid metabolism and signaling. Comprehensive Physiology, 3(3), 1191- 1212. https://doi.org/10.1002/cphy.c120023

93. Jonas A. (2000). Lecithin cholesterol acyltransferase. Biochimica et biophysica acta, 1529(1-3), 245-256.https://doi.org/10.1016/s1388-1981(00)00153-0

94. Wang J., Grishin N., Kinch L., Cohen J.C., Hobbs H.H. and Xie X.S.

(2011) Sequences in the nonconsensus nucleotide-binding domain of

ABCG5/ABCGrequired for sterol transport. J. Biol. Chem286, 7308-7314 10.1074/jbc.M110.210880

95. Altmann, S. W., Davis, H. R., Zhu, L., Yao, X., Hoos, L. M., Tetzloff, G., Sai

Prasad N. Iyer, Maguire, M., Golovko, A., Zeng, M., Wang, L., Murgolo, N., &  Graziano, M. P. (2004). Niemann-Pick C1 like 1 Protein Is Critical for Intestinal

Cholesterol Absorption. Science, 303(5661), 1201- 1204.

http://www.jstor.org/stable/3836300

96. Xavier, B. M., Zein, A. A., Venes, A., Wang, J., & Lee, J. Y. (2020).

Transmembrane Polar Relay Drives the Allosteric Regulation for ABCG5/G8

Sterol TransporterInternational journal of molecular sciences, 21(22), 8747.

https://doi.org/10.3390/ijms21228747

97. Hunt, J. F., Wang, C., & Ford, R. C. (2013). Cystic fibrosis transmembrane

conductance regulator (ABCC7) structure. Cold Spring Harbor perspectives in medicine, 3(2), a009514.https://doi.org/10.1101/cshperspect.a009514

98. Stieger B. (2011). The role of the sodium-taurocholate cotransporting polypeptide (NTCP) and of the bile salt export pump (BSEP) in physiology and

pathophysiology of bile formation. Handbook of experimental pharmacology, (201), 205-259.https://doi.org/10.1007/978-3-642-14541-4_5


 

 

 

VITA

 

1.   Born in Kalamazoo, MI.

2.  Bachelors of Science from the University of South Florida (May 2020), Enrolled at the University of Kentucky Medical Science program.

3.   American Heart Association ATVB Travel Grant for Early Career Investigators Recipient (2022)

4.   Brittney Poole