CHOLESTEROL 7 ALPHA-HYDROXYLASE IS REGULATED POST- TRANSLATIONALLY BY AMPK

Size: px
Start display at page:

Download "CHOLESTEROL 7 ALPHA-HYDROXYLASE IS REGULATED POST- TRANSLATIONALLY BY AMPK"

Transcription

1 CHOLESTEROL 7 ALPHA-HYDROXYLASE IS REGULATED POST- TRANSLATIONALLY BY AMPK A dissertation submitted to Kent State University in partial fulfillment of the requirements for the Degree of Doctor of Philosophy By Mauris E.C. Nnamani May 2009

2 Dissertation written by Mauris E. C. Nnamani B.S, Kent State University, 2006 Ph.D., Kent State University, 2009 Approved by Diane Stroup Gail Fraizer Advisor Members, Doctoral Dissertation Committee S. Vijayaraghavan Arne Gericke Jennifer Marcinkiewicz Accepted by Robert Dorman John Stalvey, Director, School of Biomedical Science, Dean, Collage of Arts and Sciences ii

3 TABLE OF CONTENTS LIST OF FIGURES..vi ACKNOWLEDGMENTS..viii CHAPTER I: INTRODUCTION....1 a. Bile Acid Synthesis..2 i. Importance of Bile Acid Synthesis Pathway....2 ii. Bile Acid Transport iii. Bile Acid Synthesis Pathway... 4 iv. Classical Bile Acid Synthesis Pathway Cholesterol 7 -hydroxylase (CYP7A1) Transcriptional Regulation of Cholesterol 7 -hydroxylase by Bile Acid-activated FXR CYP7A1 Transcriptional Repression by SHP-dependant Mechanism...10 CYP7A1 Transcriptional Repression by SHP-independent Mechanism CYP7A1 Transcriptional Repression by Activated Cellular Kinase v. Alternative/ Acidic Bile Acid Synthesis Pathway Sterol 27-hydroxylase (CYP27A1)..12 Sterol 12 -hydroxylase (CYP8B1) b. Cholesterol i. Effects of Excess Cholesterol...19 ii. Importance of Cholesterol c. Cholesterol Synthesis d. Regulation of Cholesterol Synthesis.. 25 i. HMG-CoA Reductase ii. Transcriptional Regulation of HMG-CoA Reductase iii. Regulation of HMG-CoA Reductase by Phosphorylation/Dephosphorylation Events..27 iv. Repression of HMG-CoA Reductase by Statin Drugs...27 e. Physiological Relevance and Significance of the Bile Acid Synthesis Pathway. 28 iii

4 f. Experimental Rationale i. Experimental Approach, Hypothesis, and Specific aims CHAPTER II: MATERIALS AND METHODS..37 PART 1: Cholesterol 7 -hydroxylase Specific Antibodies a. Rational for Cholesterol 7 -hydroxylase Specific Antibody Production...37 b. Antigen (C2) used for CYP7A1 Antibody Production...37 c. Sub-cloning of the C2 cdna Sequence into E. coli-expression Vector..38 d. Expression of the C2 antigen...42 i. Expression of C2 with BL21 (DE3) E. coli Cell Line...42 ii. Expression of C2 with the Roche RTS 500 E. coli HY kit..46 e. Immunization for Antibody Production...49 f. Characterization of Anti-serum from Immunized Rabbits...51 g. Pre-absorption of Rabbit Serum with E. coli lysate. 57 h. Antibody Characterization with Purified C2 Antigen i. Antibody Affinity Purification i.c2 Affinity Column Preparation..62 j. C2 Antibody Affinity Purification 63 PART 2: Expression of Recombinant CYP7A1 in E. coli...68 a. History of CYP7A1 Expression...68 b.sub-cloning WT CYP7A1 cdna into E. coli-expression Vector c. Characterization of Recombinant CYP7A1 Protein d. Characterization of Expressed 30KD Fragment..74 PART 3: CYP7A1 Enzymatic Assay a. History of CYP7A1 Assay Detection Methods...82 b. CYP7A1 Assay Detection Method..83 c. Internal Standard for Enzyme Assay 84 d. Evaluating Assay Detection.91 e. CYP7A1 Enzyme Assay..91 f. Enzymatic Activity of the 30KD Fragment..99 PART 4: Mutagenesis..100 a. Site-directed Mutagenesis Strategy b. Site-directed Mutagenesis Strategy c. Site-directed Mutagenesis Strategy d. Site directed Mutagenesis Strategy e. Mutant Sub-cloning and Characterization.116 f. Mutant Vector-construct Restriction Mapping..119 iv

5 CHAPTER III: RESULTS AND CONCLUSIONS 124 PART 1: AICAR, an AMPK Kinase Activator, does not Affect CYP7A1 mrna Steady-state Levels PART 2: CYP7A1 In-vitro Reconstituted Assay with E. coli-expressed and Microsomal HepG2 Cells Treated with AMPK, PKC, and JNK Kinases a. Rational for Kinase Selection i. AMPK ii. PKC iii. JNK b. Kinase Treatment of Microsomal HepG2 Cells c. Kinase Treatment of E. coli-expressed CYP7A1 Recombinant Protein PART 3: AMPK Kinase Treated In-vitro CYP7A1 Enzymatic Assay with Mutant CYP7A1 Recombinant Protein a. Rational for Mutation Design b. Rational for Mutation-site Selection c. Mutation of AMPK Phosphorylation Sites d. CYP7A1 In-vitro Assay with T193A /T197A Double Mutant..141 e. CYP7A1 In-vitro Assay with T80A Mutant..145 f. CYP7A1 In-vitro Assay with S252G Mutant g. CYP7A1 in-vitro assay with Truncated WT CYP7A1 ( 1-729) Mutant CHAPTER IV: DISCUSSION..154 CHAPTER V: APPENDIX APPENDIX A: EXPERIMENTAL PROTOCOLS APPENDIX B: ABBREVIATIONS 191 REFERENCES v

6 LIST OF FIGURES Figure 1. Differences Between Primary and Secondary Bile Acids 7 Figure 2. Cholic Acid and Chenodeoxycholic Acid..15 Figure 3. Schematic Diagram of the Bile Acid Biosynthesis Pathway...17 Figure 4. Cholesterol Biosynthesis 24 Figure 5. ClustalW (1.83) Multiple Sequence Alignment of C2 and Full-length CYP7A1..41 Figure 6. Western Blot Analysis of In-vitro Expressed C2 Fragment for Antibody Production..45 Figure 7. RTS 500-expressed C2 Antigen for Antibody Production Figure 8. Western Blot of C2 Samples with the First Small Bleed of Immunized Rabbits...54 Figure 9. Western Blot of C2 Samples with Serum After Two Months of Immunization.56 Figure 10. Western Blot Analysis of C2 Antigen with Pre-absorbed Rabbit Serum.58 Figure 11. Western Blot Analysis of Purified C2..61 Figure 12. Coomassie Blue Staining of Purified C2 Polyclonal Antibody...66 Figure 13. Western Blot Analysis with Affinity Purified C2 Polyclonal Antibodies 67 Figure 14. Western Blot of E. coli-expressed WT CYP7A1 Recombinant Protein..71 Figure 15. Protein Sequence Alignment of Human Cytochrome P450 7A1 (CYP7A1) and the 30KD Isolated Fragment.76 Figure 16. Protein Alignment of the C2 Polypeptide and the 30KD Fragment.78 Figure 17. Western Blot of E. coli-expressed WT CYP7A1 and Truncated CYP7A1 Recombinant Protein..81 Figure 18a. Standard Curve of 7 -hydroxycholesterol using the HPLC-MS.. 87 Figure 18b. Extracted Portion of the Standard Curve for 7 -hydroxycholesterol using the HPLC-MS..88 Figure 19. D7- and 7 -hydroxycholesterol Co-injection.90 Figure 20. Complete and Extracted Ion Traces of Samples from the CYP7A1 Enzyme Assay Figure 21. CYP7A1 Reconstituted In-vitro Assay Performed with Vector only and WT CYP7A1 E. coli-expressed Recombinant Proteins Figure 22. Schematic Diagram to Illustrate Site-directed Mutagenesis Strategy Figure 23. Schematic Diagram to Illustrate Site-directed Mutagenesis Strategy vi

7 Figure 24. Schematic Diagram to Illustrate Site-directed Mutagenesis Strategy Figure 25. Schematic Diagram to Illustrate Mutagenesis Strategy 4 used in Generate Truncated CYP7A1 polypeptide Figure 26. Mini Plasmid prep of Colonies from Mutants Transformed into E.coli DH5 Cell Lines Figure 27. Restriction Mapping of Mutant Plasmid Figure 28. Contiguous Alignment of Mutant Nucleotide Sequence Figure 29. AICAR, AMPK Kinase Activator does not Affect CYP7A1 mrna Steady-state Levels..127 Figure 30. AMPK, PKC, and JNK Kinase Activity Repressed CYP7A1 Enzymatic Activity in Microsomal HepG2 Cell Fractions 133 Figure 31. AMPK Kinase Activity Represses CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein Figure 32. Tabulated Results from Synthetic Peptides Treated with Commercially Available Protein Kinases 140 Figure 33. AMPK Kinase Activity Represses T193A/T197A Double Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein Figure 34. AMPK Kinase Activity Represses T80A Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein.147 Figure 35. AMPK Kinase Activity did not Repress S252G Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein Figure 36. AMPK Kinase Activity Stimulates WT CYP7A1 ( 1-729) Enzymatic Activity Figure 37. Crystal Structure of CYP7A1.166 Figure 38. AMPK Maintains Cholesterol Homeostasis vii

8 ACKNOWLEDGEMENTS I would like to acknowledge those that made the completion of my Doctoral study a success. I would like to express my extreme gratitude to my advisor Dr. Diane Stroup, for her wisdom, encouragement, support, patience and most importantly, her trust throughout my graduate studies. Her scientific expertise and guidance made the completion of this dissertation a success. I would like to thank my committee members: Dr. Gail C. Fraizer, Dr. Arne Gericke, Dr. Jennifer Marcinkiewicz and Dr. S. Vijayaraghavan; for the valuable time they put into my work. Their scientific suggestions, guidance and support helped improve this dissertation. I would like to thank Dr. Robert J. Twieg, for allowing me full access to the LC/MS in his lab. His support and confidence in my ability to operate the equipment, allowed me to complete my experiments in a timely fashion. I would also like to thank my colleague, Jing Zhao, for her scientific discussions and assistance with the real-time PCR experiments. Our discussions helped spark ideas for solving experimental problems. Last, but not least, I would like to thank my family; my parents Emmanuel and Martha Nnamani; my sisters, Adaeze and Maryann Nnamani; my brothers, Kim and Jason Nnamani; Alysse Ferranto and the Ferranto family. I want to thank them for believing in me and challenging me to achieve new heights. Without their love and support none of this would have been possible. viii

9 CHAPTER I INTRODUCTION The rate at which cholesterol is converted to bile acids is determined by the activity of the cholesterol 7 -hydroxylase, encoded on the CYP7A1 gene [1-3]. This rate-limiting enzyme is highly regulated on multiple levels, including control of initiation of transcription and message stability [1]. Transcriptional repression of CYP7A1 has been well documented [4-8], but less studied are the other levels of regulation that includes a possible post-translational control of the enzyme. Published studies on the transcriptional regulation of CYP7A1 often did not include measurement of enzyme activity. However, when reported, the mrna levels did not always correlate with bile acid production. Previous work in our lab show that bile acid production, in Human hepatoma liver carcinoma (HepG2) cell line, is repressed by 5`AMP-activated protein kinase (AMPK) activator, AICAR (Stroup and Ramsaran, 2005). This is the first report of regulation of bile acid production through the AMPK pathway, an important pathway that has long been known as a repressor of cholesterol synthesis by shutting down 3-hydroxyl 3-methylglutaryl-coenzyme A (HMG-CoA) reductase. 1

10 2 The hypothesis for this study is that AMPK kinase activity regulates cholesterol 7 -hydroxylase enzymatic activity via phosphorylation at the enzyme level. a. Bile Acid Synthesis i. Importance of Bile Acid Synthesis Pathway Bile acid synthesis is the major cholesterol disposal pathway and thus, influences cholesterol homeostasis. Bile acids serve as detergents in the intestine and facilitate the absorption, transport, and distribution of lipid soluble vitamins and dietary fats. They also act as signaling molecules and regulate gene expression by acting as a ligand for the nuclear hormone receptor, farnesoid X-receptor (FXR) nuclear receptor, and by activating protein cellular kinases[2]. Bile acids also induce the cytochrome P450 3A (CYP3A) family of cytochrome P450 enzyme, that play a role in drug metabolism and detoxification of xenobiotics in the liver and intestine[9]. Intermediates of the bile acids synthesis pathway and oxysterols, have a broad spectrum of biological effects on the activity of enzymes involved in cholesterol homeostasis[10]. Oxysterols, which are oxidized cholesterol, are potent signaling molecules that regulate cell proliferation, apoptosis, brain function and the immune system[2]. Several oxysterols including 20 -, 22 -, 25-, and 27-hydroxycholesterol are reported to be ligands of the nuclear hormone receptor, liver X receptor (LXR)[10]. This is significant, notably through genes whose promoters contain LXR binding elements,

11 3 and also through LXR s interaction with other transcription factors, such as PPAR and SHP. The anti-apoptotic effect of ursodeoxycholic acid (UDCA), a bile acid conjugate, has been used to reduce neurological damage following stroke and slow the progression of Huntington s disease[11, 12]. Alterations in bile acid synthesis have been associated with diabetes and cardiovascular disease. Patients with diabetes mellitus and streptozotocin-diabetic rats, show larger bile acid pools and bile acid excretion rates compared to normal individuals [2]. Disruption in the bile acid pathway has also been shown to be associated with hypercholesterolemia and cardiovascular disease. ii. Bile Acid Transport Bile acids are synthesized in the liver and stored in the gallbladder. After each meal, the gallbladder contracts and releases bile acids into the intestine to solubilize dietary fats. The bile acids are reabsorbed in the ileum and transported back to the liver by enterohepatic circulation. This process reabsorbs about 95% of bile acids in humans[9]. Studies to determine the mechanism involved in the enetrohepatic circulation, leading to bile acid turnover, is ongoing. It was suggested by Low-Beer, et al 1969[13]; that the main factors determining turnover and synthesis of the primary bile acids is the emptying of the gallbladder. This assumption was deduced by the association of delayed gallbladder emptying and increased bile acid pool in patients with celiac disease. More recently, nuclear hormone receptors have been implicated in the regulation

12 4 of bile acid synthesis through the enterohepatic circulation through the FGF-19/FGFR4 pathway [14]. iii. Bile Acid Synthesis Pathways Primary Bile acids and their conjugates There are two pathways involved in bile acid synthesis, the classical (neutral) pathway and the acidic (alternative) pathway. Both the classical and acidic bile acid pathways lead to the production of cholic and chenodeoxycholic primary bile acids, which are the most abundant bile acids produced. They differ in that cholic acid requires an additional enzyme, 12 -hydroxylase, for synthesis and has a hydroxyl group at the 12 position. Cholic and chenodeoxycholic acids are called primary bile acids because they are the principle bile acids formed form cholesterol in mammals and are precursors for the production of secondary bile acid conjugates. Portions of the cholic acid and chenodeoxycholic acid are conjugated in the liver by the addition of either glycine or taurine to the side-chain carboxyl-group of the bile acid through amidation. As bile acid is secreted into the intestines, cholic acid and chenodeoxycholic acid are converted into deoxycholic acid and lithocholic acid respectively, by bacterial 7 -dehydroxylase in the intestines[15]. The bile acids that were conjugated with glycine or taurine can be further modified, forming other forms of secondary bile acids. Bile acids in humans are

13 5 primarily, lithocholic, deoxycholic, chenodeoxycholic, and cholic acids, which can all be conjugated by either glycine or taurine[16].

14 6 Figure 1. Differences between Primary and Secondary Bile Acids Cholesterol gets converted into cholic and chenodeoxycholic primary bile acids in the liver. Portions of the primary bile acids can be conjugated by the addition of either glycine or taurine to the carboxyl-group of the primary bile acids to form the glyco- and tauro- conjugates respectively. 7 -dehydroxylation takes place in the intestines by bacteria flora, and dehydroxylate the 7 -hydroxyl positions. Image modified from Duarte et al., 2009[17].

15 Figure 1. Differences Between Primary and Secondary Bile Acids 7

16 8 The three major enzymes that play important roles in regulating bile acid production through the two pathways are: cholesterol 7 -hydroxylase (CYP7A1) is the rate limiting step in the classical pathway, sterol 27 hydroxylase (CYP27A1) is the first enzyme in the alternative pathway, and 12 -hydroxylase (CYP8B1) is the specific enzyme that determines the ratio of cholic acid and chenodeoxycholic acid[18]. iv. Classical Bile Acid Synthesis Pathway The rate at which cholesterol is converted to bile acids in the classical pathway is determined by the activity of the rate-limiting enzyme cholesterol 7 -hydroxylase[1-3]. Bile acid synthesis by this pathway has been reported to account for approximately 50% of total bile acid production in rats and mice, and greater than 90% of total bile acids in humans [19]. Cholesterol 7 -hydroxylase is found solely in the liver, and the liver is the only organ capable of producing bile acids. Cholesterol 7 -hydroxylase Cholesterol 7 -hydroxylase is a 504 amino acid cytochrome P450 monooxygenase and is believed to be anchored to the smooth endoplasmic reticulum by a 23 amino acid N-terminal internal-membrane sequence. The enzyme contains a steroid binding site, an aromatic amino acid region and a heme region. Cholesterol 7 hydroxylase hydroxylates cholesterol to 7 -hydroxycholesterol in the first step of the classical bile acid synthesis pathway. The 7 -hydroxycholesterol is converted to 7 -

17 9 hydroxy-4-cholesten-3-one (C4) by 3- -hydroxysteriod dehydrogenase (3 HSD). The pathway can then follow two different routes to produce the two major primary bile acids, cholic acid and chenodeoxycholic acid. The factor that determines whether the 3 HSD will be converted to cholic or chenodeoxycholic acid is the enzymatic activity of sterol 12 -hydroxylase (CYP8B1). A 12 hydroxylation by CYP8B1 will drive the reaction towards the formation of cholic acid. While a conversion of the 3 HSD to 5 cholestane-3, 7 -diol will drive the reaction towards the formation of chenodeoxycholic acid. This process is illustrated in Figure 3. The expression of cholesterol 7 -hydroxylase is controlled at multiple levels, including control of initiation of transcription and message stability [1]. Transcription of CYP7A1 is repressed by bile acids, phorbol esters, and insulin and is activated by cyclic- AMP[20, 21]. The transcriptional repression of cholesterol 7 -hydroxylase by bile acids has been shown to occur through a feedback regulation mechanism. Bile acids feedback repression of cholesterol 7 -hydroxylase transcriptional activity is accomplished by a bile acid associated activation of farnesoid X-receptor (FXR).

18 10 Transcriptional Regulation of Cholesterol 7 -hydroxylase by Bile Acid-activated FXR Bile acids are believed to regulate cholesterol 7 -hydroxylase gene expression by binding and activating FXR[14, 22-25] and by activating cellular kinases [23, 26-28]. FXR is expressed in the liver, intestine, kidney, and adrenal and regulates the synthesis of cholesterol 7 -hydroxylase and oxysterol 7 -hydroxylase (CYP8B1), both the rate limiting enzymes in the classical and acidic pathway for bile acid synthesis respectively[29, 30]. Bile acid activated FXR regulate bile acid synthesis through an indirect feedback repression of CYP7A1 transcriptional activity. CYP7A1 Transcriptional Repression by SHP-dependant Mechanism Bile acid activated FXR binds as a heterodimer with retinoid X receptor (RXR) to DNA, recognizing an inverted hexanucleotide repeats composed of two nuclear receptor half sites of the consensus AG(G/T)TCA, and separated by a single nucleotide (IR-1 motif) [24, 29, 30]. The activated FXR/RXR heterodimer has been shown to repress CYP7A1 promoter activity by the activation of other factors[24, 31]. Activated FXR can induce the expression of small heterodimer partner 1 (SHP-1) protein, an orphan nuclear receptor. Elevated levels of SHP-1 have been associated with repressed CYP7A1 transcription activity in mice. It has been proposed that elevated SHP-1 protein levels prevent CYP7A1 transcriptional activation by forming a heterodimer with the orphan nuclear receptor LRH-1 (liver receptor homolog 1) (CPF or FTF)[26] and by interfering with the activity of the liver X receptor (LXR). The importance of SHP in bile acid

19 11 synthesis feedback regulation was demonstrated in SHP -/- mice, where the mice were shown to have increased CYP7A1 expression, enzyme activity, and bile acid pool size[32]. It still remains controversial whether this mechanism exists in humans. Notable differences exist in bile acid biosynthesis between rodents and human, most notable is the regulation of CYP7A1 promoter by LXR [33]. LXR stimulation in rodents leads to activation of CYP7A1 transcriptional activity[9], but this effect is not noticed in humans. CYP7A1 Transcriptional Repression by SHP-independent Mechanism Activated FXR induces the expression of fibroblast growth factor-19 (FGF-19) in primary human hepatocytes culture. FGF-19 is a secreted protein that has been proposed to regulate CYP7A1 transcription through a c-jun N-terminal kinase (JNK)-dependent pathway[34]. FGF-19 selectively binds to FGF receptor 4 (FGFR4), a transmembrane receptor with tyrosine kinase activity[35]. The FXR activated FGF-19/FGFR4 pathway has been proposed to be the mechanism by which bile acid synthesis is regulated through the enterohepatic circulation. This proposal was a result of experiments performed with the mouse ortholog FGF-15. FGF-15 was induced by FXR in the small intestines of mice and repressed bile acid synthesis through a mechanism involving FGFR4 and SHP[36].

20 12 CYP7A1 Transcriptional Repression by Activating Cellular Kinase It has been long speculated that bile acids can repress cholesterol 7 -hydroxylase transcriptions through activating cellular kinases. HepG2 cells treated with bile acids resulted in the activation and translocation of protein kinase C (PKC) from the cytosol to the membrane[37, 38]. PKC inhibitors reduced the bile acid repression of transcription from the CYP7A promoter[37]. Phorbol esters activate PKC and have been shown to repress CYP7A1 transcription[37]. Bile acids have also been shown to activate c-jun N- terminal kinase (JNK) pathway and Raf-1/MEK/ERK signaling cascade in primary rat hepatocytes[26, 39]. Although the mechanism by which bile acids activate cellular kinases are not fully understood, it has been shown that transcription repression of cholesterol 7 -hydroxylase is associated with kinase activation. v. Alternative /Acidic Bile Acid Synthesis Pathway Sterol 27-hydroxylase (CYP27A1) Sterol 27-hydroxylase is the first enzyme in the acidic pathway, and in contrast to cholesterol 7 -hydroxylase, is a widely distributed mitochondrial enzyme with high activity in vascular endothelial cells[40]. In the extrahepatic tissues, the acidic pathway leads to the synthesis of 3, 7 -dihydroxy-5-cholestenoic acid, which is a precursor for chenodeoxycholic acid synthesis.

21 13 The purpose of the acidic pathway in extrahepatic tissues is apparently a salvage pathway for oxysterols. Recombinant CYP27A1 has been shown to have a wide substrate specificity[41]; it is possible that the enzyme has more roles beyond C 27 -sterol side chain cleavage. Studies have shown that oxysterol products from CYP27A1 activities, 27-hydroxycholesterol and 3 -hydroxy-5-cholestenoic acid, are ligands for the nuclear receptors LXR and LXR that are involved in transcriptional regulation of cholesterol homeostasis[42, 43]. Although a few studies argue against the regulatory role of CYP27A1[44, 45]. More recent studies have implicated CYP27A1 with activating the liver orphan receptor (LXR ) and inducing cholesterol efflux transporters ABCA1 and ABCG1 in macrophages[46]. It was also hypothesized earlier, CYP27A1 is likely to be an essential cholesterol mobilization mechanism and not primarily for producing bile acids [2]. This hypothesis is supported by numerous studies showing the insensitivity of CYP27A1 to bile acid feedback regulating [47-49] Patients with the rare inherited disease cerebrotendinous xanthomatosis (CTX), have a mutation on their CYP27A1 gene resulting in a partial or complete lack of CYP27A1 activity[50]. Clinical manifestations of CTX vary and may include tendon xanthomas, bilateral cataracts, accelerated atherosclerosis and progressive neurological impairment[51]. There have been 40 different mutations found in CYP27A1 that are associated with causing CTX, but no phenotype-genotype correlation has been established so far[52-54].

22 14 Sterol 12 -hydroxylase (CYP8B1) Sterol 12 -hydroxylase (CYP8B1) is the specific enzyme that determines the ratio of cholic acid (CA) and chenodeoxycholic acid (CDCA) produced in the cell. Both primary bile acids are formed at a ratio of ~ 2:1 in humans[50]. CYP8B1 has a broad substrate specificity and is solely expressed in the liver[55, 56]. CA is more hydrophilic than CDCA; therefore, by controlling the synthesis of CA, CYP8A1 determines the overall hydrophobicity of the bile acid production.

23 15 OH COOH COOH OH OH OH OH Cholic acid Chenodeoxycholic acid Figure 2. Cholic Acid and Chenodeoxycholic Acid. Cholic and chenodeoxycholic acid are primary bile acids produced in the liver. The ratio of the bile acids produced is determined by the enzymatic activity of CYP8B1. Cholic acid is only produced by the natural pathway and has three hydroxyl groups at positions 3, 7, and 12, making it more hydrophilic than chenodeoxycholic acid. Chenodeoxycholic acid is produced by both the natural and acidic bile acid pathways and has hydroxyl groups at positions 3 and 7.

24 16 Figure 3: Schematic Diagram of the Bile Acid Biosynthesis Pathway. In the classical pathway, cholesterol is hydroxylated at the 7 position by cholesterol 7 hydroxylase (CYP7A1) first, followed by several other oxidation reactions. What determines the ratio of cholic to chenodeoxycholic acid is the activity of sterol 12 hydroxylase (CYP8B1). In the alternative pathway, cholesterol is modified by sterol 27- hydroxylase first, and then is hydroxylated at the 7 position by oxyxterol 7 hydroxylase (CYP7B1) (Image from Chiang, 2002).

25 Figure 3. Schematic Diagram of the Bile Acid Biosynthesis 17

26 18 b. Cholesterol The levels of cholesterol in the body is determined by three major activities: uptake of dietary cholesterol and transport, cholesterol synthesized by the body and the conversion of cholesterol into other steroid compounds, such as bile acids and hormones [2, 20]. Cholesterol from the diet is absorbed into the blood from the small intesines, and transported to various tissues were they are needed. The hydrophobic cholesterol is transported throughout the body by lipoproteins. Different types of lipoproteins are classified based on their density as determined by ultracentrifugation. High density lipoproteins (HDL) are believed to function by taking cholesterol away from the blood to the liver were they are converted into bile acids and other steriod hormornes. HDL is refered to as good cholesterol because of clinical and epidemiological studies that show an inverse relationship between levels of HDL-cholesterol, and the risk of coronary heart disease (CHD) events [57]. LDL (low density lipoprotein) and VLDL (very low density lipoprotein), carries cholesterol through the blood to the tissues and. Elevated levels of LDL-cholesterol are shown to have a significantly positive correlation with a higher risk of CHD[58].

27 19 i. Effects of Excess Cholesterol Concentration of, and the ratio between, LDL and HDL are used to determine the risk of cardiovascular events. Hypercholesterolemia is a condition that results from high levels of serum cholesterol and is a strong risk factor for progression of atherosclerosis. Atherosclerosis is a slowly progressive disease of the arterial wall leading to thickening of the wall and resulting in blood flow obstruction. A Multicentral study by the Pathobiological Determinants of Atherosclerosis in Youth (PDAY), found that the atherosclerosis progression starts in childhood[59, 60], and progression accelerates up in aldulthood. The earliest pathologic finding in atherosclerosis is thought to be the fatty streaks[61], which are characterized by lipid-filled macrophages within the artery[62]. Atherosclerosis progression starts with a disruption of the endothelium layer of the arterial wall followed by the migration and accumulation of lipid-filled macrophages and poliferation of vascular smooth muscles cells. As the smooth muscles cells poliferate and migrate to the outer lining of the arteries they accumulate and form lesions called fibrous plaques[62]. As atherosclerosis progresses, the fibrous plaques build up and thicken the arterial walls causing blood flow obstruction. The plaques can also build up and rupture, breaking off into the blood stream, and are responsible for adverse clinical outcomes, such as myocardial infarctions and ischemic stroke[61].

28 20 Since cholesterol is the precursor for oxysterol, excess cholesterol leads to an excess production of oxysterols in the cell. The ratio of oxysterol:cholesterol in plaque are higher than in normal tissues or plasma, suggesting that they play a role in plaque development[63]. They have been shown to influence the molecular order of the cell membranes when compared to pure cholesterol, this suggests they might contribute to cell membrane permeability changes affecting crucial cell functions and events leading to vascular cell injury[64]. Oxysterols have also been shown to exhibit cytotoxicity in cells leading to angiotoxic and atherogenic effects; alter prostaglandin synthesis, stimulate platelet aggregation, and also alter the functionality of LDL receptors, possibly stimulating hypercholesterolemia[65]. ii. Importance of Cholesterol Despite the negative effects associated with elevated levels of cholesterol, cholesterol is still a very essential molecule for cellular survival. It is the precursor for important steroid hormones such as the estradiol, testosterone and vitamin D, which is necessary to utilize calcium and form bone. Cholesterol inserts into the lipid bilayer of the cell and makes it less susceptible to permeability from small water-soluble molecules. Cholesterol also stabilizes a cell against temperature changes. It is associated with the majority of membranes in the nervous system; in particular it is incorporated into the myelin sheath that insulates the nerves from the surrounding tissue. Cholesterol is a major part of membranes in the brain, the spinal cord and the peripheral nerves.

29 21 Cholesterol is the precursor for bile acids which have very important physiological functions, including but not limited to: cholesterol solubilization in the bile, in order to prevent precipitation in the gallbladder; facilitation of digestion of dietary triacylglycerol by acting as detergents; facilitating intestinal absorption of fat-soluble vitamins; and acting as signaling molecules in the cell. They are also oxygenated to oxysterols P450s, and function as signaling molecules in the cell. Cholesterol differs from the main steroid ring structures by having an aliphatic side chain at C17 of ring D, methyl groups at C10 and C13, a double bond in ring B, and a hydroxyl group on C3. The position of the OH group makes cholesterol a steroid alcohol or sterol. Almost every cell in the body is capable of producing cholesterol, but the liver and the intestines produce the majority of cholesterol in the body. c. Cholesterol Synthesis Cholesterol synthesis occurs in the cytosol and starts with the condensation of acetyl-coa and acetoacetyl-coa to form 3-hydroxy 3-methylglutaryl-coenzyme A (HMG-CoA), catalyzed by the enzyme HMG-CoA synthase. HMG-CoA is the precursor for cholesterol synthesis and is also an intermediate in the pathway for the synthesis of ketone bodies in the mitochondrial. The next step in cholesterol synthesis is the conversion of HMG-CoA to mevalonate. This reaction is catalyzed by the HMG-CoA reductase, which is the rate-limiting enzyme in cholesterol synthesis and utilizes NADPH. In the next step of cholesterol synthesis, mevalonate is converted to

30 22 isopentenyl pyrophosphate, and this step utilizes ATP. There are several additional steps involved to the production of cholesterol and a schematic diagram depicting major reactions in the mevalonate pathway for cholesterol synthesis is shown below [Fig. 4].

31 23 Figure 4. Cholesterol Biosynthesis. Synthesis begins with the transport of acetyl-coa from the mitochondria to the cytosol followed by condensation of acetyl-coa and Acetoacetyl-CoA into HMG-CoA. The rate-limiting step is catalyzed by HMG-CoA reductase, which is highly regulated by phosphorylation /dephosphorylation events by AMPK kinase activity, and is the key regulatory region for cholesterol lowering drugs (statins). (Cholesterol biosynthesis pathway was modified from The Medical Biochimistry Page, M. King Ph. D. IU School of Medicine).

32 Figure 4. Cholesterol Biosynthesis 24

33 25 d. Regulation of Cholesterol Synthesis i. HMG-CoA Reductase Cholesterol synthesis is regulated by HMG-CoA reductase, which is the ratelimiting enzyme in cholesterol biosynthesis. HMG-CoA reductase is regulated by protein degradation, at the transcriptional level, and at the enzyme level by phosphorylation /dephosphorylation events. ii. Transcriptional Regulation of HMG-CoA Reductase HMG-CoA reductase is regulated at the transcriptional level by a family of transcription factors known as sterol regulatory element binding proteins (SREBP). SREBPs are know to enhance transcription of over 30 genes involved in the uptake and synthesis of cholesterol, fatty acids, triglycerides, and phospholipids[66]. There are three SREBP isoforms: SREBP-1a, SREBP-1c/ADD1 and SREBP-2 encoded on two genes[67]. SREBP1a activates all SREBP-responsive genes while SREBP-1c and SREBP-2 are the predominant isoforms in the liver that regulate genes involved in fatty acid synthesis and sterol biosynthesis respectively[68]. SREBP is a membrane-bound transcription factor located in the endoplasmic reticulum (ER). The N-terminal domain of SREBP is a basic helix-loop-helix leucine zipper transcription factor and the c- terminal domain is bound to a SREBP-cleavage-activating protein (Scap) that acts as a sterol sensor[67, 69].

34 26 In the presence of cholesterol, SREBPs are localized in the ER. SREBP-Scap complex binds to cholesterol in the ER membrane and promotes binding to ER-resident protein Insig (Insulin-induced genes). The binding to Insig retains the SREBP-Scap complex in the ER by preventing the interaction of Scap with the COPII vesicleformation proteins Sar1, Sar23 and Sar24[70]. However, in the absence of cholesterol, the SREBP undergoes specific proteolytic cleavage that leads to its translocation to the nucleus. In the absence of cholesterol the SREBP-Scap complex is not bound to Insig and is therefore packaged in the COPII-coated transport vesicles. The SREBP-Scap complex is then transported to the golgi apparatus where it is proteolyticaly cleaved by two sequential proteases, site1 (S1P) and site 2 (S2P). The sequential cleavage releases the SREBP N-terminal transcription factor domain from the membrane. The SREBP N- terminal transcription factor domain is transported into the nucleus as a dimer by importin ß by interacting with the helix-loop-helix domain[71]. Once in the nucleus, SREBP activates the transcription of target genes involved in cholesterol and lipid metabolism such as HMG-CoA reductase, by binding to the sterol regulatory element (SRE) sequences in their promoter regions. The activation of SREBP is inhibited by feedback regulation by increased synthesis and uptake of cholesterol as well as ubiquitin-dependent proteasomal degradation.

35 27 Events iii. Regulation of HMG-CoA Reductase by Phosphorylation/Dephosphorylation HMG-CoA reductase is highly regulated by phosphorylation / dephosphorylation events at the enzyme level by AMP-activated protein kinase (AMPK) activity. The effect of AMPK on cholesterol synthesis is well characterized[72-75]. Phosphorylated AMPK regulates cholesterol synthesis by phosphorylating and inactivating HMG-CoA reductase. iv. Repression of HMG-CoA Reductase by Statin Drugs Pharmacological drugs are used to lower cholesterol levels by targeting HMG- CoA reductase. Statin drugs lower cholesterol levels by acting as a competitive inhibitor of HMG-CoA reductase. Statin drugs mimic HMG-CoA molecular structure and bind to HMG-CoA reductase, thereby inhibiting the synthesis of cholesterol in the cell. These drugs have been proven to lower total cholesterol levels; low-density lipoprotein cholesterol (LDL-C) by increasing LDL receptor synthesis; triglyceride; and reduce the occurrence of coronary events[76-79]. Statin drugs were also shown to slightly increase HDL levels[79].

36 28 e. Physiological Relevance and Significance of the Bile Acid Synthesis Pathway Type II diabetes and dyslipidemia are common metabolic disorders. Individuals with type II diabetes are at a higher risk of cardiovascular disease and have abnormal serum lipid profile. Patients with diabetes mellitus and streptozotocin-diabetic rats, show larger bile acid pools and bile acid excretion rates compared to normal individuals [2]; suggesting an association between abnormal bile acid and cholesterol biosynthesis with type II diabetes and cardiovascular disease. Metformin, one of the most commonly used drugs for the treatment of type II diabetes, lowers serum glucose levels by activating 5`AMP-activated kinase (AMPK) [80, 81]. AMPK is a key kinase involved in many metabolic pathways including but not limited to: energy metabolism, stimulation of fatty acid oxidation, ketogenesis, cellular uptake of glucose, repression of gluconeogenisis, insulin secretion, cholesterol synthesis and more recently, the repression of bile acid production. Understanding the cross talk between AMPK and these pathways will help us better understand the progression, and treatment of type II diabetes. The levels of lipoproteins are closely associated with the atherosclerotic vascular process. Elevated levels of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein AI (apo AI) in plasma indicate a low probability of coronary heart disease (CHD) together with enhanced longevity. In contrast, elevated levels of low-density lipoprotein cholesterol (LDL-C) and apo B indicate an increased risk of CHD, cerebrovascular events, other atherosclerotic diseases and death [82-84]. The discovery that plasma levels of LDL-C decreased with increasing P450 activity in the liver provides

37 29 a new avenue for fighting against atherosclerotic vascular process. P450- enzymes are important in maintaining cholesterol homeostasis by eliminating excess cholesterol. The most important P450-isoenzymes in the formation and metabolism of oxysterols in humans are: CYP7A1, CYP27A1, CYP46A1 and CYP3A4 [85]. Many pharmacological drugs indicated for the treatment of lipid disorders, such as statins, fibrates, cholestyramine and niacin; have been shown to activate genes that affect the fate of lipids [86, 87]. Recent results published in a European Journal of Clinical Pharmacology review article, 2008 [82], showed that lipid lowering drugs resulted in an activation of P450, ABCA1, and Apo AI-S as well as regression of atherosclerosis. CYP7A1 is the rate-limiting enzyme in the classical bile acid pathway and is the key P450-enzyme in maintaining cellular cholesterol levels. Disruption of the classical bile acid pathway in human patients, as a result of a mutation on the CYP7A1 gene, resulted in hypertriglyceridemia, premature gallstone disease, premature coronary disease, peripheral vascular disease, high levels of LDL, double the amount of normal hepatic cholesterol content, a markedly deficient rate of bile acid excretion, and evidence for up-regulation of the acidic bile acid pathway[88]. In-vivo experiments performed with homozygous CYP7A1 -/- mutant mice, had a very poor survival rate with 40% of the animals dieing between 1 and 4 days, and 45% between 11 and 18 days. Survival rates were increased by vitamin and cholic acid supplementations to nursing mothers at early and late developmental stages respectively. Control mice where nursing mothers were not feed cholic acid supplemented chow showed phenotypic characteristics of oily coats, hyperkeratosis, and apparent vision

38 30 defects[89]. Fully understanding the regulation mechanisms involved with P450s will provide insight on how metabolic disorders, cardiovascular disease and atherosclerosis can be prevented. f. Experimental Rationale Bile acid feedback repression of the cholesterol 7 -hydroxylase transcriptional activity has been well studied and documented[4-7, 90], but less studied is the possible post-translational control of the enzyme. The cholesterol 7 -hydroxylase mrna levels have been shown to correlate with repression by bile acids, phorbol esters, and insulin; and activated by camp, but in many studies the mrna levels of the enzyme does not always correlate with the enzymatic activity when measured. Pandak, et al., 1994[91], tested the effects of different bile salts on steady-state mrna levels and transcriptional activity of cholesterol 7 -hydroxylase. The experiments were conducted on male Sprague-Dawley rats and showed that feeding with chenodeoxycholic acid and deoxycholic acid resulted in a significant increase in cholesterol 7 -hydroxylase mrna levels with no correlating increase in enzymatic activity. Drover, et al., 2004 [92], by inducing hypo and hyperthyroidism in transgenic mice carrying the human cholesterol 7 -hydroxylase were able to show that the mrna

39 31 levels of the cholesterol 7 -hydroxylase gene was reduced 37% in hyperthyroid male mice, while the cholesterol 7 -hydroxylase enzymatic activity measured in the same group was 2-fold greater than control mice. Charles, et al., 1982 [93] showed using rat liver cholesterol 7 -hydroxylase, that treatment with NaF, an inhibitor of phosphoprotein phosphatases, increased the cholesterol 7 -hydroxylase activity by 80%, compared to treated liver with NaCl. They also demonstrated that the hydroxylase activity decreased by 40% when microsomes were incubated with bacterial alkaline phosphatase, compared to incubation with phosphatase inhibitors. These papers clearly show that the activity of cholesterol 7 -hydroxylase can also be regulated at a level other than that of transcription. It is reasonable to suggest that if bile acid synthesis is regulated by post-transcriptional events, it is most likely to do so through the regulation of the rate-limiting enzyme cholesterol 7 -hydroxylase. The activity of cholesterol 7 -hydroxylase (gpcyp7a1) has been postulated to be regulated by phosphorylation / dephosphorylation events[94, 95]. Previous data from our lab, Stroup and Ramsaran, 2005 [1] showed that cholesterol 7 -hydroxylase can be covalently modified by the addition of phosphate groups and exists as a phosphoprotein in the cell. An In-vitro kinase assays was performed on 45 synthetic 15-mer peptides based on the human cholesterol 7 -hydroxylase amino acid sequence with 12 protein kinases (AMPK, PKA, JNK, PKC, PKC I, PKC II, PKC, PKC, PKC, PKC, PKC, and PKC ) to determine whether the peptides could be modified by phosphate transfer.

40 32 Three of the 15-mer polypeptides were phosphorylated by almost all the kinase tested. Polypeptide F188/I202, which has 2 threonines at position 193 and 197, and its mutant, F188/I202B6, were phosphorylated by 9 out of 12 kinases (JNK, PKC, and PKC being the exceptions). C69/L83, which also has a threonine at position 80, was phosphorylated by 11 out of 12 kinases (all except PKA). Other peptides showed more specificity: L22/N36 and A106/N120 associated with PKA; L247/M261 and N380/M394 with PKA and AMPK; L428/T442 with JNK; and D342/L356 and I349/R364 recognized by PKC, PKC, and PKC. By purifying phosphorylated proteins from metabolically labeled HepG2 cells, and immunoblot analysis, cholesterol 7 -hydroxylase was shown to exist as a phosphoprotein in the cell. These results indicate that cholesterol 7 hydroxylase can be covalently modified by the addition of phosphate groups and exists as a phosphoprotein in the cell. Results from total bile acid assays from HepG2 cells treated with AMPK, PKC, and PKA kinase activators showed a repression in bile acid production[1]. PKC and PKA kinase activity have been shown to affect the transcriptional activity of CYP7A1, but there is no known mechanism of AMPK transcriptional repression of CYP7A1. Activation of AMPK is associated with low energy cell states and inhibits anabolic processes to prevent energy consumption while activating catabolic processes to provide cellular energy. Both cholesterol synthesis and bile acid synthesis are energy requiring processes. It is therefore reasonable to suggest that AMPK kinase activation will inhibit bile acid synthesis to conserve energy. It is also reasonable to suggest that the

41 33 mechanism by which AMPK represses bile acid production could be in the same manner that cholesterol synthesis is regulated, by phosphorylation of the rate-limiting enzyme in the pathway. g. Experimental Approach, Hypothesis and Specific Aims Cholesterol 7 -hydroxylase is the rate-limiting enzyme in bile acid biosynthesis and highly influences cholesterol homeostasis. The bile acid pathway is very important in maintaining normal cholesterol levels and disruption in the pathway can lead to hypercholesterolemia and cardiovascular diseases. Individuals with type II diabetes are at a higher risk of cardiovascular disease and show larger bile acid pools and bile acid excretion rates compared to normal individuals[2]. Transcriptional regulation of CYP7A1 has been well documented, but less studied is possible post-translational regulation of the enzyme. Previous studies have shown CYP7A1 to be phosphorylated by kinase activity and exist as a phosphoprotein in the cell. The discovery that AMPK kinase activity phosphorylates CYP7A1 and represses bile acid production suggests that another mechanisms of regulation other than transcriptional might exist. Hypothesis: Cholesterol 7 -hydroxylase enzyme activity is regulated by phosphorylation events at the enzyme level and that AMPK kinase activity regulates cholesterol 7 -hydroxylase enzymatic activity by direct phosphorylation. To test this hypothesis, the experimental design was divided into 2 specific aims.

42 34 Specific Aim 1: Characterize the Effects of Phosphorylation on the Activity of Recombinant Cholesterol 7 -hydroxylase. The hypothesis is that cholesterol 7 -hydroxylase activity changes upon phosphorylation. To test this hypothesis, E. coli expressed recombinant cholesterol 7 hydroxylase, was characterized as substrate for purified kinases. The kinases used were commercially available AMPK, JNK and PKC kinases. The activity of the phosphorylated enzyme was determined by using the phosphorylated enzyme in the cholesterol 7 -hydroxylase in-vitro reconstitution assays. The amount of 7 hydroxycholesterol produced in the assay was determined with a newly developed detection method with a HPLC/MS. Commercially available D7 7 -hydroxycholesterol was used as an internal standard to quantitatively determine the amount of 7 hydroxycholesterol produced in the assay.

43 35 Specific Aim 2: Characterize the Specific Sites of Phosphorylation Responsible for the Regulation of Cholesterol 7 -hydroxylase Enzymatic Activity. Once the effects of kinase activity on cholesterol 7 -hydroxylase enzyme activity has been determined, the specific amino acids responsible for the measured effect will be mapped. a. Selecting Possible Phosphorylation Sites Responsible for Changes in Enzyme Activity. By using the map generated from the in-vitro kinase assay on the human cholesterol 7 -hydroxylase polypeptide sequence[1], the specific amino acids that are susceptible to kinase activity will be determined. Threonine 193 and 197 (T193 and T197) and serine 80 (S80) of cholesterol 7 -hydroxylase polypeptide sequence have been shown in kinase reactions to be phosphorylated by multiple kinases. Other sites showed more specificity to the different kinases. After the in-vitro reconstituted cholesterol 7 -hydroxylase enzymatic assay with the different kinases are performed, the phosphorylation sites that corresponds with the kinase activity that affected CYP7A1 enzymatic activity will be mutated.

44 36 b. Determining the Effects of the Mutated Sites on CYP7A1 Enzymatic Activity. The effects that the mutations have on phophorylated cholesterol 7 -hydroxylase enzymatic activity will be determined in the in-vitro reconstituted enzymatic assay. This study will answer the question of whether a post-transcriptional mechanism of CYP7A1 regulation exists. It has long been hypothesized that cholesterol synthesis and bile acid synthesis are co-regulated; the effects that AMPK has on bile acid synthesis will be able to address the co-regulation between the two pathways. AMPK kinase activity is involved in many cellular metabolic pathways, this study will also provide information about the cross-talk between cholesterol synthesis, bile acid synthesis, and energy metabolism pathways in the cell.

45 CHAPTER II MATERIALS AND METHODS PART 1: Cholesterol 7 -hydroxylase Specific Antibodies a. Rationale for Cholesterol 7 -hydroxylase Antibody Production Antibodies for cholesterol 7 -hydroxylase are commercially available (Santa Cruz Biotechnology, Inc. CYP7A1 (H-58): sc-25536), but did not show strong affinity or specificity for the antigen when tested by immunoblot analysis. In order to identify E. coli-expressed recombinant cholesterol 7 -hydroxylase antigens, anti-cyp7a1 antibodies were developed. b. Antigen (C2) used for CYP7A1 Antibody Production For CYP7A1 polyclonal antibody production, a 20.3 Kb polypeptide fragment (C2) was generated as antigen for immunization. The C2 nucleic acid sequence was cloned into the Roche pivex2.4b Nde vector. The expression vector is under the control of the T7 promoter and has an N-terminal linker 6X His-tag for purification. The C2 polypeptide is 175 amino acids long 37

46 38 corresponding to N148-G323, which is located at the central region of the 504aa CYP7A1 polypeptide sequence. The large antigen used for our antibody production is an improvement over the antigen that was used for the production of the commercially available antibody. The commercial antibody was raised against a smaller peptide corresponding to amino acids The larger C2 antigen provides more epitopes for antibody generation, and is located in the central region of the enzyme. The location of the epitopes will allow detection of both full-length and degraded E. coli-expressed recombinant CYP7A1, with the goal of developing affinity purified antibody prep with higher affinity/avidity and in sufficient quantity for the project. c. Sub-cloning of the C2 cdna Sequence into E. coli-expression Vector The C2 pivex2.4b Nde vector construct was transformed into the E. coli DH5 cell line, and plated on 15% agar LB-antibiotic plates with 50mg/ml ampicillin. As a negative control, the DH5 E. coli cell line was transformed with the pivex2.4b Nde vector construct only. Transformation procedure was performed as described on page 126. Plates with transformed cells were incubated at 37 o C overnight. Colonies were selected from transformed plates and inoculated into 2ml LB + ampicillin broth, and grown overnight. DNA mini plasmid prep isolation was performed on 1ml of the cultured sample, and isolated DNA was separated by electrophoresis on a 1% agarose gel. Cultures that yielded plasmids that migrated slower than empty vector on agarose gels, were streak plated and incubated overnight at 37 o C in order to isolate single

47 39 colonies. The colonies were inoculated in 2ml LB + ampicillin overnight, and mini plasmid preps were performed on the samples. The plasmid DNA isolated was separated by electrophoresis on a 1% agarose gel to confirm plasmid size. Plasmid samples that were positive for fragment insertion were restriction mapped by digesting with restriction enzymes flanking the insert. The restriction-digested samples were separated by electrophoresis on a 1% agarose gel to confirm insert size. Freezer stocks were made of samples that had the appropriate fragment size, and stored at -70 o C. Procedures for freezer stock preparation are described in the material and methods section. Positive samples were grown in a larger volume of LB + 50mg/ml ampicillin overnight, and plasmid preparation was performed using the Nucleobond DNA purification kit from Clontech (Palo Alto, CA). The plasmid prep kit provided a higher concentration of DNA plasmid that was clean from contaminants. This sample was sequenced to confirm that the inserted fragment was in-frame with the vector-construct; and no unwanted mutations were made to the sequence. The C2 cdna sequencing was performed by the Ohio State University Plant-Microbe Genomics Facility using T7 forward (T7P) and T7 reverse (T7T) primers. Contiguous alignment of the sequence was generated using both Map Vector 2.3 and ClustalW (1.83) multiple sequence alignment programs [Fig. 5]. The sequence alignment showed that the C2 fragment was in-frame with the vector initiation site, and no unwanted mutations were made to the DNA sequence.

48 40 Figure 5. ClustalW (1.83) Multiple Sequence Alignment of C2 and Full-length CYP7A1 C2 sequence alignment with full-length CYP7A1 cdna sequence. C2 sequencing was performed using T7 promoter and T7 terminator primers provided by sequencing company. * Indicates consensus sequences between the nucleic acid sequence from full-length CYP7A1 cdna sequence and sequence results from the C2 fragment. A 100% homology was obtained from the multiple sequence alignments.

49 41 Figure 5. ClustalW (1.83) Multiple Sequence Alignment of C2 and Full-length CYP7A1

50 42 d. Expression of the C2 Antigen For the expression of the C2 fragment, the vector-construct was transformed into the E. coli BL21 (DE3) expression system. The BL21 (DE3) cell line allows for high protein expression of vector constructs under the control of the T7 promoter and have a ribosome-binding site. The BL21 (DE3) cell line contains a T7 bacteriophage gene that encodes for T7 RNA polymerase under the control of the lac operon. Expression of the gene of interest can be induced by the addition of isopropyl -D-1-thiogalactopyranoside (IPTG), which will trigger the transcription of the T7 RNA polymerase and promote transcription of the gene of interest. i. Expression of C2 with BL21 (DE3) E. coli Cell Line An expression time course, using different concentrations of IPTG, was performed to determine the appropriate parameters for C2 expression. Figure 6 is the results from the immunoblot analysis of the IPTG induced expression of the C2 polypeptide. Fig. 6A is the induction of the C2 pivex 2.4b Nde construct with 1mM IPTG for 0, 2, and 4 hrs, and Fig. 6B is the induction of the C2 pivex 2.4b Nde construct with 2.5mM IPTG for 0, 2, 4, and 6 hrs. The immunoblot detection was performed with anti-his antibodies. The results showed that 2.5mM of IPTG was capable of inducing expressing of the C2 fragment. Expression proceeded for 4hrs of induction and decreased at the 6 hrs time point. The expression of the C2 fragment was

51 43 very temperamental as 0 time point, with no IPTG, showed leaky expression in gel B, but there was no detection in gel A. Scaling up the expression did not produce substantial C2 antigen for antibody production.

52 44 Figure 6. Western Blot Analysis of In-vitro Expressed C2 Antigen for Antibody Production. 6A: Induction of C2 pivex 2.4b Nde BL21 (DE3) vector construct with 1 mm IPTG for 0, 2, and 4 hrs, corresponding to lanes 2, 3 and 4 respectively. 6B: Induction of C2 pivex 2.4b Nde BL21 (DE3) vector construct with 2.5 mm IPTG for 0, 2, 4, and 6 hrs, corresponding to lanes 6, 7, 8 and 9 respectively. Lanes 1 and 5 are pre-stained low molecular weight markers. Western blot was performed with anti-his antibodies.

53 45 KD A B Figure 6. Western Blot Analysis of In-vitro Expressed C2 Antigen for Antibody Production.

54 46 ii. Expression of C2 with the Roche RTS 500 E. coli HY kit The Roche RTS 500 E. coli HY cell free system was used to improve expression of the C2 antigen. The RTS 500 is a coupled transcription/translation reaction, which utilizes the T7 RNA polymerase. The transcription and translation machinery that are used in the expression system are present in the E.coli lysate provided. The expression system consists of two chambers, a 1ml chamber that serves as the reaction compartment were expression takes and a feeding compartment that supplies the necessary amino acids, nucleotides and energy substrates. A semi permeable dialysis membrane is used to separate the two chambers and it keeps the waste components at a minimum in the reaction chamber. With this system, the C2 antigen was expressed to a maximum of 5 mg per ml at 20 hrs of expression [Fig. 7]. The C2 RTS 500 expression system produced substantial amounts of C2 that was used for antibody production.

55 47 Figure 7. RTS 500-expressed C2 Antigen for Antibody Production. In-vitro expression of C2 in the RTS 500 E. coli expression system for 0, 2, and 20 hrs corresponding to lanes 1, 2, and 3 respectively. Western blot was performed with anti-his primary antibodies at a 1: 6,000 dilution and goat-antimouse-hrp secondary at 1:10,000 dilutions. Solid Arrow on the right indicates C2 antigen detection.

56 48 KD Figure 7. RTS 500-expressed C2 Antigen for Antibody Production.

57 49 e. Immunization for Antibody Production Two New Zealand White Female Rabbits, tagged and 20966, were used for antibody production. Pocono Rabbit Farm & Laboratory Inc. (PRF&L), located in Canadensis, PA, performed the rabbit s Immunizations μg of the C2 antigen was used for each immunization. The timeline and procedures for the immunizations are outlined in table 1. Rabbits and were maintained on a four-week cycle of immunizations with the C2 antigen. A small bleed of 3-5ml were collected from the rabbits and characterized for production of C2 specific polyclonal antibodies.

58 50 Day 0 Day 14 Day 28 Day 42 Day 44 Day 56 Day 70 Day 72 Day 84 Small preimmune bleed (3-5ml sera). Immunize with μg/rabbit antigen in complete freund s adjuvant (CFA) intradermally. Immunize with μg/rabbit antigen in incomplete freund s adjuvant (IFA) intradermally. Immunize with μg/rabbit antigen in IFA subcutaneously Small bleed 3-5ml sera Receive small bleeds from preimmune and immune serum Immunize with μg/rabbit antigen in IFA subcutaneous Small bleed (3-5ml sera), continuing every 28 days Received small bleed Immunize with μg/rabbit antigen in IFA subcutaneous and continuing every 28 days. Table 1. Polyclonal Antibody Production Schedule Table 1 shows the timeline and procedure for rabbit immunization μg of the C2 antigen was used for each immunization and subsequent antibody boosts. The rabbits are maintained on a four-week cycle of immunizations and small bleeds, until antibody specificity for C2 is determined.

59 51 f. Characterization of Anti-serum from Immunized Rabbits The serum was characterized for each rabbit small bleed after the immunization cycle was completed (refer to Table 1). For the rabbit serum characterization, serum from rabbits and were used as primary antibodies (dilutions from 1:500 to 1:20,000), with goat anti-rabbit-hrp secondary antibodies (1:10,000) for western blot analysis of the RTS 500-expressed C2 antigen [Fig. 8 and Fig. 9]. In figure 8, the C2 antigen sample was overloaded; necessary adjustments were made on subsequent western blots. Figure 8, Lane 1 and 6 are western blot analysis performed with pre-immune serum as the primary antibody from rabbits and respectively. Dilution used for the pre-immune serum was 1:500. Western blot showed that the serum had no specificity for the C2 antigen prior to immunization of the rabbits. Lanes 2-5, and lanes 7-10, is the western blots that were performed with serum from the immunized rabbits and respectively. From left right, dilutions of the serum used for the western blot analysis are 1:100, 1:500, 1:1,000, and 1:5,000. Arrow on the right side of the gel indicates C2 migration. Figure 9 is the western blot analysis of the C2 antigen with rabbits and serum after two months of immunization. Lanes 1-4, from the left right, rabbit Pre-immune serum (1:500), immunized serum, 1:1,000, 1:5,000, 1:10,000 dilutions. Lanes 5-8, from left right, rabbit pre-immune serum (1:500), followed by immunized serum from rabbit in the same dilution order as the previous rabbit. The serum from the immunized rabbits showed strong affinity for the C2 polypeptide even at a 1:10,000 dilution. Indicating that the rabbits were producing

60 52 antibodies against the C2 antigen. The serum also showed affinity for non-specific E. coli proteins, which were present in the RTS 500 C2 assay sample used for immunization. The following experiments were performed to confirm the presence of C2 specific antibodies in the rabbit serum.

61 53 Figure 8. Western Blot of C2 Samples with Serum from the First Small Bleed of Immunized rabbits Lanes 1-5 from left right, Pre-immune (1:500 dilution) lane1, immunized rabbit serum lanes 2-5 are 1:100, 1:500, 1:1,000, 1:5,000 dilutions. Lanes 6-10 is for rabbit in the same order with lane 6 being the pre-immune serum. Arrow on the right indicates C2 migration.

62 54 KD Figure 8. Western Blot of C2 Samples with Serum from the First Small Bleed of Immunized rabbits

63 55 Figure 9. Western Blot of C2 Samples with Serum After Two Months of Immunization Lanes 1-4 from left right, Pre-immune (1:500 dilution) lane1, immunized rabbit serum lanes 2-4 are 1:1,000, 1:5,000, 1:10,000 dilutions. Lanes 5-8 is for rabbit in the same order with lane 5 being the pre-immune serum. Arrow on the right indicates C2 migration.

64 56 KD Figure 9. Western Blot of C2 Samples with Serum After Two Months of Immunization

65 57 g. Pre-absorption of Rabbit Serum with E. coli Lysate In order to determine whether the serum contained antibodies specific to C2, and that the band detected in the western blot analysis was not a co-migrating E. coli protein, the rabbit serum was pre-absorbed with E. coli lysate from the RTS cell free system. The rationale for this experiment is: the E. coli lysate will absorb any E. coli specific antibodies present in the serum, but will not have an effect on C2 specific antibodies. The serum for rabbits and were pre-incubated with the RTS 500 E. coli lysate, for 15 min in TTBS buffer. The pre-absorbed serum was used as the primary antibody, in a western blot analysis of the C2 antigen prepared with the RTS 500 assay [fig. 10]. The experiment showed that the pre-incubation of the serum with E. coli lysate, pre-absorbed the E. coli specific antibodies. Thereby, diminished the specificity of the serum to the E. coli proteins. Fig. 10 shows the band intensity of the E. coli proteins diminish in the western blot analysis when comparing complete serum detection lane 1 and 3 to pre-absorbed serum detection lanes 2 and 4, for rabbits and respectively. The E. coli antibody pre-absorption had no effect on the specificity and affinity of the serum to the C2 polypeptide. The band intensity of the C2 antigen remained the same, when comparing complete serum to pre-absorbed serum western blots. This indicates that the serum contains antibodies that react to the C2 polypeptide, and the band observed is not a co-migrating E. coli protein.

66 Figure 10. Western Blot Analysis of C2 Antigen with Pre-absorbed Rabbit Serum The rabbit serum was pre-absorbed with E. coli lysate prior to using it as the primary antibody for C2 RTS samples at a dilution of 1:10,000. Lanes 1 and 2 are un-absorbed and pre-absorbed antibody for rabbit Arrow indicates migration of the C2 polypeptide.

67 59 h. Antibody Characterization with Purified C2 Antigen To further confirm that the rabbit serum contained antibodies specific for the C2 polypeptide, the serum was tested against purified C2 antigen. The C2 antigen was purified as described in the protein purification section of this chapter. The serum was used for a western blot analysis of the purified C2 antigen. Western blot analysis of the purified and unpurified C2 antigen was also performed with anti-his antibody as a positive control. Figure 11 shows the results from the western blot analysis of unpurified C2 antigen (lanes 1, 3, and 5) and purified C2 antigen (lanes 2, 4, and 6). Primary antibodies used were: anti-his antibodies for lanes 1 and 2 at a concentration of 1:3,000, lanes 3 and 4 was with Rabbit serum and lanes 5 and 6 was with rabbit serum, both at a concentration of 1:10,000. The western blot analysis shows strong affinity and specificity of the serum to the purified C2 antigen. The experiment provided additional evidence that the serum is capable of reacting with the C2 polypeptide. The positive bands on the anti-his western blot shows that the isolated protein was fused with the 6x His-tag, and is therefore a product from the vector construct. The blot with the two rabbit serum (20966 and 20967) showed that the same peptide reacts with the rabbit serum, confirming that the serum contains anti-c2 antibodies.

68 60 Figure 11. Western Blot Analysis of Purified C2 Western blots of unpurified C2 (lanes 1, 3, and 5) and purified C2 (lanes 2, 4, and 6). Primary antibodies used were: Anti-His antibody for lanes 1 and 2 at a concentration of 1:3,000, lanes 3 and 4 was with Rabbit serum and lanes 5 and 6 were with rabbit serum, both at a concentrations of 1:10,000.

69 61 KD Figure 11. Western Blot Analysis of Purified C2

70 62 i. Antibody Affinity Purification: i. C2 Affinity Column Preparation In order to purify C2 antibodies from the rabbit serum, a C2 affinity column was prepared using a cyanogen bromide activated matrices. The cyanogen bromide reacts with hydroxyl groups on agarose in alkaline solutions, to form very active cyanate esters, and a small portion of slightly active imidocarbonates. These groups react with the primary amines of the ligand at very mild conditions, forming a covalent coupling of the ligand to the agarose matrix. The purified C2 polypeptide was dialyzed against cyanogen-bromide activated matrix coupling buffer (0.1M NaHCO 3 containing 0.5M NaCl, ph ). The cyanogen-bromide activated resin was washed and swelled in cold 1 mm HCl for 30 minutes with continuous buffer exchange. The resin was then washed in distilled water, 5-10 column volumes, and then washed in the coupling buffer (5 ml per gram dry gel). After adding the coupling buffer, the purified C2 polypeptide was immediately added (5-10mg protein per ml of gel) and rocked at room temperature for 2 hours. Un-reacted ligands were washed away with two washes using the coupling buffer at a volume that is two times the column volume. The un-reacted groups on the resin were blocked with 0.2 M glycine, ph 8.0 for 2 hours at room temperature. The column was washed extensively to remove the blocking solution, first with basic coupling buffer at ph ~ 8.5, then with acetate buffer (0.1 M, ph4) containing NaCL (0.5 M). The wash cycle of high and low

71 63 ph buffer was completed five times and the C2 column was stored in 1.0 M NaCL at 4 o C with 0.05% of a suitable bacteriostat. j. C2 Antibody Affinity Purification The C2 cyanogen-bromide activated matrix column was first washed twice with distilled water. The resin was transferred into a 50 ml sterol tube and washed five times with 1X TBS (tris-buffered saline, 20 mm tris 50 mm NaCL, Ph 7.5). The resin was then washed two times with 20 ml of 100 mm glycine HCL/150 mm NaCL, followed by two washes with 20 ml of 1X TBS. Serum was spun down at 3K RPM for 1 Min to remove precipitation and then added to the equilibrated C2 column for binding. The column was mixed gently for 30 min by gently rolling and allowing settling before rolling again. The column was then settled and the serum was allowed to flow through. The column was then washed with 100-times the resin volume with 1x TBS. The antibodies were eluted with 10 ml of 100 mm glycine HCL ph 2.4/ 150 mm NaCL and 1 ml fractions were collected in eppendorf tubes containing 200 μl of 1M tris ph 9 to neutralize the eluted samples. The protein concentration was determined using a BCA (bicinchoninic acid) protein assay and the apparent molecular weight of the eluted protein was determined by electrophoresis on a 10% SDS polyacrylamide gel, and stained with coomassie blue [Fig. 12]. The eluted fractions peaked at fraction 3. The band noticed indicated the heavy chain of the eluted antibody. The affinity of the purified antibodies was determined by

72 64 western blot analysis of purified C2. The western blot analysis showed that the purified antibodies had strong affinity and specificity for the purified C2 antigen [Fig. 13].

73 65 Figure 12. Coomassie Blue Staining of C2 Polyclonal Antibody Purification Lanes 1-8 is collected fractions from the C2 cyanogens-bromide activated matrixes column. Coomassie stain of eluted fractions separated on a 10% SDS- PAGE. Fractions peaked at fraction 3.

74 KD Figure 12. Coomassie Blue Staining of Purified C2 Polyclonal Antibody

75 KD Figure 13. Western Blot Analysis with Affinity Purified C2 Polyclonal Antibodies Affinity purified polyclonal antibodies eluted from the C2 cyanogen-bromide activated matrix column were used for a western blot analysis of purified C2 polypeptide. The antibodies showed strong specificity for the C2 antigen

76 68 PART 2: Expression of Recombinant CYP7A1 in E. coli a. History of CYP7A1 Expression The difficulties with expressing cholesterol 7 -hydroxylase have been well documented. The low levels of CYP7A1 expression in COS cells has been reported[96, 97]. Expression of recombinant CYP7A1 in E. coli cell lines has also been a challenge. The enzyme has been successfully expressed in E. coli cells, but the levels of expression are low. It has been suggested that the membrane-binding nature of CYP7A1, make it difficult to express in E. coli cell, due to the lack of an internal membrane system for the enzyme to insert into[98]. b. Sub-cloning WT CYP7A1 cdna into E. coli-expression Vector To overcome the difficulties in CYP7A1 in-vitro expression, the invitrogen ptrc- His2b expression vector was chosen to express CYP7A1 recombinant protein in E.coli Top 10 cells. The ptrc-his2b vector was chosen because of its advantageous features. It is not under the control of the T7 promoter but instead a Trc promoter (a hybrid of the trp and lac promoters for high-level expression of fusion proteins); it also contained a mini cistron, and a re-initiation ribosome binding site up-stream of the transcription start site. The mini-cistron loads multiple ribosomes, and brings them in close proximity to the transcription start site. Because the expression vector is not under the control of the T7 promoter, the plasmids were amplified and expressed in E. coli cell lines with high

77 69 transformation efficiencies (eg. DH5 and Top 10 cell lines). The vector also has a c- terminal 6X His-tag for purification of in-frame recombinant protein. The full-length cdna sequence of CYP7A1 was sub-cloned into the ptrc-his2b vector using 5`GGGACCATGGCTACCACATCTTTGATTTGG-`3 (ptrc-5`[ncoi]-f) forward and 5`GGGTAGAAAGCTTCGCAAATGCTTGAATTTATA-`3(pTrc- 3`[HindIII]-R) reverse primers. E. Coli Top 10 cells were transformed with either the ptrc-his2b vector only, or vector sub-cloned with the CYP7A1 cdna sequence. Expression of the CYP7A1 recombinant protein was performed as described in the materials and methods section. Isolated protein samples were separated by electrophoresis on a 12.5% polyacrylamide SDS-PAGE gel, and transferred onto PVDF transfer membrane for immunoblot assay detection. Both anti-his and anti-cyp7a1 antibodies were used to confirm protein expression off the plasmid construct [Fig. 14].

78 70 Figure 14. Western Blot of E. coli-expressed WT CYP7A1 Recombinant Protein Lane 1-pre-stained low molecular weight markers. Lane 2- C2 His tagged positive control. Lane 3- E. coli expressed vector only negative control. Lane 4- E. coli expressed CYP7A1 recombinant protein. Fig. 14A and 14B are immunoblot analysis with anti-his and anti-cyp7a1 antibodies respectively.

79 71 KD A c2 C WT KD B c2 C WT Figure 14. Western Blot of E. coli-expressed WT CYP7A1 Recombinant Protein

80 72 c. Characterization of Recombinant CYP7A1 Protein Figure 14 shows the immunoblot analysis of recombinant protein expression from the CYP7A1 cdna sequence. Both gels were loaded in the same order with equal amounts of total protein, using BSA as the mass standard. Lane 1 is a pre-stained low molecular weight marker. Lane 2 is the positive control for the western blot analysis, using the C2 His-tagged polypeptide that was previously described in the production of anti-cyp7a1 antibodies section. Lane 3 is the negative control, using expressed proteins from the E. coli Top 10 cells transformed with vector only construct. Lane 4 is the expressed sample from the CYP7A1 cdna vector construct. Immunoblot analysis of the E. coli Top 10 cells transformed with the CYP7A1- ptrc-his2b vector construct, showed positive detection of expressed proteins. In contrast, the vector-only construct showed no detectable bands. This indicated that the observed proteins expressed were from the CYP7A1 cdna construct. The positive bands detected from the CYP7A1 cdna construct with the anti-his antibodies, confirm that the expressed protein was in-frame with the c-terminal 6X His-tag. Multiple bands were detected in the CYP7A1 sample. The approximate molecular weight for the full-length CYP7A1 and the linker-his tag was determined with Science Gateway molecular weight calculator to be 60KD. Five high molecular weight bands corresponding to 60, 49, 47.6, 46.4, and 44.5 KD, and a predominant 30KD fragment, were detected in the immunoblot analysis of recombinant protein expressed off the CYP7A1 cdna vector construct. The approximate molecular weights of the detected

81 73 bands were determined by using the Kodak 1D imaging software. The bands from the molecular weight markers were used to determine the approximate molecular weights of the detected bands. The higher molecular weight bands, from 49 to 44.5 KD, were attributed to protein degradation because of the laddering effect, and the positive detection with the anti-his and anti-cyp7a1 antibodies. The positive detection with the anti-cyp7a1 antibodies, confirmed that the expressed proteins had the same specific epitopes as the C2 polypeptide that was used to generate the anti-cyp7a1 antibodies. The predominant 30KD fragment was not detected in the anti-cyp7a1 immunoblot assay, but was detected in the anti-his immunoblot assay. The detection with the anti-his antibody indicated that the expressed 30KD fragment was in frame with the CYP7A1 full-length cdna construct. The fact that the fragment could have been a product of degradation was considered, but this hypothesis was not consistent with the observed results. If the 30KD polypeptide were a product of degradation, there would be expected laddering of various molecular weights migrating faster and slower than the prominent band, as seen in the higher molecular weight bands. The two other explanations could be; the 30KD fragment is a result of a single proteolytic cleavage, resulting in two bands of the same intensity and molecular weight; secondly, the 30KD fragment is a result of an internal start in the CYP7A1 cdna sequence.

82 74 d. Characterization of Expressed 30KD Fragment Manual sequence analysis of the CYP7A1 cdna sequence revealed a sequence with homology to an E. coli consensus ribosome binding site located between nucleic acids T965 to A970, approximately in the middle of the cdna sequence. The presence of an internal start site was further confirmed by CYP7A1 cdna sequence analysis with the Gene Locator and Interpolated Markov ModelER (GLIMMER) software. The GLIMMER software was also used to predict the efficiency of the open reading frames (ORF). The efficiency of the ORF as determined by the GLIMMER software, starting at amino acids 965, was 1.44 times greater than that of the full-length CYP7A1 cdna sequence. The higher efficiency of the internal start site would explain the approximate 1:2 ratio in expression between the higher molecular weight bands compared to the 30KD fragment. The ratios between the expressed proteins were determined by comparing triplicate densitometry measurements of the higher molecular weight bands to the 30KD fragment from anti-his western blot analysis. Method for determining specific protein amounts using the densitometry measurements are described in the appendix experimental protocols. An internal start site from amino acids 965 would result in a fragment approximately the same size as the band observed in the western blot. This was determined by using a molecular weight calculator. To confirm that the predominant 30KD fragment was a result of an internal start site, the polypeptide was isolated and sequenced. The 30 KD fragment was purified with a Ni-NTA Purification system and separated by electrophoresis on a 12.5% SDS-PAGE gel. The gel was stained and the

83 75 30KD fragment was cut out and sequenced by The Ohio State, Plant-Microbe Genomics Facility. The sequencing result from the 30KD fragment was aligned with the CYP7A1 full-length protein sequence and resulted in a 964-point identity [Fig.15]. The sequencing results confirmed that the 30KD fragment was a truncated peptide from the CYP7A1 polypeptide and also confirmed that is was a result of an internal start from the CYP7A1 cdna sequence. The immunoblot with the anti-cyp7a1 antibodies did not detect the 30KD fragment. The 30KD fragment was not detected in the immunoblot with the anti- CYP7A1 antibody because of the lack of specific epitopes. The anti-cyp7a1 antibodies were produced with the C2 antigen as described in the material and methods section. Protein alignment of the C2 polypeptide and the 30KD fragment showed only a 16 amino acid overlap between the two polypeptides [Fig.16].

84 76 1 MMTTSLIWGI AIAACCCLWL ILGIRRRQTG EPPLENGLIP YLGCALQFGA 51 NPLEFLRANQ RKHGHVFTCK LMGKYVHFIT NPLSYHKVLC HGKYFDWKKF 101 HFATSAKAFG HRSIDPMDGN TTENINDTFI KTLQGHALNS LTESMMENLQ 151 RIMRPPVSSN SKTAAWVTEG MYSFCYRVMF EAGYLTIFGR DLTRRDTQKA 201 HILNNLDNFK QFDKVFPALV AGLPIHMFRT AHNAREKLAE SLRHENLQKR 251 ESISELISLR MFLNDTLSTF DDLEKAKTHL VVLWASQANT IPATFWSLFQ 301 MIRNPEAMKA ATEEVKRTLE NAGQKVSLEG NPICLSQAEL NDLPVLDSII 351 KESLRLSSAS LNIRTAKEDF TLHLEDGSYN IRKDDIIALY PQLMHLDPEI 401 YPDPLTFKYD RYLDENGKTK TTFYCNGLKL KYYYMPFGSG ATICPGRLFA 451 IHEIKQFLIL MLSYFELELI EGQAKCPPLD QSRAGLGILP PLNDIEFKYK 501 FKHL Figure 15. Protein Sequence Alignment of Human Cytochrome P450 7A1 (CYP7A1) and the 30KD Isolated Fragment Amino acids represent the full-length protein sequence of CYP7A1. Bold red lettering represents positive sequence alignment between WT CYP7A1 and the 30KD fragment. The alignment match score was 964.

85 77 Figure 16. Protein Alignment of the C2 Polypeptide and the 30KD Fragment Sequence alignment of the C2 polypeptide used for anti-cyp7a1 antibody production and the 30KD fragment from in-vitro expressed recombinant CYP7A1 revealed a 16 amino acid overlap. The bold underline represents the sequence identity between the two polypeptides.

86 Figure 16. Protein Alignment of the C2 Polypeptide and the 30KD Fragment 78

87 79 To farther confirm that the 30KD fragment was in-fact a product from an internal start, the CYP7A1 cdna sequence was truncated at the N-terminus and sub-cloned into the ptrc-his2b vector. 729 nucleotides were deleted from the N-terminus. The N- terminus was truncated out-of-frame from the WT CYP7A1 cdna reading frame. The purpose of truncating the CYP7A1 cdna sequence out-of-frame was to insure that the CYP7A1 cdna sequence was not read in-frame with the linker 6x His tag. If the construct resulted in the synthesis of a polypeptide from the vectors initiation site, it would result in a polypeptide that is out-of-frame with the 6X linker His-tag, and therefore will not be detected with the anti-his antibodies. If the 30KD fragment is a result from an internal start, expression should proceed from the AUG down stream of the ribosome-like binding site, and the expressed polypeptide will be in-frame with the 6X linker. Figure 17 shows the immunoblot analysis from the expression of the CYP7A1 full length and truncated ( 1-729) vector constructs. Lane 1 is the pre-stained low molecular weight marker, lanes 2 and 3 are the recombinant proteins expressed from the full length and truncated ( 1-729) CYP7A1 cdna vector constructs respectively. Immunoblot analysis was performed with anti-his antibodies. The immunoblot shows the detection of the 30KD fragment in both the full length and truncated constructs. This result provided additional evidence that the 30KD fragment was a product of an internal start. The immunoblot also provides evidence that the higher molecular weight polypeptides that are detected in the CYP7A1 full-length vector construct are products from the CYP7A1 full-length cdna sequence.

88 80 Figure 17. Western Blot of E. coli-expressed WT CYP7A1 and Truncated CYP7A1 Recombinant Protein Lane 1-pre-stained low molecular weight marker. Lane 2- E. Coli expressed WT CYP7A1 recombinant protein. Lane 3- E. Coli expressed truncated ( 1-729) CYP7A1. Immunoblot detection was performed with anti-his antibodies. Arrow labeled A shows the expression of the higher molecular weight proteins from the CYP7A1 fulllength construct. Arrow labeled B shows the expression of the 30KD fragment from the CYP7A1 full-length construct and the truncated ( 1-729) CYP7A1construct.

89 81 KD WT WT A B Figure 17. Western Blot of E. coli-expressed WT CYP7A1 and Truncated CYP7A1 Recombinant Protein

90 82 PART 3: CYP7A1 Enzymatic Assay a. History of CYP7A1 Assay Detection Methods The difficulty in assaying the enzyme activity of cholesterol 7 -hydroxylase has been well documented [98, 99]. Problems arising from the low levels of the cholesterol 7 -hydroxylase enzyme peptide and activity, compounded by issues of sensitivity of detecting the end product in the enzyme assay. Recent techniques for quantifying CYP7A1 enzymatic activity involves the quantification of 7 -hydroxy-4-cholesten-3-one (C4), the product of the next oxidative enzymatic reaction after CYP7A1 in the classical bile acid synthesis pathway[19]. It has been reported that serum concentration of C4 and 7 -hydroxycholesterol, the immediate product from CYP7A1 enzyme activity, reflects both CYP7A1 enzymatic activity and bile acid synthesis in humans [19, 100, 101]. Because of the low detection sensitivity of 7 -hydroxycholesterol, C4 has been the intermediate of choice for characterizing the enzyme activity of cholesterol 7 hydroxylase. Gälman, et al.[102], measured CYP7A1 enzymatic activity by monitoring C4, using the conventional HPLC-ultraviolet method. With their technique, they were able to separate C4 from peripheral blood and used it to quantify the activity of cholesterol 7 hydroxylase. Akira, et al.[19], developed a new technique to quantify serum C4 by liquid chromatography-tandem mass spectrometry (LC-MS/MS). In their method, C4 was derivated into the picolinoyl ester (C4-7 -picolinate), and then purified using a C18 cartridge. The derivation made the C4 picolinoyl ester more sensitive (with a reported

91 83 detection limit of 100ƒg) than the detection limits of C4 with a conventional HPLCultraviolet method. b. CYP7A1 Assay Detection Method A new detection method, using HPLC-MS, was developed in this study to quantify cholesterol 7 -hydroxylase enzyme activity by directly measuring 7 hydroxycholesterol. Measuring 7 -hydroxycholesterol will be a direct measurement of cholesterol 7 -hydroxylase enzyme activity. 7 -hydroxycholesterol is the immediate product from the enzymatic reaction of CYP7A1 activity. The MS used in the assay, uses an orthogonal atmospheric pressure chemical ionization source (APCI) that improves sensitivity and reproducibility over a wide range of liquid chromatography (LC) operating conditions. The detection limit of the new method was evaluated by analyzing commercially available 7 -hydroxycholesterol (Steraliods, Inc. Newport, RI) that was diluted stepwise and analyzed in quadruplicates. The detection was linear down to 0.4 moles of 7 hydroxycholesterol with an R 2 = [Fig. 18a and 18b]. Figure 18a shows the complete standard curve for 7 -hydroxycholesterol, and figure 18b shows the extracted linear portion of the standard curve. The assay was shown to be just as sensitive for 7 hydroxycholesterol, as the detection reports of C4 by Gälman, et al. This method eliminates, the derivation steps required to detect C4 in the LC MS/MS method presented by Akira, et al., and the purification steps required for detecting C4 with the conventional

92 84 HPLC-ultraviolet method. By eliminating these steps, the new method is less time consuming, and less expensive than the previously proposed methods. c. Internal Standard for Enzyme Assay For the quantification of 7 -hydroxycholesterol, a stable isotopic form of 7 hydroxycholesterol is needed as an internal standard. Deuterium ( 2 H or D) compounds have been used as an internal standard for quantifying C4[19]. 7-Deuterated ( 2 H 7 ) 7 hydroxycholesterol (D7), is commercially available from Steraloids inc. Newport, RI, and was used as the internal standard for quantifying CYP7A1 enzymatic activity. An alternative to purchasing D7 was to perform a hydrogen/deuterium exchange on exchangeable hydrogens of 7 -hydroxycholestrol. Exchangeable hydrogens are those bond to nitrogen, oxygen, and sulfer[103]. In the case of 7 -hydroxycholesterol, the hydroxyl groups will exchange its hydrogen with the deuterium. Unlike the commercially available D7, which the deuterium is covalently linked to the hydrocarbon ring (C- 2 H bond), the hydrogen/deuterium exchange will replace hydrogen atoms on the hydroxyl-groups of 7 -hydroxycholesterol (O- 2 H). The hydroxyl group is a commonly lost group during mass spectrometry; therefore, the O- 2 H 7 -hydroxycholesterol will not make a good internal standard. Upon fragmentation, the O- 2 H group will be lost, and the internal standard will not be distinguishable from the assay produced 7 -hydroxycholesterol.

93 85 The detection sensitivity of the internal standard was analyzed by co-injecting triplicate samples of the commercially available 7 -hydroxycholesterol and D7 in stepwise dilutions [fig. 19]. The internal standard was determined to elute at the same time as 7 -hydroxycholesterol when injected through the HPLC-MS equipped with a C18 column, and had a molecular weight of 392 m/z. The concentration of D7 was normalized so the relative intensity between the D7 internal standard and 7 hydroxycholesterol were comparable. The co-injected samples had the same slope, and R 2 values of 1 and , for D7 and 7 -hydroxycholesterol respectively. Because of the sensitivity of the MS to the internal standard; ƒmoles quantities of the D7 compound was sufficient for the quantification of assay produced 7 -hydroxycholesterol.

94 86 Figure 18a. Complete Standard Curve of 7 -hydroxycholesterol using the HPLC-MS Graph representation of intensity verses concentration ( moles) of commercially purchased 7 -hydroxycholesterol. Extracted portion of the graph is enlarged in figure 18b. Detection limits of the HPLC-MS for 7 -hydroxycholesterol was linear down to 0.4 moles at 385 m/z with an R 2 value of

95 87 Figure 18a. Complete Standard Curve of 7 -hydroxycholesterol using the HPLC- MS

96 88 Figure 18b. Extracted Portion of the Standard Curve for 7 -hydroxycholesterol using the HPLC-MS

97 89 Figure Deuterated ( 2 H 7 ) 7 -hydroxycholesterol (D7) and 7 hydroxycholesterol Co-injection Graph representation of intensity verses concentration ( moles) of co-injecting triplicate samples of the commercially available 7 -hydroxycholesterol and D7 in stepwise dilutions. Internal standard eluted at the same time as 7 -hydroxycholesterol from the HPLC-MS equipped with a C18 column. The internal standard is easily distinguishable from 7 -hydroxycholesterol because of an additional +7 molecular weight for the internal standard. Sensitivity and intensity of both compounds was similar, therefore, no additional standardization was necessary

98 90 Figure Deuterated ( 2 H 7 ) 7 -hydroxycholesterol (D7) and 7 hydroxycholesterol Co-injection

99 91 d. Evaluating Assay Detection E. Coli-expressed recombinant cholesterol 7 -hydroxylase has been previously reported to be catalytically active [98]. The following experiments were performed to determine whether the product from the enzymatic assay, 7 -hydroxycholesterol, could be detected with the new detection method. Two in-vitro reconstituted CYP7A1 enzymatic assays were performed using recombinant proteins expressed from E. coli Top 10 cells, transformed with either vector only or vector containing full-length CYP7A1 cdna. The CYP7A1 in-vitro reconstituted assay was performed as described below. e. CYP7A1 Enzyme Assay The enzyme assay was carried out in 500 μl containing mg of either HepG2 cell microsomal protein or E. coli membrane fractions, 100 μm cholesterol in 3% Triton X-100, 0.015% CHAPS, 0.1 M sodium phosphate buffer, ph 7.4, 1 mm EDTA, 5 mm DTT, 100 μm ATP, 100 μm MgCl 2, 200 M NADPH (in a regenerative system containing 5 mm isocitric acid, 0.01 U of isocitrate dehydrogenase). For CYP7A1 enzymatic assays performed with kinase treated samples, the protein samples were incubated at 30 o C for five minutes with μg of JNK, PKC, or AMPK kinase, prior to the addition of the reducing agent. In AMPK kinase assays, adenosine 5`monophosphate (AMP) is reported to increase enzymatic activity by greater than twofold. This increase was not noticed in the CYP7A1 enzymatic assay, when compared to

100 92 AMPK treated assay without AMP. Therefore, AMP was omitted from the enzymatic assay. The kinases used for the in-vitro assay were commercially available from Millipore, Temecula, CA. For the assay with the microsomal proteins, the macrolide antibiotic, oleandomycin, was added to a final concentration of 50- M to inhibit synthesis of the down stream products following 7 -hydroxycholesterol production. Oleandomycin is not widely used in the CYP7A1 enzymatic assays, but has been reported to correlated with an increase in 7 -hydroxycholesterol [104]. In my experiments, with microsomal fractions, oleandomycin decreased the production of secondary products and correlated with an increase in production of 7 hydroxycholesterol. The use of the antibiotic had no effect the production of 7 hydroxycholesterol in the recombinantly expressed protein assay because none of the enzymes necessary for catalyzing the down stream reactions are present in the assay. Therefore, it was not necessary to include the oleandomycin in the E. coli-expressed CYP7A1 enzymatic assay. Samples that were not treated with kinases were also incubated for five-minute at 30 o C prior to the addition of NADPH. For the negative control, the assay was performed with 1 mg of E. coli membrane fractions that were prepared from cells transformed with vector alone. After the addition of the reducing agent, the assay was incubated at 37 o C for 20 minutes. The reaction was stopped by the addition of anhydrous ether and was immediately ether extracted.

101 93 The reaction sample was ether extracted five times and the combined ether layers were dried in a speed vacuum and suspended in 100 μl of methanol. The sample was then centrifuged to pellet the insoluble compounds. 10 μl of the sample was used per injected into a HPLC/MS equipped with a reverse phase C18 column. A quadruplicate injection of each sample was performed with an internal standard of 0.12 nmoles of D7 7- hydroxycholesterol added to the fraction before injecting into the HPLC/MS. The HPLC was connected to a Bruker Esquire 3000 plus-ms (APCI) system. Total ion and extracted ion chromatographs of the injected samples were obtained for the production of 7 -hydroxycholesterol (385 m/z) and the internal standard D7 7 -hydroxycholesterol (392 m/z). Figure 20 shows the complete and extracted ion chromatograph traces of samples from the CYP7A1 reconstituted in-vitro enzyme assay. Fig. 20(a) represents the complete ion chromatograph traces of samples from the CYP7A1 enzymatic assay performed with E. coli-expressed recombinant protein from negative control vector only, and WT CYP7A1 respectively. Fig. 20(b) and 20(c) represent traces of the extracted ion chromatograph from both vector only and WT CYP7A1 respectively. The peak at 385m/z represents 7 -hydroxycholesterol extracted ion trace, and the 392m/z peak represents the internal standard D7 7 -hydroxycholesterol extracted ion trace. The specific enzymatic activity was determined per mg of total E. coli protein. E. coli cells harboring the vector only did not have significant measurable enzymatic activity. This was concluded by the absence of the 385m/z peak, which indicates the production of 7 -

102 94 hydroxycholesterol. The extracted ion chromatograph for the internal standard, D7 7 hydroxycholesterol (392 m/z), in the vector only assay was detectable as shown in Fig. 20b, with no detectable peak for 7 -hydroxycholesterol (385m/z). In contrast the E. coli cells that was transformed with the WT CYP7A1 cdna construct, produced a significant amount of 7 -hydroxycholesterol (385m/z) [Fig 20c]. The results from this experiment indicated that the assay was successful in producing 7 -hydroxycholesterol. It also confirmed that the new enzymatic detection method was successful in detecting the production of 7 -hydroxycholesterol from the assay. To confirm that 7 -hydroxycholesterol was produced from the enzymatic activity of the recombinant protein; the reducing agent was omitted from the assay. The 7 hydroxycholesterol peak that was measured in the NADPH absent assay was comparable to that of the assay performed with vector only. D7 was used to quantify the amount of 7 -hydroxycholesterol produced in the assay. The results were normalized to reflect the specific activity per min per mg of total E. coli protein used in the assay. Figure 21 is the graphical representation of the specific enzymatic activity of the in-vitro expressed recombinant CYP7A1 protein compared to assay performed with E. coli cells harboring the vector only. Specific enzymatic activity measured was 2.2 ± 0.3 and 174 ± 27.6 moles/min/mg of total protein for the vector only and WT CYP7A1 enzyme assay respectively. The error bars indicate the variation in measurements associated with a quadruplicate injection of the same assay and is recorded as the standard deviation of the mean.

103 95 Figure 20. Complete and Extracted Ion Traces of Samples from the CYP7A1 Enzyme Assay. Fig. 20(a)- traces represent the complete ion chromatograph representing samples from CYP7A1 enzymatic assay performed with E. coli-expressed recombinant protein expressing the negative control vector only and WT CYP7A1 respectively. Fig. 20(b) and 20(c)- traces represents the extracted ion chromatograph from assay performed with vector only and recombinant CYP7A1respectively. Arrows within the extracted ion chromatograph indicate the peaks for 7 -hydroxycholesterol (385m/z) and the internal standard D7 7 -hydroxycholesterol (392 m/z). The only peak present in the vector only chromatograph (20(b)) represents D7 7 -hydroxycholesterol. Extracted ion chromatograph from the assay performed with recombinant CYP7A1 protein (20(c)) shows the appearance of the 385m/z peak corresponding to 7 -hydroxycholesterol.

104 96 A D7 7 -hydroxycholesterol 7 -hydroxycholesterol 392 m/z B C 385 m/z Time D7 392 m/z Figure 20. Complete and Extracted Ion Traces of Samples from the CYP7A1 Enzyme Assay

105 97 Figure 21. CYP7A1 Reconstituted In-vitro Assay Performed with Vector only and WT CYP7A1 expressed protein. Enzymatic assay was normalized to reflect the amount of total protein used per assay. Solid bar represents the measured enzymatic activity for vector only. The white bar represents the measured enzymatic activity for the E. coli-expressed recombinant CYP7A1 protein. Specific activity measured were 2.2 ± 0.3 and 174 ± 27.6 moles/min/mg for the vector only and WT CYP7A1 enzyme assay respectively. The error bars represent standard deviation associated with quadruplicate injection measurements of samples.

106 98 Figure 21. CYP7A1 Reconstituted In-vitro Assay Performed with Vector Only and WT CYP7A1 E. coli-expressed Recombinant Protein.

107 99 f. Enzymatic Activity of the 30KD Fragment The purpose of this experiment was to determine if the enzymatic activity measured in the in-vitro reconstituted assay was from the 30KD fragment or the higher molecular weight proteins. To answer this question, two in-vitro reconstituted CYP7A1 enzymatic assays were performed with recombinant proteins expressed from E. coli Top 10 cells transformed with either, vector containing full-length CYP7A1 cdna, or vector containing the truncated ( 1-729) CYP7A1 construct. The CYP7A1 in-vitro reconstituted assay was performed as previously described, and the assay was normalized to reflect the amount of specific proteins expressed. The 30 KD fragment was shown to have enzymatic activity, but the measured activity was significantly lower than that measured from the WT CYP7A1. The 30KD fragment had an activity that was 30% ± 4.1 RSD of the measured enzymatic activity of WT CYP7A1 100% ± 5.7 RSD.

108 100 PART 4: MUTAGENESIS To generate the individual mutations, site directed mutagenesis was performed on the cholesterol 7 -hydroxylase sequence, using a primer-directed PCR. Invitrogen Life Technologies synthesized eighteen oligonucleotide primers that were used for the site directed mutagenesis [table 2]. Primer T193A-F contained a single nucleotide mismatch that resulted in a change from Threonine (T) Alanine (A) at position 193. This single mismatch is the same for other primers, with the numbers corresponding to the position of the amino acid change. Primer T193A/T197A-F is a double mutation at positions 193 and 197. Primers T193A- F, T197A-F and T193A/T197A-F all have a BgIII site at the 5` end of the primer which was used as a fusion point for the mutations. Primers C1-F (Not I) and C3-R (Sac I) are primers for generating the full-length CYP7A for the pivex 2.3mcs vectors with extended restriction sites at the 5` and 3` positions respectively. Primers ptrc-his2b- [NcoI]-F and ptrc-his2b[hindiii]-r are primers for generating the full-length CYP7A for the ptrc-his2b vector. The C1-F (Not I) primer was constructed to introduce a start (ATG) at the 5` amino terminus, and the C3-R (Sac I) primer was constructed to delete the stop at the 3` carboxyl terminus of the CYP7A gene. The stop codon on the CYP7A1 gene was deleted to allow fusion of the linker His-tag to the expressed polypeptide. The stop codon down stream from the Linker His-tag was used to terminate the translation of the gene product.

109 101 Table 2. Synthesized Primers used for Site-directed Mutagenesis. Table 2, shows eighteen oligonucleotide primers that were used for the site directed mutagenesis and cloning into expression vectors. Primers were synthesized by Invitrogen Life Technologies.

110 102 Table 2. Synthesized Primers used for Site-directed Mutagenesis. Oligonucleotide Primers (F=forward and R=reverse) 1 5`GGCAGAGATCTTGCAAGGCGGGACACACAG3` (T193A-F [BglII]) 2 5`GGCAGAGATCTTACAAGGCGGGACGCACAG3` (T197A- F[BglII]) 3 5`GGCAGAGATCTTGCAAGGCGGGACGCACAG3` (T193A/T197A-F[BglII]) 4 5`CAAGCGGGTGAACCACCTCTAGAG3` (T29A-F), 5 5`CTCTAGAGGTGGTTCACCCGCTTG3` (T29A-R), 6 5`GTCCATTTCATCGCAAATCCC3` (T80A-F), 7 5`GGGATTTGCGATGAAATGGAC3` (T80A-R), 8 5`GGGAGCTCGCAAATGCTTGAATTTATATTT3` (1574-R [sac1]), 9 5`TTTTTTTTTGCGGCCGCACCACATCTTTGATTTGGGGG3`(C1-66-F[Not1]), 10 5`GGGCTCGAGTTAGACTGGAGGTCTCATGATACG3` (C R[Xho1]),

111 103 Table 2. Synthesized Primers used for Site-directed Mutagenesis. Continued `TTTTTTTTTGCGGCCGCATGACCACATCTTTGATTTGGGGG3` (C1-F [not1],+ ATG), 12 5`GGGGGGAGCTCCAAATGCTTGAATTTATATTTAAATTC3` (C3- R[sac1],-ACT) 13 5`GGGACCATGGCTACCACATCTTTGATTTGG 3` (ptrc-his2b[ncoi]- F) 14 5`GGGTAGAAAGCTTCGCAAATGCTTGAATTTATA3` (ptrc- His2b[HindIII]-R) 15 5`CTCCAAAAGAGGGAAGGCATCTCAGAACTG3` (S252G-F) 16 5`CAGTTGTGAGATGCCTTCCCTCTTTTGGAG3` (S252G-R) 17 5`ATCCGAAAAGATGACATCATAGCTCTTTAC3` (S385D-F) 18 5`GTAAAGAGCTATGATGTCATCTTTTCGGAT3` (S385D-R)

112 104 Due to the positions of the nucleic acids, various strategies were used to generate the site directed mutations. Four different strategies were used to generate the sitedirected mutations. The strategies were adjusted accordingly to suit the site of the enzyme that was to be mutated. Adjustments made include: additional PCR purification steps, adjusting PCR conditions, and adjusting primer lengths a. Site-directed Mutagenesis Strategy 1 Step A This strategy was used to take of the Bgl II site that is up-stream from the desired mutation site. The full-length cdna sequence of the CYP7A1 gene was cloned into the pivex2.3mcs vector. Restriction digestion was performed on the construct with Bgl II and Nde1 restriction enzymes. This restriction digest liberated 3 fragments because the Bgl II restriction sites were present in both the CYP7A1 cdna sequence and the vector sequence up stream of the multiple cloning sites. The fragment of interest, CYP7A1 N- terminus fragments with an Nde1 5` and a Bgl II 3` restriction site, was separated from the other fragments on a 1% agarose gel. The fragment was cut out from the gel and electro-eluted out of the gel. Ethanol precipitation was performed in order to purify and concentrate the DNA fragment. The N-terminus fragment was saved for the next step in the mutation design.

113 105 Step B In step two, the C-terminus fragment with the mutation was produced. Forward primers with the single nucleotide mismatch were used for PCR with the reverse primer of the full-length CYP7A1 sequence. The forward primers were designed with the Bgl II restriction site on the N-terminus in order to fuse the fragment to the gel isolated N- terminus fragment isolated from step 1. After the PCR was performed with the mutant forward primers and the full-length reverse primers, the resulting fragment was separated on a 1% agarose gel. The DNA fragment was extracted by electro-elution and the isolated DNA fragment was concentrated by ethanol precipitation. Step C In step three, the C-terminus fragment was digested with the Bgl II restriction enzyme and ligated with the N-terminus fragment from step one to produce the fulllength cdna CYP7A1 sequence with the desired mutation. The full-length cdna sequence with the mutation was digested with SacI and ligated into the pivex2.3mcs vector that was digested with Nde1 and Sac1 restriction enzymes. Figure 22 is a schematic diagram of the procedures used in strategy 1 to produce the desired site directed mutation.

114 Figure 22. Schematic Diagram to Illustrate Site-directed Mutagenesis Strategy 1 106

115 107 b. Site-directed Mutagenesis Strategy 2 Step A This strategy was designed as an improvement of strategy 1, and was used in the double mutant design. Digestion of the CYP7A1 full-length cdna construct to produce the N-terminus fragment, in step one of strategy 1, resulted in a low concentration of isolated N-terminus fragment. The Nde1 restriction site, when digested, produced a twonucleotide overhang that also made ligation difficult. Therefore, the restriction site was changed. Strategy 2 also took advantage of the Bgl II restriction site, which is up-stream from the desired mutation sites. In step one; the N-terminus fragment was produced using the full-length CYP7A1 forward primer with the Not1 restriction site at the 5` region, with a reverse primer that is down stream from the internal Bgl II restriction site. The reverse primer was designed to incorporate the internal Bgl II restriction site in order to use it for fusing in subsequent steps. The PCR product was gel isolated from a 1% agarose gel by electro-elution and the extracted DNA was concentrated by ethanol precipitation. Step B In step two, a second PCR fragment was performed to produce the C-terminus fragment with the desired mutations. Forward primers with the single nucleotide

116 108 mismatch were used for PCR with the reverse primer of the full-length CYP7A1 sequence. The forward primers were designed with the Bgl II restriction site on the N- terminus in order to fuse the fragment to the gel-isolated N-terminus PCR fragment from step one. After the PCR was performed with the mutant forward primers and the fulllength reverse primers, the resulting fragment was separated on a 1% agarose gel. The DNA fragment was extracted by electro-elution and the isolated DNA fragment was concentrated by ethanol precipitation. Step C In step three, the two PCR fragments from step one and two were digested and then ligated together. The PCR fragment from step one was digested with Not1 and Bgl II restriction enzymes. The PCR fragment form step two was digested with Bgl II and Sac1 restriction enzymes. The two fragments were ligated together by the Bgl II site and the Not1 and Sac1 restriction sites were used to ligate the full-length CYP7A1 fragment, with the desired mutation, into the pivex2.3mcs vector. Figure 23 is a schematic diagram of the procedures used in strategy 2 to produce the desired site directed mutation.

117 Figure 23. Schematic Diagram to Illustrate Site-directed Mutagenesis Strategy 2 109

118 110 c. Site-directed Mutagenesis Strategy 3 Step A This strategy was used for designing mutations on sites that were not in close proximity to a single internal restriction site. In this site directed mutation strategy, the forward and reverse mutant primers were complimentary sequences. In step one, a PCR reaction was performed with the forward full-length CYP7A1 primer and the mutant reverse primer. This PCR reaction will generate an N-terminus fragment with the desired mutation at the C-terminus. The fragment was separated on a 1% agarose gel and gel isolated by electro-elution. The fragment was concentrated by ethanol precipitation and saved for subsequent steps. Step B In step two, a second PCR reaction was performed to produce the N-terminus fragment. The PCR was performed with the mutant forward primer and the full-length CYP7A1 reverse primer. The resulting fragment from this PCR reaction had the desired mutation at the N-terminus of the fragment. The fragment was purified as previously described and saved for subsequent steps.

119 111 Step C In step three, the PCR fragments that were generated in steps one and two above, were used as mega-primers for another PCR reaction. Because of the complimentary sequence present in both fragments, two annealing positions are possible. The first is the annealing of the 3` coding strand of the N-terminus fragment to the 3` template strand of the C-terminus fragment. This annealing results in an available 3` hydroxyl ends. The second possible annealing position is the 5` template strand of the N-terminus fragment annealing with the 5` coding strand of the C-terminus fragment. This annealing position results in open 5` phosphate ends. Of the two possible annealing positions, only the first position described will be able to elongate. Step D The resulting full-length CYP7A1 cdna sequence from step three is further amplified by a PCR reaction with the full-length forward and reverse primers. The amplified fragment is separated on a 1% agarose gel and isolated by electro-elution. The fragment was digested with Not1 and Sac1 restriction enzymes and ligated into the pivex2.3mcs vector. Figure 24 is a schematic diagram of the procedures used in strategy 3 to produce the desired site directed mutation.

120 112 Figure 24. Schematic Diagram to Illustrate the Site-directed Mutagenesis Strategy 3

121 113 d. Site-directed Mutagenesis Strategy 4 Step A This strategy was used for designing the truncated CYP7A1 polypeptide. In this truncated mutation strategy no primers were necessary. In step one, the full-length CYP7A1 ptrc-his2b vector construct was digested with BamH I and Stu I restriction enzymes. The BamH I site was located at the multiple cloning site of the vector construct. The Stu I restriction site is in an internal restriction site in the CYP7A1 cdna sequence, and is located at nucleotide 720. The product from this restriction digest were two fragments, the n-terminal CYP7A1 cdna sequence, approximately 729 nucleic acids and the ptrc vector construct with the c-terminal CYP7A1 cdna sequence. The linearized vector construct was separated on a 1% agarose gel and gel isolated by electroelution. The fragment was concentrated by ethanol precipitation and saved for subsequent steps. Step B In step two, the 5` overhang produced from the BamH I restriction digest was filled-in, to produce a blunt end, by using a bacteriophage T4 DNA polymerase with a high concentration of datp, dttp, dgtp, dctp, in a low Mg2+ ionic buffer. The high concentration of dntps facilitates polymerization of the single-strand overhang. An alternative approach will be to exploit the exonuclease activity of the T4 DNA

122 114 polymerase. Bacteriophage T4 DNA polymerase has a very strong exonuclease activity. By omitting the dntps from the reaction, the T4 polymerase will cleave off the singlestranded DNA overhang produced from the restriction enzyme digestion. The polymerase activity of T4 was chosen in this experiment in order to cause a frame-shift in the CYP7A1 cdna sequence. Step C The two blunt ends from the Stu I restriction digest and the 5` overhang from the BamH I restriction digest that was filled in with the bacteriophage T4 DNA polymerase, were ligated together. Figure 25 is a schematic diagram of the procedures used in strategy 4 to produce the desired truncation mutation.

123 Figure 25. Schematic Diagram to Illustrate the Mutagenesis Strategy 4 used to Generate Truncated CYP7A1 Polypeptide 115

124 116 e. Mutant Sub-cloning and Characterization Each mutant-vector construct was transformed into the E. coli DH5 cell line, plated on LB + amp agar plate, and incubated overnight at 37 o c. Single colonies were selected from the plates and grown in 2ml LB + amp overnight. 1 ml of the cell culture was used to perform a plasmid mini prep analysis to confirm fragment ligation into plasmids [fig. 26]. In the figure, lane 1 is the DNA molecular weight marker, lane 2 is the E. coli cell line transformed with vector only, Lanes 3-17 are mutant-vector constructs transformed into E. coli DH5 cell line. The arrow on the left shows the migration of the empty vector used as a negative control. Arrows on the right, from top to bottom, show the migration of the positive CYP7A1 DNA fragment-vector construct and the vector without fragment insert respectively. The colonies that showed a slower migration when separated on a agarose gel were streak plated to get single colonies.

125 117 Figure 26. Mini Plasmid prep of Colonies from Mutants Transformed into E.coli DH5 Cell Lines. Lane1: DNA molecular weight markers, lane 2: control plasmid (E. coli cells transformed with vector only without insert), lanes 3-17: colonies from mutant transformation, shift in plasmid migration indicates plasmid contains insert. Arrow on the left, labeled A shows migration of empty vector. Arrows on the right labeled B and C, from top to bottom, show the migration of positive insert-vector construct and plasmids that do not have an insert respectively.

126 Figure 26. Mini Plasmid prep of Colonies from Mutants Transformed into E.coli DH5 Cell Lines. 118

127 119 f. Mutant Vector-construct Restriction Mapping Colonies were selected from the streak plates and grown in 2ml LB + amp overnight. 1ml of the cell culture were used for mini plasmid prep, and isolated plasmid DNA was used for restriction mapping. Restriction mapping of the mutant plasmid was performed using Hind III and Not I restriction enzymes and separated by electrophoresis on a 1% agarose gel [fig. 27]. The Not I restriction site was created during the mutant design and was therefore not present in the starting WT CYP7A1 cdna fragment. The Not 1 site was used to distinguish newly generated cdna fragments from starting templates. In the figure, lanes 2,4,6, and 8 were plasmid DNA digested with only Hind III restriction enzyme. The two fragments that were produced from the digestion migrated at 2695 and 2426 bp, which is a sum of the insert with the His-tag (1551bp), and the pivex 2.3 MCS vector (3553bp). Lanes 3,5,7 and 9 were plasmid DNA digested with Hind III and Not I restriction enzymes. The liberated fragments were 2695, 2129 and 297 bp. The digestion of the Not 1 restriction enzyme confirmed that the cdna fragments were from the mutant design and not the starting template cdna. Freezer stocks of the positive plasmid preps were made and plasmid prep was performed using Nucleobond DNA purification kit from Clontech (Palo Alto, CA) for DNA sequencing. Cleveland Genomics sequenced the purified plasmid, and a contiguous alignment of the fragments was used to ensure that no additional mutation was made in the sequence. Four primers were used for the sequencing, two forward primers and two reverse primers. The sequencing results from the four primers were aligned to form overlapping sequences using map vector and Clustal W (1.83)

128 120 sequencing alignment programs. Figure 28 illustrates the positions of the sequence alignments used to confirm positive mutation.

129 121 Figure 27. Restriction Mapping of Mutant Plasmid Lane1: DNA lambda HindIII and EcoR I molecular weight marker, lanes 2,4,6 and 8, are plasmids digested with only HindIII Lanes 3,5, 7 and 9 are plasmids digested with HindIII and Not1. (Digestion with only HindIII liberates 2695 & 2426 bp, Hind III and Not1 liberates 2695,2129 &297 bp)

130 Kbp 5Kbp 3Kbp 2Kbp 2695bp 2129bp 564bp 297bp Figure 27. Restriction Mapping of Mutant Plasmid

131 123 T193/197A S252G S385D TMD Cholesterol 7 -Hydroxylase 504aa s long -60 T C1-66-F C2-R C3-R 1551 Figure 28. Contiguous Alignment of Mutant Nucleotide Sequence Four primers were used to sequence the mutant CYP7A1 cdna sequence. Two end primers and two internal primers were used to create overlapping sequences in order to confirm sequence identity.

132 CHAPTER III RESULTS AND CONCLUSIONS PART 1: AICAR, an AMPK Kinase Activator, does not Affect CYP7A1 mrna Steady-state Levels. AICAR, an AMPK kinase activator, has been shown to repress bile acid production in HepG2 cell culture. To determine if the bile acid repression observed by the activation of AMPK, was a result of transcriptional control, the mrna levels of CYP7A1 in treated HepG2 cells was measured using real-time PCR. Figure 29 shows a time course of the effects of AICAR and CDCA, on the steady-state mrna levels of CYP7A1 in HepG2 cells. CDCA was used as a positive control because of its well-established repressive effects on CYP7A1 transcription. HepG2 cells were treated with 20μM AICAR and 25μM CDCA for 0h, 0.5h to 24h as indicated. Relative mrna levels of CYP7A1 were calculated with respect to UBC mrna. The mrna expression at 0h was set as 1. With treatment of HepG2 cells 124

133 125 with CDCA, the repression on the CYP7A1 transcript happens quickly and is maximal at the 2h point. The repression of CYP7A1 transcription by CDCA is through a feedback repression and involves the activation of FTF, therefore, the mrna levels of FTF in cells treated with CDCA was also measured. The repression of the mrna steady-state levels of CYP7A1, when treated with CDCA, correlates with the increase in FTF mrna expression in the same cells. The activation of FTF transcription by CDCA is as previously reported and supports the repression of CYP7A1, at the transcriptional level, by bile acid mediated FTF activation[9, 18]. Activation of AMPK did not change the transcription of CYP7A1 within the time course of the experiment. This result in combination with the total bile acid production results from AICAR treated HepG2 cell, supports the hypothesis that CYP7A1 enzymatic activity can be regulated at a level other than that of transcription.

134 126 Figure 29. AICAR, AMPK Kinase Activator does not Affect CYP7A1 mrna Steady-state Levels Real-time PCR data for hepg2 cells treated with AICAR and CDCA. HepG2 cells were treated with 20μM AICAR and 25μM CDCA for 0h, 0.5h, up to 24h as indicated. The total RNA was prepared and measured with real-time PCR. Relative mrna levels of CYP7A1 were calculated with respect to UBC mrna. The mrna expression at 0h was set as 1. A rapid repression of CYP7A1 is observed at the 2h time point and this correlates with an increase in FTF mrna levels. AICAR treatment did not have an effect on CYP7A1 mrna levels throughout the time course of the experiment.

135 127 Figure 29. AICAR, AMPK Kinase Activator does not affect CYP7A1 mrna Steady-state Levels

136 128 PART 2: CYP7A1 In-vitro Reconstituted Assay with E. coli-expressed and Microsomal HepG2 Cells Treated with AMPK, PKC, and JNK Kinases To determine if kinase activity had an effect on CYP7A1 enzymatic activity, microsomal proteins from HepG2 cells and E. coli-expressed recombinant CYP7A1, were treated with various commercially available kinases prior to performing the in-vitro reconstituted CYP7A1 enzymatic assay. The effects of the kinases on enzymatic activity were measured with the new detection method described in the material and methods section. AMPK, PKC and JNK kinases were used in the in-vitro enzymatic assay because of their already well-established effects on cholesterol and bile acid synthesis, and feedback regulation of bile acid synthesis. a. Rationale for Kinase Selection i. AMPK The effect of AMPK on cholesterol synthesis is well characterized[72-75]. Phosphorylated AMPK regulates cholesterol synthesis by phosphorylating and inactivating 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase. The ability for AMPK to regulate cholesterol synthesis, and also phosphorylate cholesterol 7 -hydroxylase, the rate-limiting enzyme in bile acid synthesis, suggests a possible mechanism by which bile acid synthesis and cholesterol synthesis could be co-regulated through phosphorylation events. AMPK is also a well-established energy sensor in the

137 129 cell. The effects of AMPK on cholesterol 7 -hydroxylase enzymatic activity will also provide insight on the cross talk between bile acid synthesis and energy metabolism. ii. PKC Bile acids have been shown to regulate gene expression of CYP7A1 both by binding farnesoid X receptor (FXR)[14, 22, 24, 25] and by activating cellular Kinases [23, 26, 27]. Both bile acids and phorbol esters activate protein kinase C (PKC). Treatment of HepG2 cells with bile acids resulted in the translocation of PKC, and PKC inhibitors reduced the bile acid repression of transcription from the CYP7A promoter [105]. The regulation of cholesterol 7 -hydroxylase by PKC kinase activity will provide an additional mechanism by which bile acids regulate CYP7A1 enzymatic activity at the enzyme level. iii. JNK The activation of FXR by bile acids has also been shown to indirectly repress CYP7A1 transcriptional activity by activating small heterodimer proteins (SHP-1) [26]. It has been proposed that SHP-1 can repress transcription of CYP7A1 through the activation of the JNK/c-jun pathway. Primary rat hepatocyte cultures treated with taurocholate (TCA) was shown to strongly activate JNK in a time- and concentrationdependant manner[26, 28]. This activation of JNK correlated with SHP-1 activation and CYP7A1 transcriptional repression. In a more recent study using human primary

138 130 hepatocytes, activation of JNK by chenodeoxycholic acid (CDCA) and interleukin-1 (IL-1 ) represses CYP7A1 transcriptional activity by interacting with HNF4 [106]. It was proposed in this study that the activation of JNK indirectly repressed CYP7A1 gene transcription by blocking HNF4 recruitment of PPAR coactivator-1 (PGC-1 ) to the CYP7A1 chromatin. This study will address the possible direct effect that JNK kinase has on CYP7A1 enzymatic activity. b. Kinase Treatment of Microsomal HepG2 Cells The purpose of this experiment is to determine if kinase activity has a direct effect on CYP7A1 enzymatic activity. HepG2 cells were used to prepare microsomal fractions, as described in the material and methods section, for the CYP7A1 in-vitro reconstituted assay. The cells were grown to confluency in dulbecco s modified eagle s medium (DMEM): F12 with no phenol red, 10% fetal calf serum, 100units/ml penicillin, in 100mm tissue culture plates incubated at 3% CO 2 at 37 o c. After cells reached confluency, the overlay medium was changed to medium without fetal calf serum. The cells were monitored and the medium was changed twice within four hours. The removal of the medium is to remove any bile acids accumulated over the cells. The presence of bile acids in the medium will repress the transcriptional activity of the CYP7A1 gene. The cells were collected after the four-hour incubation and microsomal fractions were prepared. The CYP7A1 in-vitro enzymatic assay was performed as described in the material and methods section. Microsomal fractions were treated with AMPK, PKC, or JNK

139 131 kinases, and compared to untreated assay. Percent activity was determined by combining results from 3-4 individual enzyme assays. Comparisons were only made between assays performed with the same protein preps. Specific enzyme activity was determined for pmoles of 7 -hydroxycholesterol produced per min per milligram of total microsomal protein, by using the D7 internal standards. Because of the day-to-day variation in assay activity, the statistical significance of the treatments were determined by using a single classification ANOVA analysis of the percentage of specific enzymatic activity measured in each assay. In the CYP7A1 in-vitro reconstituted assay performed with microsomal HepG 2 cells [Fig. 30], kinase treatment significantly repressed CYP7A enzymatic activity with a p-value < AMPK, JNK, and PKC all repressed CYP7A1 enzymatic activity by 51% ± 5, 57% ± 11, and 48% ± 6 respectively when compared to WT CYP7A1 enzymatic activity. Error bars represent standard error of the mean between experimental samples. This result provided clear evidence that endogenous CYP7A1 enzymatic activity can be regulated by phosphorylation events at the post-translational level. This data is supported by the real-time PCR data of HepG2 cells treated with kinase activators. The repression of bile acid production in the AICAR treated HepG2 cells was not a result of limited cholesterol, but a result of phosphorylation events at the protein level.

140 132 Figure 30. AMPK, PKC and JNK Kinase Activity Repressed CYP7A1 Enzymatic Activity in Microsomal HepG2 Cell Fractions In-vitro CYP7A1 reconstituted enzymatic assay performed with microsomal HepG2 cells pretreated with AMPK, PKC, and JNK kinases for 5 min prior to the addition of NADPH. Percent activity was determined for 3-4 individual assays that were combined for statistical analysis. AMPK, JNK, and PKC all significantly repressed CYP7A enzymatic activity with a p-value of < ** = p-value <0.01, and *** = p-value < Error bars represent standard error of the mean.

141 133 *** *** *** Figure 30. AMPK, PKC and JNK Kinase Activity Repressed CYP7A1 Enzymatic Activity in Microsomal HepG2 Cell Fractions

142 134 c. Kinase Treatment of E. coli-expressed CYP7A1 Recombinant Protein To confirm the regulation of CYP7A1 enzymatic activity by phosphorylation events, the in-vitro reconstituted assay was performed with CYP7A1 E. coli-expressed recombinant protein. Full-length CYP7A1 cdna sequence was cloned into the ptrc- His2b expression vector. The sample preparation of the recombinant protein and CYP7A assay were performed as previously described with a few changes. In the in-vitro assay performed with E. coli-expressed recombinant proteins, oleandomycin was omitted from the assay. The oleandomycin was omitted because it served no function in the recombinant CYP7A1 enzyme assay, since the enzymes required for proceeding through the classical bile acid pathway are absent in the reaction mixture. Also the addition of oleandomycin did not have an effect on measured enzymatic activity. In contrast to the in-vitro assay with HepG2 cell microsomal fractions, in the invitro enzymatic assay using E. coli-expressed CYP7A1 recombinant protein, AMPK kinase activity was the only kinase capable of repressing the enzymatic activity of CYP7A1 [Fig. 31]. AMPK significantly repressed CYP7A enzymatic activity by 63% ± 6 s.e.m with a p-value <0.01. PKC showed a tendency of increasing enzymatic activity in the E. coli in-vitro assay, with a 23% ± 19 s.e.m. The effects measured for both JNK and PKC were not significant, both having a p-value >0.05. This result, in combination with the in-vitro assay performed with the HepG2 cell microsomal fraction, and the realtime PCR results from treated HepG2 cells, confirmed that CYP7A1 enzymatic activity is directly regulated by phosphorylation at the post-translational level.

143 135 Figure 31. AMPK Kinase Activity Represses CYP7A1 Enzymatic Activity in E. coliexpressed Recombinant Protein. In-vitro CYP7A1 reconstituted enzymatic assay was performed with E. coli-expressed CYP7A1 recombinant protein that was pretreated with AMPK, PKC, and JNK kinases for 5 min prior to the addition of NADPH to the reaction mixture. Percent activity was determined for 3-4 individual assays that were combined for statistical analysis. AMPK was the only kinase that significantly repressed CYP7A enzymatic activity with a p-value of <0.01. PKC and PKA did not have a significant effect on CYP7A1 enzymatic activity. Both PKC and PKA had a p-value >0.05. ** = p-value <0.01, and *** = p-value < Error bars represent standard error of the mean.

144 136 n/s n/s ** Figure 31. AMPK Kinase Activity Represses CYP7A1 Enzymatic Activity in E. coliexpressed Recombinant Protein.

145 137 PART 3: AMPK Kinase Treated In-vitro CYP7A1 Enzymatic Assay with Mutant CYP7A1 Recombinant Protein a. Rationale for Mutation Design To expand on the discovery that AMPK represses CYP7A1 enzymatic activity, both in the microsomal HepG2 cell and the E. coli-expressed recombinant protein, the following experiments were designed to determine the specific sites on the CYP7A1 polypeptide that was responsible for the AMPK repressive effect. Various AMPK phosphorylation sites were mutated on the enzyme and an in-vitro reconstitution assay was performed to determine the effects of the mutations. The sites that were mutated were chosen from a CYP7A1 phosphorylation map previously published from our lab (Stroup and Ramsaran; 2005). b. Rationale for Mutation-site Selection Stroup et al[1], performed an in-vitro kinase assays where 45 synthetic 15-mer peptides, based on the human cholesterol 7 -hydroxylase amino acid sequence, were challenged with 12 protein kinases (AMPK, PKA, JNK, PKC, PKC I, PKC II, PKC, PKC, PKC, PKC, PKC, and PKC ). In the kinase assay, three of the 15-mer polypeptides were phosphorylated by almost all the kinase tested. Polypeptide F188/I202, which has 2 threonines at position 193 and 197, and its mutant F188/I202B6 were phosphorylated by 9 out of 12 kinases (JNK, PKC, and PKC being the

146 138 exceptions). C69/L83, which also has a threonine at position 80, was phosphorylated by 11 out of 12 kinases (all except PKA). Other peptides showed more specificity: L22/N36 and A106/N120 associated with PKA; L247/M261 and N380/M394 with PKA and AMPK; L428/T442 with JNK; and D342/L356 and I349/R364 recognized by PKC, PKC, and PKC. This result provided the starting point for choosing AMPK phosphorylation sites. AMPK was shown to strongly phosphorylate threonines at positions 80, 193 and 197, and serines at positions 252 and 385 [Fig. 32].

147 139 Figure 32. Tabulated Results from Synthetic Peptides versus Commercially Available Protein Kinases. Data from autoradiography of SDS-PAGE gels of 45 synthetic peptides reacted with 12 protein kinases. Shading of boxes corresponds to the relative amount of radioactive phosphorous incorporated into each peptide. From the far left column, sequence of peptides, results from individual kinases: 95% gray, most intense labeling for given kinases; 50% gray, less than the strongest signal, but significantly above background; and lightest gray, peptide detectable, but close to background (marked to donate specificity of labeling) D. Stroup, J.R. Ramsaran, Cholesterol 7 -hydroxylase is phosphorylated at multiple amino acids, BBRC 329 (2005)

148 140 D. Stroup, J.R. Ramsaran, Cholesterol 7 -hydroxylase is phosphorylated at multiple amino acids, BBRC 329 (2005) Figure 32. Tabulated Results from Synthetic Peptides Treated with Commercially Available Protein Kinases

149 141 c. Mutations of AMPK Phosphorylation Sites The threonine (T) at position 80 was mutated to alanine (A). Threonines at positions 193 and 197 were mutated as a double mutant, because both sites are in close proximity of each other and were shown in the kinase assay to be non-specifically phosphorylated by kinase activity. Only when both sites were removed, was phosphate transfer not observed in the assay. Serine (S) at position 252 was mutated to glycine (G) and the 30 KD fragment, described in the materials and methods section, was used for S385. The 30KD fragment was shown to have catalytic activity and S385 was the only AMPK phosphorylation site present on the polypeptide. Four different strategies were used to design the mutations and are described in detail in the material and methods section. The mutated CYP7A1 recombinant protein was used in an in-vitro reconstituted assay to determine which site(s) is responsible for the AMPK repressive effect that was observed in the CYP7A1 reconstituted enzymatic assay. d. CYP7A1 In-vitro Assay with T193A/T197A Double Mutant Site directed mutagenesis strategy two, described in the material and methods section, was used to create the T193A/T197A double mutant. The double mutant design was confirmed by restriction mapping of the vector construct followed by cdna sequencing using 5` and 3` full-length primers, and forward and reverse internal primers. The multiple sequence results were aligned to provide an overlapping sequence, confirming sequence identity and positive mutation sites. The CYP7A1 T193A/T197A double mutant was cloned into the ptrc-his2b expression vector and transformed into the

150 142 E. coli Top10 cell line as described in the material and methods section. The CYP7A1 T193A/T197A double mutant was expressed and E. coli protein prep was performed for the CYP7A1 in-vitro reconstituted enzyme assay. The CYP7A1 in-vitro assay with E. coli-expressed T193A/T197A double mutant recombinant protein showed an approximately two-fold less enzymatic activity when compared to WT CYP7A1. Assay was normalized to reflect total amount of specific protein used in each assay. The enzymatic activity of the T193A/T197A CYP7A1 mutant was 58% ± 6 RSD of the enzymatic activity measured for that of WT, 100% ± 16.2 RSD. Although enzymatic activity of the double mutant was lower than WT CYP7A1, the mutant was still able to convert cholesterol to 7 -hydroxycholesterol in the enzymatic assay, and AMPK kinase activity repressed CYP7A1 enzymatic activity [Fig. 33]. AMPK repressed the double mutants enzymatic activity by 43% ±1.7 RSD, when compared to untreated double mutant recombinant protein. This result suggested that threonines at positions 193 and 197 were not responsible for the repressive effect of AMPK kinase activity on CYP7A1 enzymatic activity.

151 143 Figure 33. AMPK Kinase Activity Represses T193A/T197A Double Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein. In-vitro CYP7A1 reconstituted enzymatic assay was performed with E. coli-expressed T193A/T197A double mutant CYP7A1 recombinant protein. Enzymatic activity of the double mutant was 58% ± 6 RSD of the enzymatic activity measured in WT CYP7A1, 100% ± 16.2 RSD. Double mutant was pretreated with AMPK kinase for 5 min prior to the addition of NADPH to the reaction mixture. AMPK kinase activity repressed total enzymatic activity of the double mutant by 43% ± 1.7 RSD. Mutations at T193 and T197 did not abolish the repressive effect observed by AMPK kinase activity. Error bars represent relative standard deviation between quadruplicate injections of the samples measured.

152 144 *** Figure 33. AMPK Kinase Activity Represses T193A/T197A Double Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein.

153 145 e. CYP7A1 In-vitro Assay with T80A Mutant Threonine at amino acid position 80 was another site that showed low specificity to kinase activity. The site was strongly phosphorylated by seven of the twelve kinases used in the in-vitro assay. Site directed mutagenesis strategy three described in the materials and methods section was used to generate the desired T80A mutation. Restriction mapping and cdna sequencing, as previously described, was used to confirm the mutation. The CYP7A1 in-vitro assay with E. coli-expressed T80A mutant recombinant protein, had a total enzymatic activity that was 34% ± 11 RSD of the enzymatic activity measured for WT CYP7A1, 100 ± 9 RSD. The measured enzymatic activity was normalized to reflect equal amount of specific protein used per assay. Upon AMPK kinase treatment, the enzyme activity of the T80A mutant protein was repressed by 24% ± 5.5 RSD when compared to untreated T80A mutant [Fig. 34]. Noticeably, the repression observed with the T80A mutant was two times less than the repressive effect of AMPK on WT CYP7A1 enzymatic activity. This result suggested that AMPK phosphorylation at T80 might be required for complete suppression of WT CYP7A1 enzymatic activity, but is not sufficient in preventing the repressive effect of AMPK kinase activity on CYP7A1 enzymatic activity.

154 146 Figure 34. AMPK Kinase Activity Represses T80A Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein. In-vitro CYP7A1 reconstituted enzymatic assay was performed with E. coli-expressed T80A mutant CYP7A1 recombinant protein. Enzymatic activity of the mutant was 34% ± 11 RSD of the enzymatic activity measured in WT CYP7A1, 100% ± 9 RSD. The T80A mutant was pretreated with AMPK kinase for 5 min prior to the addition of NADPH to the reaction mixture. AMPK kinase activity repressed total enzymatic activity of the mutant by 24% ± 5.5 RSD. Mutation at T80A did not abolish the repressive effect observed by AMPK kinase activity. Error bars represent relative standard deviation between quadruplicate injections of the samples measured.

155 147 * Figure 34. AMPK Kinase Activity Represses T80A Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein.

156 148 f. CYP7A1 In-vitro Assay with S252G Mutant The other sites that were tested showed more specificity to kinase activity in the in-vitro kinase assay. S252 and S385 were both heavily phosphorylated by AMPK kinase activity. The two sites are located towards the central and n-terminal region of the CYP7A1 polypeptide sequence respectively. The S252G CYP7A1 mutant was designed with the site directed mutagenesis strategy three. S252 mutant was characterized in the same manner as previously described for the other mutations. The CYP7A1 in-vitro assay, with E. coli-expressed S252G mutant recombinant protein, showed comparable enzymatic activity compared to WT CYP7A1 recombinant protein. Unlike previous results that showed repression of CYP7A1 enzymatic activity upon AMPK kinase treatment, the S252G CYP7A1 mutant enzymatic activity was not repressed by AMPK kinase activity [Fig. 35]. The measured enzymatic activity was the same for treated and untreated samples with percent activity of 100% ± 6.3RSD and 100% ± 1.4RSD respectively. This result provided evidence that serine at position 252 was responsible for the AMPK repressive effect on CYP7A1 enzymatic activity.

157 149 Figure 35. AMPK Kinase Activity did not Repress S252G Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein. In-vitro CYP7A1 reconstituted enzymatic assay was performed with E. coli-expressed S252G mutant CYP7A1 recombinant protein. Enzymatic activity of the S252G mutant was comparable to activity measured with WT CYP7A1. S252G mutant was pretreated with AMPK kinase for 5 min prior to the addition of NADPH. AMPK kinase activity did not have a significant effect on the total enzymatic activity of the S252G mutant when compared to untreated mutant. Mutation at S252 abolished the repressive effect observed by AMPK kinase activity. Error bars represent relative standard deviation between quadruplicate injections of the samples measured.

158 150 Figure 35. AMPK Kinase Activity did not Repress S252G Mutant CYP7A1 Enzymatic Activity in E. coli-expressed Recombinant Protein.

159 151 g. CYP7A1 In-vitro Assay with Truncated WT CYP7A1 ( 1-729) Mutant The 30KD fragment was previously shown to have enzymatic activity, therefore the effects of AMPK on the fragments activity was determined in the CYP7A1 in-vitro assay. The fragment consists of amino acids methionine (M) 308 to leucine (L) 504 and covers the catalytic region of the CYP7A1 polypeptide that includes: the steroid-binding site, the aromatic amino acid region and the heme region. The fragment consists of only one of the sites that are phosphorylated by AMPK, S385. By using the fragment in this experiment, the effects of AMPK on S385 can be determined. This experiment will also be used to confirm whether S252 is needed for AMPK repression of CYP7A1 enzymatic activity, since the site is missing from the 30 KD fragment. In the CYP7A1 in-vitro reconstituted assay with E. coli-expressed 30KD fragment recombinant protein, AMPK kinase activity did not repress enzyme activity, rather treatment of the 30KD fragment with AMPK kinase increased enzymatic activity by 46% ± 6.8 RSD, when compared to untreated 30KD fragment [Fig. 36]. This experiment provided evidence that S385 phosphorylation was not responsible for the repressive effect on CYP7A1 enzymatic activity by AMPK. It also provided supporting evidence for the conclusions made about AMPK kinase activity regulating CYP7A1 through the phosphorylation of S252. The increase in enzymatic activity suggests that phosphorylation could also have a stimulatory effect on CYP7A1 enzymatic activity.

160 152 Figure 36. AMPK Kinase Activity Stimulates WT CYP7A1 ( 1-729) Enzymatic Activity In-vitro CYP7A1 reconstituted enzymatic assay was performed with E. coliexpressed WT CYP7A1 ( 1-729) truncated recombinant protein. Enzymatic activity measured for the truncated fragment was 30% ± 4.1 RSD of the enzymatic activity measured in WT CYP7A1, 100% ± 5.7 RSD. The 30KD fragment has only one known AMPK phosphorylation site at position S385. The fragment was pretreated with AMPK kinase for 5 min prior to the addition of NADPH to the reaction mixture. AMPK kinase activity stimulated enzymatic activity of the mutant by 46% ± 6.8 RSD. Error bars represent relative standard deviation between quadruplicate injections of the samples measured.

161 153 *** Figure 36. AMPK Kinase Activity Stimulates WT CYP7A1 ( 1-729) Enzymatic Activity

162 CHAPTER IV DISCUSSION Cholesterol and bile acid synthesis are major metabolic pathways, and disruptions in these pathways are associated with metabolic disorders and progression of atherosclerosis. Bile acid synthesis is the major cholesterol disposal pathway, and influences cholesterol homeostasis in the cell. Understanding the cross talk between cholesterol and bile acid synthesis will help in the treatment of metabolic disorders and prevention of atherosclerosis. The goal of this study is to determine if cholesterol 7 -hydroxylase enzymatic activity is regulated by phosphorylation events at the post-translational level, and whether AMPK kinase activity represses bile acid production by direct phosphorylating CYP7A1. To answer this question, the experiments were divided into two specific aims. Specific aim 1 was designed to address the question of whether CYP7A1 enzymatic activity changed upon phosphorylation events. Specific aim 2 was designed to determine the specific sites, on the CYP7A1 polypeptide, that are responsible for the effects noticed by kinase activity. The 154

163 155 difficulty in assaying the enzyme activity of cholesterol 7 -hydroxylase has been well documented [98, 99]. This study presents a new technique using HPLC-MS to quantify cholesterol 7 -hydroxylase enzymatic activity, by directly measuring 7 hydroxycholesterol. The new detection method proved to be very sensitive, and was capable of detecting 7 -hydroxycholesterol in the moles range. Published studies on transcriptional regulation of CYP7A1 have shown mrna levels to correlate with repression by bile acids, phorbol esters, and insulin, and activated by camp. The transcriptional studies often did not include measurements of enzyme activity, and when reported, the mrna levels did not always correlate with bile acid production. Results from our lab demonstrated bile acid repression in HepG2 cells treated with various kinase activators [1]. HepG2 cells treated with camp, a PKA activator; AICAR, an AMPK activator; and PMA, a PKC activator, showed a 19%, 28%, and 35% repression in bile acid production respectively, when compared to untreated HepG2 cells. PKC and PKA have been previously shown to affect the transcriptional level of the CYP7A1 gene. Unlike PKC and PKA, this was the first report of bile acid repression by an AMPK kinase pathway. There have been suggestions that cholesterol synthesis and cholesterol disposal, via bile acid synthesis, are coordinated in the cell. If cholesterol and bile acid synthesis are co-regulated, there are two possible scenarios by which this would be possible: first, repression of cholesterol biosynthesis with a stimulation in bile acid synthesis, to facilitate the removal of excess cholesterol in the cell; second, repression/stimulation of

164 156 cholesterol biosynthesis associated with similar repression/stimulation of bile acid synthesis, in order to maintain cholesterol homoeostasis in the cell. The discovery that bile acid synthesis can be repressed by AMPK kinase activity will provide a possible mechanism by which the two pathways are co-regulated in the cell. Specific Aim 1: CYP7A1 Enzymatic Activity is Regulated by Phosphorylation Events To determine whether the repression of bile acid production in HepG2 cell treated with AMPK kinase activator was a result of transcriptional control, the mrna level of CYP7A1 was measured by real-time PCR. The real-time PCR results showed that the repression of bile acid production was not a result of transcriptional control. AICAR treatment repressed bile acid production in a 90 min time course, but the mrna level of CYP7A1 did not change in the treated cells [fig. 29]. This provided evidence that the repressive effect of AMPK kinase activity on bile acid synthesis, was occurring at a level other than that of transcription. A possible mechanism for the AMPK repressive effect on bile acid synthesis could be explained by the AMPK-dependant repression of HMG-CoA reductase. Phosphorylation of HMG-CoA reductase, by AMPK, shuts down cholesterol synthesis. Lowering the amount of cholesterol present would result in decreased bile acid production. The in-vitro reconstituted CYP7A1 enzymatic assay experiments were performed to address the question of whether AMPK was phosphorylating CYP7A1 directly or was the effects of bile acid repression a result of a reduction in cholesterol

165 157 levels via phosphorylation of HMG-CoA. In the in-vitro reconstituted CYP7A1 enzymatic assay, cholesterol will not be rate limiting in the reaction because excess cholesterol will be added to the assay. To investigate the possibility of post-translational regulation by phosphorylation events, CYP7A1 in-vitro assays were compared between microsomal HepG2 cells and E. coli-expressed CYP7A1 recombinant protein. In the in-vitro assay using microsomal HepG2 cells [Fig. 29], AMPK, JNK, and PKC kinases all significantly repressed CYP7A1 enzymatic activity, with a p-value <0.001 when compared to untreated control. This was the first direct evidence that CYP7A1 enzymatic activity was regulated by phosphorylation events at the enzyme level. The in-vitro CYP7A1 enzymatic assay with E. coli-expressed recombinant protein was performed in the same manner as described for the in-vitro assay with microsomal fractions, with the exception of omitting oleandomycin from the reaction. E. Coliexpressed CYP7A1 recombinant protein was treated with AMPK, JNK, and PKC kinases, and the enzymatic activity was compared to untreated assay samples. In contrast to the results from the in-vitro enzyme assay performed with microsomal HepG2 cells, AMPK was the only kinase capable of repressing enzymatic activity [Fig. 31]. AMPK repressed CYP7A1 enzymatic activity by 63% ± 6 S.E.M with a p-value <0.01. Although PKC showed a tendency of increasing enzymatic activity by 23% ± 19 S.E.M, both PKC and JNK did not have a significant effect on CYP7A1 enzymatic activity.

166 158 The difference in enzymatic activity noticed upon kinase treatment, between the microsomal fractions and recombinant protein assays, suggests that multiple kinase activities could be involved in regulating CYP7A1 enzymatic activity. Endogenous CYP7A1 has been shown to exist as a phosphoprotein in the cell [1]. Although other post-translational modification has not been identified for the enzyme, this cannot be discounted. The effects of the kinase activity on endogenous CYP7A1 enzymatic activity could be a result of a combination of native post-translational modifications with kinase activity, and not just phosphorylation events. E. coli-expressed CYP7A1 recombinant protein has been previously shown to be catalytically active (Chiang et al. 1991). The recombinant protein is hypophosphorylated, therefore, the effects of kinase activity on the enzymatic activity represents phosphorylation events alone. The in-vitro enzyme assay experiments performed with recombinant CYP7A1 confirm that CYP7A1 enzymatic activity could be regulated by phosphorylation events alone. The in-vitro assays also suggest that the regulation of the enzymatic activity of CYP7A1 by kinase activity could result in either a repressive or stimulatory effect. Phosphorylation by JNK kinase repressed CYP7A1 enzyme activity in microsomal fractions but had no effect on E. coli-expressed recombinant protein. This Suggests that other forms of post-translational modifications might be required to observe the repressive effect by JNK kinase activity. Phosphorylation by PKC kinase activity also repressed enzymatic activity in HepG2 microsomal fractions, but slightly increased enzymatic activity in E. coli-expressed recombinant protein assays. Even though the measured increase in activity was not significant; the slight increase suggests that some

167 159 kinase activity might have a stimulatory effect on CYP7A1 enzymatic activity. This avenue of investigation was not followed up in this study; rather, the definite repressive effect of AMPK on enzymatic activity was further pursued. AMPK was the only kinase tested capable of significantly repressing CYP7A1 enzymatic activity in the in-vitro assay with both recombinant protein and microsomal fractions. AMP is reported to increase enzymatic activity of AMPK by greater than two fold. This increase was not noticed in the CYP7A1 enzymatic assay, when compared to AMPK treated assay without AMP. Therefore, AMP was omitted from the enzymatic assay. AMPK activity has been reported to be stimulated in muscle cells by anti-diabetic agents through an AMP-independent pathway[107]. The enzymatic activity of the commercially available AMPK was reported to be 636 U/mg, where 1 unit of AMPK activity is defined as 1 nmole phosphate incorporated into 200 μm substrate per minute at 30 o C with a final ATP concentration of 100 μm. The amount of AMPK kinase used in the in-vitro assay was in excess, therefore, the absence of AMP in the in-vitro assay does not influence the final observations. The next experiments were designed to determine the specific sites responsible for the repressive effects of AMPK on CYP7A1 enzymatic activity.

168 160 Specific Aim 2: Determine Sites Responsible for AMPK Repressive Effects The in-vitro assay with CYP7A1 recombinant protein was used to determine the site(s) necessary for AMPK repression of CYP7A1 enzymatic activity. The advantage of using the recombinant protein is that the specific sites on the human cholesterol 7 hydroxylase polypeptide, which are phosphorylated by AMPK kinase, have been mapped in an in-vitro kinase assays[1]. The recombinant protein is also hypophosphorylated, therefore, effects of AMPK alone on the enzymatic activity can be determined for the specific site(s). T193, T197, T80, S252, and S385 were determined in the kinase assays to be strong AMPK phosphorylation sites. T193 and T197 were mutated as a double mutant to alanines, T80 was mutated to an alanine, and S252 was mutated to glycine. For S385, the truncated 30KD polypeptide was used for the in-vitro assay. S385 was the only AMPK phosphorylation site present on the polypeptide. The mutated recombinant proteins were used in an AMPK kinase treated assay, to determine which site(s) is involved in the repressive effects observed by AMPK kinase activity. T193/T197, and T80 were non-specific to kinase activity in the in-vitro kinase assay previously reported. AMPK kinase activity repressed the double mutant and T80A mutant enzymatic activity by 43% ±1.7 and 24% ± 5.5 RSD respectively, when compared to untreated assay. The reduction in enzymatic activity noticed with untreated mutants compared to WT, and the non-specificity of the sites to kinase activity, suggests that T193, T197 and T80 are more likely involved in enzyme activation. The lower activity

169 161 measured in the double mutant and T80A mutant suggests that these sites might be needed for complete enzyme activity. CYP7A1 exists as a phosphoprotein in the cell; therefore, it is reasonable to assume the sites that are non-specific to kinase phosphorylation will most likely be phosphorylated in the cell. If T193, T197 and T80 were phosphorylation repressive sites, then the enzymatic activity of CYP7A1 would be inactive at a high regularity in the cell. This is not the case because bile acid production is measured in HepG2 cell at short time courses. A simpler explanation would be that the enzymatic activity of CYP7A1 is regulated by the phosphorylation of specific sites, which have a stronger effect on the enzyme activity than the effects by phosphorylations that already exist on the enzyme. These non-specific sites might serve to enhance effects of other specific phosphorylation sites. The other prominent AMPK phosphorylation sites, S252 and S385, showed more specificity in the in-vitro kinase assays. S252 was mutated to glycine and the recombinant protein was used in the CYP7A1 enzymatic assay. The specific enzymatic activity measured from the S252 mutant was comparable to that of WT. Upon AMPK kinase treatment, there was no significant difference between treated and untreated assays. The measured enzymatic activity for treated and untreated samples were 100% ± 6.3RSD and 100% ± 1.4RSD respectively. This result suggested that S252 is the phosphorylation site needed for AMPK repression of CYP7A1 enzymatic activity.

170 162 The 30KD fragment, which was previously characterized in this study, was used to test S385 as a possible site for the AMPK repressive effect on CYP7A1 enzymatic activity. The fragment served two purposes in this experiment. First, the fragment only has one of the AMPK phosphorylation sites, S385. The effects of AMPK on its enzymatic activity will address the importance of the site for AMPK repression. Secondly, since S252 is missing from the 30KD fragment, the effects that AMPK kinase activity has on the fragment will help support the conclusions made about S252. AMPK kinase activity on the 30KD fragment, interestingly resulted in an increase in specific enzymatic activity. Phosphorylation by AMPK resulted in a 46% increase in enzymatic activity, when compared to the untreated assay. This result confirmed S385 was not responsible for the AMPK repression of CYP7A1 enzymatic activity. It also provided additional evidence to support the hypothesis that S252 is the site responsible for the repression of CYP7A1 enzymatic activity by AMPK kinase. The result also suggests that other sites might be required for maintaining CYP7A1 enzymatic activity. It was only when the N-terminus was removed did enzymatic activity increase upon phosphorylation. The N-terminus could be functioning as a regulatory region on the CYP7A1 polypeptide. The fragment does not have S252, which was shown in this study to be responsible for the repressive effect of AMPK on CYP7A1 enzyme activity. The 30KD fragment has all the catalytic regions of the CYP7A1 polypeptide. Losing the regulatory region of the enzyme could result in an increase in total enzymatic activity upon phosphorylation events.

171 163 Mapping of the S252 AMPK Phosphorylation Sites to the CYP7A1 Crystal Structure In an attempt to understand the mechanism by which AMPK represses CYP7A1 enzymatic activity through the phosphorylation of S252, the AMPK phosphorylation site was mapped to the CYP7A1 crystal structure. Seven residues on the CYP7A1 polypeptide have been mapped as the steroid binding pocket, and shown to interact with the cholesterol molecule[108]. More recently, the crystal structure of CYP7A1 was determined, and is available on the Protein Data Bank (PDB), EC no: Figure 37 shows the crystal structure of CYP7A1, which was modified to illustrate the locations of AMPK phosphorylation sites and important amino acids involved in cholesterol binding. AMPK phosphorylation sites S80 (cyan), T193 and T197 (magenta), S252 (yellow), S385 (red), the steroid binding site (black), and the amino acids that interact with cholesterol, cholesterol binding-pocket (green). The cholesterol binding-pocket is located in the center of the crystal structure, and consists of amino acid residues L280, L283, W284, A288, S358, S360, G485, and I488 located on two different alpha-helix strands. The cholesterol molecule was determined to bind in the pocket with its 7 -hydroxyl group oriented towards the heme group[108]. The steroid binding-pocket also has a helix-loop-helix structure sitting on top of it. S252 is located on the outer part of the crystal structure, a position suitable for phosphorylation events. The S252 site is located on a loop that is located between the helix-loop-helix structure, (blocking the cholesterol binding-pocket) and one of the alpha helix strands harboring the

172 164 amino acids involved in cholesterol interaction with CYP7A1. S252 is also located adjacent to two hydrophobic amino acids isoleucine 187 and phenylalanine 188. The locations of the S252 on the crystal structure, suggests a possible mechanism by which AMPK kinase activity regulates CYP7A1 enzymatic activity. The phosphorylation of S252 could result in a structural change by which the helix-loop-helix structure, blocking the cholesterol binding-pocket, is moved further into the grove. The movement of the helix-loop-helix structure, upon phosphorylation, might be a result of the proximity of the phosphorylated serine 252 and the hydrophobic amino acids adjacently located. The blocking of the cholesterol binding-pocket will influence the accessibility of the binding-pocket to its ligand.

173 165 Figure 37. Crystal Structure of CYP7A1 The crystal structure of CYP7A1 was obtained from the Protein Data Bank (PDB), EC no: The structure was modified to illustrate the AMPK phosphorylation sites and important amino acids involved in cholesterol binding. S80 (cyan), T193 and T197 (magenta), S252 (yellow), S385 (red), the steroid binding site (black), and the amino acids that interact with cholesterol, cholesterol binding-pocket (green). Attention is drawn to the helix-loop-helix structure (blue) that is attached to S252 and blocking the cholesterol binding-pocket. Alternative views of the structure are provided.

174 S25 T193 Cholesterol Binding 166 T197 Steroid Binding Site T80 S38 Figure 37. Crystal Structure of CYP7A1

175 167 It has long been hypothesized that there exists a mechanism by which cholesterol synthesis and cholesterol disposal is coordinated in the cell. The evidence provided here, that AMPK kinase activity represses the enzymatic activity of CYP7A1 by phosphorylation, is the first step to answering that question. The regulation of cholesterol synthesis by AMPK phosphorylation of HMG-CoA has been well documented. Here AMPK is shown to also regulate cholesterol 7 -hydroxylase, the rate-limiting enzyme in bile acid production through the classical pathway. This study provides evidence of how cholesterol homoeostasis is maintained in the cell by kinase activity. AMPK phosphorylation of both HMG-CoA and cholesterol 7 -hydroxylase, the two rate limiting enzymes in cholesterol synthesis and bile acid synthesis respectively, result in a repression of their enzymatic activities, thereby maintaining cholesterol homeostasis in the cell [fig.38]. The discovery that CYP7A1 enzymatic activity can be modified by phosphorylation events, presents a rapid method of regulation bile acid biosynthesis. This study also provides evidence of how cellular energy homeostasis is co-regulated with cholesterol homeostasis and bile acid synthesis. AMPK kinase is a serine/threonine protein kinase that is highly conserved in higher eukaryotes, yeast and plants[109, 110] and serves as an energy gauge for the cell. During energy starvation conditions such as anoxia or exercise-induced muscle contraction, ATP concentrations are decreased resulting in an increase in cellular AMP levels. The fluctuations in the AMP:ATP ratios result in the activation of AMPK. Activated AMPK leads to increase in cellular energy

176 168 by inhibiting anabolic energy consuming pathways such as fatty acid, cholesterol, and amino acid synthesis, and stimulates catabolic energy producing pathways such as fatty acid oxidation, glycolysis and glucose transport. By regulating both anabolic and catabolic pathways, AMPK is able to maintain the energy homeostasis in the cell when in low energy states. The ability for AMPK kinase activity to repress CYP7A1 enzymatic activity is consistent with energy preservation during low cellular energy states. Cholesterol and bile acid synthesis are both energy-consuming pathways. HMG-CoA reductase and CYP7A1 are the rate-limiting enzymes in cholesterol synthesis and bile acid production respectively. In low cellular energy states, the activation of AMPK will represses both cholesterol and bile acid synthesis to preserve energy, and the repression of both pathways together maintains cholesterol homeostasis in the cell. In summary, the present study provides evidence to support the hypothesis that CYP7A1 enzymatic activity is regulated at the post-translational level by phosphorylation events. AMPK kinase activity was shown to repress CYP7A1 enzymatic activity by directly phosphorylating the enzyme at S252. This study provides a mechanism by which cholesterol synthesis and bile acid biosynthesis is co-regulated in the cell. It also provides information about the cross talk between cholesterol synthesis, bile acid production, and energy homeostasis.

177 169 Cholesterol Synthesis Cholesterol Disposal P P AMPK Figure 38. AMPK Maintains Cholesterol Homeostasis Figure 38 is a schematic depiction to illustrate a mechanism by which cholesterol homeostasis is maintained in the cell. The phosphorylation of HMG-CoA reductase and cholesterol 7 -hydroxylase, both the rate-limiting enzymes in cholesterol synthesis and cholesterol disposal pathways respectively, represses both pathways and thereby maintains the cellular cholesterol levels.

Cholesterol and its transport. Alice Skoumalová

Cholesterol and its transport. Alice Skoumalová Cholesterol and its transport Alice Skoumalová 27 carbons Cholesterol - structure Cholesterol importance A stabilizing component of cell membranes A precursor of bile salts A precursor of steroid hormones

More information

Companion to Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism Lecture Notes

Companion to Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism Lecture Notes Companion to Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism Lecture Notes The major site of acetoacetate and 3-hydorxybutyrate production is in the liver. 3-hydorxybutyrate is the

More information

Cholesterol metabolism. Function Biosynthesis Transport in the organism Hypercholesterolemia

Cholesterol metabolism. Function Biosynthesis Transport in the organism Hypercholesterolemia Cholesterol metabolism Function Biosynthesis Transport in the organism Hypercholesterolemia - component of all cell membranes - precursor of bile acids steroid hormones vitamin D Cholesterol Sources: dietary

More information

Unit IV Problem 3 Biochemistry: Cholesterol Metabolism and Lipoproteins

Unit IV Problem 3 Biochemistry: Cholesterol Metabolism and Lipoproteins Unit IV Problem 3 Biochemistry: Cholesterol Metabolism and Lipoproteins - Cholesterol: It is a sterol which is found in all eukaryotic cells and contains an oxygen (as a hydroxyl group OH) on Carbon number

More information

cholesterol structure Cholesterol FAQs Cholesterol promotes the liquid-ordered phase of membranes Friday, October 15, 2010

cholesterol structure Cholesterol FAQs Cholesterol promotes the liquid-ordered phase of membranes Friday, October 15, 2010 cholesterol structure most plasma cholesterol is in the esterified form (not found in cells or membranes) cholesterol functions in all membranes (drives formation of lipid microdomains) cholesterol is

More information

Bile acid metabolism. doc. Ing. Zenóbia Chavková, CSc.

Bile acid metabolism. doc. Ing. Zenóbia Chavková, CSc. Bile acid metabolism doc. Ing. Zenóbia Chavková, CSc. Bile acid metabolism Importance: Availability for fat & cholesterol absorption Regulates total body pool of cholesterol Factors that synthesis promote

More information

Cellular control of cholesterol. Peter Takizawa Department of Cell Biology

Cellular control of cholesterol. Peter Takizawa Department of Cell Biology Cellular control of cholesterol Peter Takizawa Department of Cell Biology Brief overview of cholesterol s biological role Regulation of cholesterol synthesis Dietary and cellular uptake of cholesterol

More information

Oxidation of Long Chain Fatty Acids

Oxidation of Long Chain Fatty Acids Oxidation of Long Chain Fatty Acids Dr NC Bird Oxidation of long chain fatty acids is the primary source of energy supply in man and animals. Hibernating animals utilise fat stores to maintain body heat,

More information

Chapter 26 Biochemistry 5th edition. phospholipids. Sphingolipids. Cholesterol. db=books&itool=toolbar

Chapter 26 Biochemistry 5th edition. phospholipids. Sphingolipids. Cholesterol.   db=books&itool=toolbar http://www.ncbi.nlm.nih.gov/sites/entrez? db=books&itool=toolbar 1 The surface of a soap bubble is a bilayer formed by detergent molecules 2 Chapter 26 Biochemistry 5th edition phospholipids Sphingolipids

More information

Cholesterol Metabolism

Cholesterol Metabolism Cholesterol Metabolism Lippincott s Illustrated Review Chapter 18 Steroid Nucleus 1 2 Cholesterol was isolated from gall bladder stones in 1774 3 Sources and Elimination of Cholesterol Synthesis: 1000

More information

Acetyl CoA HMG CoA Mevalonate (C6) Dimethylallyl Pyrophosphate isopentenyl Pyrophosphate (C5) Geranyl Pyrophosphate (C10) FarnesylPyrophosphate (C15) Squalene (C30) Lanosterol (C30) 7 Dehydrocholesterol

More information

Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism- 2015

Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism- 2015 Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism- 2015 The major site of acetoacetate and 3-hydorxybutyrate production is the liver. They are preferred substrates for myocardiocytes

More information

MCB130 Midterm. GSI s Name:

MCB130 Midterm. GSI s Name: 1. Peroxisomes are small, membrane-enclosed organelles that function in the degradation of fatty acids and in the degradation of H 2 O 2. Peroxisomes are not part of the secretory pathway and peroxisomal

More information

Bile acids and the brain. Suggested pathogenetic mechanism in connection with formation of brain xanthomas in patients with CTX

Bile acids and the brain. Suggested pathogenetic mechanism in connection with formation of brain xanthomas in patients with CTX Bile acids and the brain. Suggested pathogenetic mechanism in connection with formation of brain xanthomas in patients with CTX Ingemar Björkhem, Marjan Shafaati, Ulla Andersson, Ute Panzenboeck, Magnus

More information

Glossary For TheFatNurse s For All Ages Series Adipocytes, also known as lipocytes and fat cells, are the cells that primarily compose adipose tissue, specialized in storing energy as fat. Apolipoprotein

More information

number Done by Corrected by Doctor

number Done by Corrected by Doctor number 29 Done by Ali Yaghi Corrected by Shahd Alqudah Doctor Faisal Al-Khatibe In this lecture we will continue the steps of synthesizing cholesterol. in the previous sheet we reached the step of forming

More information

23.1 Lipid Metabolism in Animals. Chapter 23. Micelles Lipid Metabolism in. Animals. Overview of Digestion Lipid Metabolism in

23.1 Lipid Metabolism in Animals. Chapter 23. Micelles Lipid Metabolism in. Animals. Overview of Digestion Lipid Metabolism in Denniston Topping Caret Copyright! The McGraw-Hill Companies, Inc. Permission required for reproduction or display. Chapter 23 Fatty Acid Metabolism Triglycerides (Tgl) are emulsified into fat droplets

More information

Chemical Classification of Hormones

Chemical Classification of Hormones Steroid Hormones Chemical Classification of Hormones Hormones are chemical messengers that transport signals from one cell to another There are 4 major chemical classes of hormones steroid hormones - i.e.

More information

Pathophysiology of Lipid Disorders

Pathophysiology of Lipid Disorders Pathophysiology of Lipid Disorders Henry Ginsberg, M.D. Division of Preventive Medicine and Nutrition CHD in the United States CHD is the single largest killer of men and women 12 million have history

More information

Niacin Metabolism: Effects on Cholesterol

Niacin Metabolism: Effects on Cholesterol Niacin Metabolism: Effects on Cholesterol By Julianne R. Edwards For Dr. William R. Proulx, PhD, RD Associate Professor of Nutrition and Dietetics In partial fulfillments for the requirements of NUTR342

More information

BIOL2171 ANU TCA CYCLE

BIOL2171 ANU TCA CYCLE TCA CYCLE IMPORTANCE: Oxidation of 2C Acetyl Co-A 2CO 2 + 3NADH + FADH 2 (8e-s donated to O 2 in the ETC) + GTP (energy) + Heat OVERVIEW: Occurs In the mitochondrion matrix. 1. the acetyl portion of acetyl-coa

More information

Lipid metabolism in familial hypercholesterolemia

Lipid metabolism in familial hypercholesterolemia Lipid metabolism in familial hypercholesterolemia Khalid Al-Rasadi, BSc, MD, FRCPC Head of Biochemistry Department, SQU Head of Lipid and LDL-Apheresis Unit, SQUH President of Oman society of Lipid & Atherosclerosis

More information

Regulating Hepatic Cellular Cholesterol

Regulating Hepatic Cellular Cholesterol Under circumstances of cholesterol deficiency, Sterol Regulatory Element Binding Proteins (SREBPs) via binding to DNA nuclear response elements set off genomic production of proteins and enzymes that induce

More information

Lecture 3 6/28/10. Membrane Lipids. Importance of Membranes. Categories of Lipids. Lipids: Chapter 20 Sections 4-7. ! Membranes are important in

Lecture 3 6/28/10. Membrane Lipids. Importance of Membranes. Categories of Lipids. Lipids: Chapter 20 Sections 4-7. ! Membranes are important in Lecture 3 Lipids: Chapter 20 Sections 4-7! The most polar lipids are found in the membranes of cells and organelles! Why?! These lipids are amphipathic! Membranes are complex and have many components Membrane

More information

2.5. AMPK activity

2.5. AMPK activity Supplement Fig. A 3 B phos-ampk 2.5 * Control AICAR AMPK AMPK activity (Absorbance at 45 nm) 2.5.5 Control AICAR Supplement Fig. Effects of AICAR on AMPK activation in macrophages. J774. macrophages were

More information

LIPID METABOLISM. Sri Widia A Jusman Department of Biochemistry & Molecular Biology FMUI

LIPID METABOLISM. Sri Widia A Jusman Department of Biochemistry & Molecular Biology FMUI LIPID METABOLISM Sri Widia A Jusman Department of Biochemistry & Molecular Biology FMUI Lipid metabolism is concerned mainly with fatty acids cholesterol Source of fatty acids from dietary fat de novo

More information

number Done by Corrected by Doctor Faisal Al-Khatibe

number Done by Corrected by Doctor Faisal Al-Khatibe number 24 Done by Mohammed tarabieh Corrected by Doctor Faisal Al-Khatibe 1 P a g e *Please look over the previous sheet about fatty acid synthesis **Oxidation(degradation) of fatty acids, occurs in the

More information

Chapter VIII: Dr. Sameh Sarray Hlaoui

Chapter VIII: Dr. Sameh Sarray Hlaoui Chapter VIII: Dr. Sameh Sarray Hlaoui Lipoproteins a Lipids are insoluble in plasma. In order to be transported they are combined with specific proteins to form lipoproteins: Clusters of proteins and lipids.

More information

ANTIHYPERLIPIDEMIA. Darmawan,dr.,M.Kes,Sp.PD

ANTIHYPERLIPIDEMIA. Darmawan,dr.,M.Kes,Sp.PD ANTIHYPERLIPIDEMIA Darmawan,dr.,M.Kes,Sp.PD Plasma lipids consist mostly of lipoproteins Spherical complexes of lipids and specific proteins (apolipoproteins). The clinically important lipoproteins, listed

More information

Chapter 10. Regulatory Strategy

Chapter 10. Regulatory Strategy Chapter 10 Regulatory Strategy Regulation of enzymatic activity: 1. Allosteric Control. Allosteric proteins have a regulatory site(s) and multiple functional sites Activity of proteins is regulated by

More information

Plasma lipoproteins & atherosclerosis by. Prof.Dr. Maha M. Sallam

Plasma lipoproteins & atherosclerosis by. Prof.Dr. Maha M. Sallam Biochemistry Department Plasma lipoproteins & atherosclerosis by Prof.Dr. Maha M. Sallam 1 1. Recognize structures,types and role of lipoproteins in blood (Chylomicrons, VLDL, LDL and HDL). 2. Explain

More information

Propagation of the Signal

Propagation of the Signal OpenStax-CNX module: m44452 1 Propagation of the Signal OpenStax College This work is produced by OpenStax-CNX and licensed under the Creative Commons Attribution License 3.0 By the end of this section,

More information

Integration Of Metabolism

Integration Of Metabolism Integration Of Metabolism Metabolism Consist of Highly Interconnected Pathways The basic strategy of catabolic metabolism is to form ATP, NADPH, and building blocks for biosyntheses. 1. ATP is the universal

More information

Is it really that simple? Alyssa Hasty, PhD Associate Professor Molecular Physiology and Biophysics

Is it really that simple? Alyssa Hasty, PhD Associate Professor Molecular Physiology and Biophysics Alyssa Hasty, PhD Associate Professor Molecular Physiology and Biophysics Why we care about hepatic lipogenesis Control of lipid synthesis What can go wrong in humans Animal models dlto study lipoprotein

More information

Lipid Metabolism. Catabolism Overview

Lipid Metabolism. Catabolism Overview Lipid Metabolism Pratt & Cornely, Chapter 17 Catabolism Overview Lipids as a fuel source from diet Beta oxidation Mechanism ATP production Ketone bodies as fuel 1 High energy More reduced Little water

More information

Lipoproteins Metabolism Reference: Campbell Biochemistry and Lippincott s Biochemistry

Lipoproteins Metabolism Reference: Campbell Biochemistry and Lippincott s Biochemistry Lipoproteins Metabolism Reference: Campbell Biochemistry and Lippincott s Biochemistry Learning Objectives 1. Define lipoproteins and explain the rationale of their formation in blood. 2. List different

More information

Lipid Metabolism. Remember fats?? Triacylglycerols - major form of energy storage in animals

Lipid Metabolism. Remember fats?? Triacylglycerols - major form of energy storage in animals Remember fats?? Triacylglycerols - major form of energy storage in animals Your energy reserves: ~0.5% carbs (glycogen + glucose) ~15% protein (muscle, last resort) ~85% fat Why use fat for energy? 1 gram

More information

Summary and concluding remarks

Summary and concluding remarks Summary and concluding remarks This thesis is focused on the role and interaction of different cholesterol and phospholipid transporters. Cholesterol homeostasis is accomplished via a tightly regulated

More information

BCM 221 LECTURES OJEMEKELE O.

BCM 221 LECTURES OJEMEKELE O. BCM 221 LECTURES BY OJEMEKELE O. OUTLINE INTRODUCTION TO LIPID CHEMISTRY STORAGE OF ENERGY IN ADIPOCYTES MOBILIZATION OF ENERGY STORES IN ADIPOCYTES KETONE BODIES AND KETOSIS PYRUVATE DEHYDROGENASE COMPLEX

More information

Biochemistry: A Short Course

Biochemistry: A Short Course Tymoczko Berg Stryer Biochemistry: A Short Course Second Edition CHAPTER 28 Fatty Acid Synthesis 2013 W. H. Freeman and Company Chapter 28 Outline 1. The first stage of fatty acid synthesis is transfer

More information

Topic 11. Coronary Artery Disease

Topic 11. Coronary Artery Disease Topic 11 Coronary Artery Disease Lipid metabolism http://news.bbc.co.uk/2/hi/health/7372495.stm Sterol Metabolism and Coronary Artery Disease Big Picture: Exogenous Cholesterol and Fat Metabolism Fats-Triglycerides

More information

ab SREBP-2 Translocation Assay Kit (Cell-Based)

ab SREBP-2 Translocation Assay Kit (Cell-Based) ab133114 SREBP-2 Translocation Assay Kit (Cell-Based) Instructions for Use For analysis of translocation of SREBP-2 into nuclei. This product is for research use only and is not intended for diagnostic

More information

Cholest s er e o r l o ١

Cholest s er e o r l o ١ Cholesterol ١ Contents of The Lecture What is Cholesterol? Structure of Cholesterol Structure of Cholesteryl Ester Normal Cholestrol Level Sources of Cholesterol What Are The Exogenous Sources Of Cholesterol?

More information

Published on Second Faculty of Medicine, Charles University (http://www.lf2.cuni.cz )

Published on Second Faculty of Medicine, Charles University (http://www.lf2.cuni.cz ) Published on Second Faculty of Medicine, Charles University (http://www.lf2.cuni.cz ) Biochemistry Submitted by Marie Havlová on 8. February 2012-0:00 Syllabus of Biochemistry Mechanisms of enzyme catalysis.

More information

1- Which of the following statements is TRUE in regards to eukaryotic and prokaryotic cells?

1- Which of the following statements is TRUE in regards to eukaryotic and prokaryotic cells? Name: NetID: Exam 3 - Version 1 October 23, 2017 Dr. A. Pimentel Each question has a value of 4 points and there are a total of 160 points in the exam. However, the maximum score of this exam will be capped

More information

Signal Transduction Cascades

Signal Transduction Cascades Signal Transduction Cascades Contents of this page: Kinases & phosphatases Protein Kinase A (camp-dependent protein kinase) G-protein signal cascade Structure of G-proteins Small GTP-binding proteins,

More information

LIPID METABOLISM

LIPID METABOLISM LIPID METABOLISM LIPOGENESIS LIPOGENESIS LIPOGENESIS FATTY ACID SYNTHESIS DE NOVO FFA in the blood come from :- (a) Dietary fat (b) Dietary carbohydrate/protein in excess of need FA TAG Site of synthesis:-

More information

CHM333 LECTURE 34: 11/30 12/2/09 FALL 2009 Professor Christine Hrycyna

CHM333 LECTURE 34: 11/30 12/2/09 FALL 2009 Professor Christine Hrycyna Lipid Metabolism β-oxidation FA Acetyl-CoA Triacylglycerols (TAGs) and glycogen are the two major forms of stored energy in vertebrates Glycogen can supply ATP for muscle contraction for less than an hour

More information

Chemistry 107 Exam 4 Study Guide

Chemistry 107 Exam 4 Study Guide Chemistry 107 Exam 4 Study Guide Chapter 10 10.1 Recognize that enzyme catalyze reactions by lowering activation energies. Know the definition of a catalyst. Differentiate between absolute, relative and

More information

Lipid Metabolism in Familial Hypercholesterolemia

Lipid Metabolism in Familial Hypercholesterolemia Lipid Metabolism in Familial Hypercholesterolemia Khalid Al-Rasadi, BSc, MD, FRCPC Head of Biochemistry Department, SQU Head of Lipid and LDL-Apheresis Unit, SQUH President of Oman society of Lipid & Atherosclerosis

More information

Receptor mediated Signal Transduction

Receptor mediated Signal Transduction Receptor mediated Signal Transduction G-protein-linked receptors adenylyl cyclase camp PKA Organization of receptor protein-tyrosine kinases From G.M. Cooper, The Cell. A molecular approach, 2004, third

More information

In glycolysis, glucose is converted to pyruvate. If the pyruvate is reduced to lactate, the pathway does not require O 2 and is called anaerobic

In glycolysis, glucose is converted to pyruvate. If the pyruvate is reduced to lactate, the pathway does not require O 2 and is called anaerobic Glycolysis 1 In glycolysis, glucose is converted to pyruvate. If the pyruvate is reduced to lactate, the pathway does not require O 2 and is called anaerobic glycolysis. If this pyruvate is converted instead

More information

Cholesterol metabolism Ι

Cholesterol metabolism Ι Sheet # 22 Cholesterol metabolism Ι Today is the first lecture in the Cholesterol metabolism and you can refer to chapter 18 in Lippincott illustrated review Q: Why Cholesterol was written in 3 different

More information

The endoplasmic reticulum is a network of folded membranes that form channels through the cytoplasm and sacs called cisternae.

The endoplasmic reticulum is a network of folded membranes that form channels through the cytoplasm and sacs called cisternae. Endoplasmic reticulum (ER) The endoplasmic reticulum is a network of folded membranes that form channels through the cytoplasm and sacs called cisternae. Cisternae serve as channels for the transport of

More information

7.06 Cell Biology EXAM #3 April 24, 2003

7.06 Cell Biology EXAM #3 April 24, 2003 7.06 Spring 2003 Exam 3 Name 1 of 8 7.06 Cell Biology EXAM #3 April 24, 2003 This is an open book exam, and you are allowed access to books and notes. Please write your answers to the questions in the

More information

Nafith Abu Tarboush DDS, MSc, PhD

Nafith Abu Tarboush DDS, MSc, PhD Nafith Abu Tarboush DDS, MSc, PhD natarboush@ju.edu.jo www.facebook.com/natarboush Lipids (cholesterol, cholesterol esters, phospholipids & triacylglycerols) combined with proteins (apolipoprotein) in

More information

Chapter 10. Introduction to Nutrition and Metabolism, 3 rd edition David A Bender Taylor & Francis Ltd, London 2002

Chapter 10. Introduction to Nutrition and Metabolism, 3 rd edition David A Bender Taylor & Francis Ltd, London 2002 Chapter 10 Introduction to Nutrition and Metabolism, 3 rd edition David A Bender Taylor & Francis Ltd, London 2002 Chapter 10: Integration and Control of Metabolism Press the space bar or click the mouse

More information

/ The following functional group is a. Aldehyde c. Carboxyl b. Ketone d. Amino

/ The following functional group is a. Aldehyde c. Carboxyl b. Ketone d. Amino Section A: Multiple Choice Select the answer that best answers the following questions. Please write your selected choice on the line provided, in addition to circling the answer. /25 1. The following

More information

UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY

UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 1 UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY GLUCOSE HOMEOSTASIS An Overview WHAT IS HOMEOSTASIS? Homeostasis

More information

Name: Date: Block: Biology 12

Name: Date: Block: Biology 12 Name: Date: Block: Biology 12 Provincial Exam Review: Cell Processes and Applications January 2003 Use the following diagram to answer questions 1 and 2. 1. Which labelled organelle produces most of the

More information

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM. Triacylglycerol and Fatty Acid Metabolism

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM. Triacylglycerol and Fatty Acid Metabolism ANSC/NUTR 618 LIPIDS & LIPID METABOLISM II. Triacylglycerol synthesis A. Overall pathway Glycerol-3-phosphate + 3 Fatty acyl-coa à Triacylglycerol + 3 CoASH B. Enzymes 1. Acyl-CoA synthase 2. Glycerol-phosphate

More information

Cell Quality Control. Peter Takizawa Department of Cell Biology

Cell Quality Control. Peter Takizawa Department of Cell Biology Cell Quality Control Peter Takizawa Department of Cell Biology Cellular quality control reduces production of defective proteins. Cells have many quality control systems to ensure that cell does not build

More information

REGULATION OF ENZYME ACTIVITY. Medical Biochemistry, Lecture 25

REGULATION OF ENZYME ACTIVITY. Medical Biochemistry, Lecture 25 REGULATION OF ENZYME ACTIVITY Medical Biochemistry, Lecture 25 Lecture 25, Outline General properties of enzyme regulation Regulation of enzyme concentrations Allosteric enzymes and feedback inhibition

More information

Arteriosclerosis & Atherosclerosis

Arteriosclerosis & Atherosclerosis Arteriosclerosis & Atherosclerosis Arteriosclerosis = hardening of arteries = arterial wall thickening + loss of elasticity 3 types: -Arteriolosclerosis -Monckeberg medial sclerosis -Atherosclerosis Arteriosclerosis,

More information

Regulation of P450. b. competitive-inhibitor but not substrate. c. non-competitive inhibition. 2. Covalent binding to heme or protein

Regulation of P450. b. competitive-inhibitor but not substrate. c. non-competitive inhibition. 2. Covalent binding to heme or protein 10. Inhibition of P450 Regulation of P450 a. competitive-2 chemicals metabolized by same isoform b. competitive-inhibitor but not substrate c. non-competitive inhibition 11. Induction of P450 1. Non-covalent

More information

PLASMA LIPOPROTEINS AND LIPIDS DETERMINATION OF PLASMA CHOLESTEROL AND TRIGLICERIDE LEVEL

PLASMA LIPOPROTEINS AND LIPIDS DETERMINATION OF PLASMA CHOLESTEROL AND TRIGLICERIDE LEVEL PLASMA LIPOPROTEINS AND LIPIDS DETERMINATION OF PLASMA CHOLESTEROL AND TRIGLICERIDE LEVEL Lipids are characterized by low polarity and limited solubility in water. Their plasma concentration is about 500-600

More information

Membrane Lipids & Cholesterol Metabolism

Membrane Lipids & Cholesterol Metabolism Membrane Lipids & Cholesterol Metabolism Learning Objectives 1. How Are Acylglycerols and Compound Lipids Produced? 2. The synthesis of Sphingolipids from Ceramide 3. Diseases due to Disruption of Lipid

More information

ENERGY FROM INGESTED NUTREINTS MAY BE USED IMMEDIATELY OR STORED

ENERGY FROM INGESTED NUTREINTS MAY BE USED IMMEDIATELY OR STORED QUIZ/TEST REVIEW NOTES SECTION 1 SHORT TERM METABOLISM [METABOLISM] Learning Objectives: Identify primary energy stores of the body Differentiate the metabolic processes of the fed and fasted states Explain

More information

Roles of Lipids. principal form of stored energy major constituents of cell membranes vitamins messengers intra and extracellular

Roles of Lipids. principal form of stored energy major constituents of cell membranes vitamins messengers intra and extracellular Roles of Lipids principal form of stored energy major constituents of cell membranes vitamins messengers intra and extracellular = Oxidation of fatty acids Central energy-yielding pathway in animals. O

More information

Practice Exam 2 MCBII

Practice Exam 2 MCBII 1. Which feature is true for signal sequences and for stop transfer transmembrane domains (4 pts)? A. They are both 20 hydrophobic amino acids long. B. They are both found at the N-terminus of the protein.

More information

FAD FADH2. glycerol-3- phosphate. dehydrogenase. This DHAP is metabolically no different from that produced in glycolysis.

FAD FADH2. glycerol-3- phosphate. dehydrogenase. This DHAP is metabolically no different from that produced in glycolysis. 1 Lipid Metabolism: ow that we are aware of the types of lipids in our bodies, it is important to see how we make them or break them. We will start our discussion with triacylglyceride degradation, and

More information

Metabolic integration and Regulation

Metabolic integration and Regulation Metabolic integration and Regulation 109700: Graduate Biochemistry Trimester 2/2016 Assistant Prof. Dr. Panida Khunkaewla kpanida@sut.ac.th School of Chemistry Suranaree University of Technology 1 Overview

More information

General Principles of Endocrine Physiology

General Principles of Endocrine Physiology General Principles of Endocrine Physiology By Dr. Isabel S.S. Hwang Department of Physiology Faculty of Medicine University of Hong Kong The major human endocrine glands Endocrine glands and hormones

More information

By: Dr Hadi Mozafari 1

By: Dr Hadi Mozafari 1 Biological lipids are a chemically diverse group of compounds, the common and defining feature of which is their insolubility in water. By: Dr Hadi Mozafari 1 Fats and oils are the principal stored forms

More information

Enzyme-coupled Receptors. Cell-surface receptors 1. Ion-channel-coupled receptors 2. G-protein-coupled receptors 3. Enzyme-coupled receptors

Enzyme-coupled Receptors. Cell-surface receptors 1. Ion-channel-coupled receptors 2. G-protein-coupled receptors 3. Enzyme-coupled receptors Enzyme-coupled Receptors Cell-surface receptors 1. Ion-channel-coupled receptors 2. G-protein-coupled receptors 3. Enzyme-coupled receptors Cell-surface receptors allow a flow of ions across the plasma

More information

BIOL212 Biochemistry of Disease. Metabolic Disorders - Obesity

BIOL212 Biochemistry of Disease. Metabolic Disorders - Obesity BIOL212 Biochemistry of Disease Metabolic Disorders - Obesity Obesity Approx. 23% of adults are obese in the U.K. The number of obese children has tripled in 20 years. 10% of six year olds are obese, rising

More information

AP Biology Cells: Chapters 4 & 5

AP Biology Cells: Chapters 4 & 5 AP Biology Cells: Chapters 4 & 5 Multiple Choice Identify the choice that best completes the statement or answers the question. 1. The was the first unifying principle of biology. a. spontaneous generation

More information

Close to site of release (at synapse); binds to receptors in

Close to site of release (at synapse); binds to receptors in Chapter 18: The Endocrine System Chemical Messengers 1. Neural 2. Endocrine 3. Neuroendocrine 4. Paracrine 5. Autocrine Endocrine System --Endocrine and nervous systems work together --Endocrine vs. Nervous

More information

Chapter 11: Enzyme Catalysis

Chapter 11: Enzyme Catalysis Chapter 11: Enzyme Catalysis Matching A) high B) deprotonated C) protonated D) least resistance E) motion F) rate-determining G) leaving group H) short peptides I) amino acid J) low K) coenzymes L) concerted

More information

Chapt. 10 Cell Biology and Biochemistry. The cell: Student Learning Outcomes: Describe basic features of typical human cell

Chapt. 10 Cell Biology and Biochemistry. The cell: Student Learning Outcomes: Describe basic features of typical human cell Chapt. 10 Cell Biology and Biochemistry Cell Chapt. 10 Cell Biology and Biochemistry The cell: Lipid bilayer membrane Student Learning Outcomes: Describe basic features of typical human cell Integral transport

More information

CELLS. Cells. Basic unit of life (except virus)

CELLS. Cells. Basic unit of life (except virus) Basic unit of life (except virus) CELLS Prokaryotic, w/o nucleus, bacteria Eukaryotic, w/ nucleus Various cell types specialized for particular function. Differentiation. Over 200 human cell types 56%

More information

Lipid Chemistry. Presented By. Ayman Elsamanoudy Salwa Abo El-khair

Lipid Chemistry. Presented By. Ayman Elsamanoudy Salwa Abo El-khair Lipid Chemistry Presented By Ayman Elsamanoudy Salwa Abo El-khair 6 1. By the end of this chapter the student should be able to: define lipids. describe the biological importance of lipids. point out basic

More information

SREBP2 AND REVERB-ALPHA REGULATION OF HUMAN CYP8B1. A thesis submitted. to Kent State University in partial. fulfillment of the requirements for the

SREBP2 AND REVERB-ALPHA REGULATION OF HUMAN CYP8B1. A thesis submitted. to Kent State University in partial. fulfillment of the requirements for the SREBP2 AND REVERB-ALPHA REGULATION OF HUMAN CYP8B1 A thesis submitted to Kent State University in partial fulfillment of the requirements for the Degree of Master of Sciences By Hailin Feng December, 2009

More information

Part 1 Risk Factors and Atherosclerosis. LO1. Define the Different Forms of CVD

Part 1 Risk Factors and Atherosclerosis. LO1. Define the Different Forms of CVD Week 3: Cardiovascular Disease Learning Outcomes: 1. Define the difference forms of CVD 2. Describe the various risk factors of CVD 3. Describe atherosclerosis and its stages 4. Describe the role of oxidation,

More information

Chapter 8. Functions of Lipids. Structural Nature of Lipids. BCH 4053 Spring 2001 Chapter 8 Lecture Notes. Slide 1. Slide 2.

Chapter 8. Functions of Lipids. Structural Nature of Lipids. BCH 4053 Spring 2001 Chapter 8 Lecture Notes. Slide 1. Slide 2. BCH 4053 Spring 2001 Chapter 8 Lecture Notes 1 Chapter 8 Lipids 2 Functions of Lipids Energy Storage Thermal Insulation Structural Components of Membranes Protective Coatings of Plants and Insects Hormonal

More information

1. (a. Homeostasis / b. Feedback) is a state of constancy of conditions inside the human body

1. (a. Homeostasis / b. Feedback) is a state of constancy of conditions inside the human body PLEASE BE AWARE CONTENT COVERED ON EXAMS VARIES FROM ONE SEMESTER TO ANOTHER. THIS EXAM MAY NOT CONTAIN MATERIAL THAT WILL BE ON YOUR EXAM THIS SEMESTER, AND/OR MAY CONTAIN MATERIAL THAT WILL NOT BE COVERED

More information

Lecture 5: Drug targets (continued)

Lecture 5: Drug targets (continued) Lecture 5: Drug targets (continued) IIa. Enzymes as drug targets (HMG-CoA example) Many drugs are inhibitors of enzymes that catalyze biologically important reactions. The conversion of HMG-CoA to mevalonic

More information

GENERAL CHARACTERISTICS OF THE ENDOCRINE SYSTEM FIGURE 17.1

GENERAL CHARACTERISTICS OF THE ENDOCRINE SYSTEM FIGURE 17.1 GENERAL CHARACTERISTICS OF THE ENDOCRINE SYSTEM FIGURE 17.1 1. The endocrine system consists of glands that secrete chemical signals, called hormones, into the blood. In addition, other organs and cells

More information

- Biosignaling: Signal transduction. References: chapter 8 of Lippincots chapter 1 3 of Lehningers

- Biosignaling: Signal transduction. References: chapter 8 of Lippincots chapter 1 3 of Lehningers Basic concepts of Metabolism Metabolism and metabolic pathway Metabolic Map Catabolism Anabolism - Regulation of Metabolism Signals from within the cell (Intracellular) Communication between cells. - Biosignaling:

More information

FIRST BIOCHEMISTRY EXAM Tuesday 25/10/ MCQs. Location : 102, 105, 106, 301, 302

FIRST BIOCHEMISTRY EXAM Tuesday 25/10/ MCQs. Location : 102, 105, 106, 301, 302 FIRST BIOCHEMISTRY EXAM Tuesday 25/10/2016 10-11 40 MCQs. Location : 102, 105, 106, 301, 302 The Behavior of Proteins: Enzymes, Mechanisms, and Control General theory of enzyme action, by Leonor Michaelis

More information

BCMB 3100 Fall 2013 Exam III

BCMB 3100 Fall 2013 Exam III BCMB 3100 Fall 2013 Exam III 1. (10 pts.) (a.) Briefly describe the purpose of the glycerol dehydrogenase phosphate shuttle. (b.) How many ATPs can be made when electrons enter the electron transport chain

More information

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM The LDL Receptor, LDL Uptake, and the Free Cholesterol Pool

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM The LDL Receptor, LDL Uptake, and the Free Cholesterol Pool ANSC/NUTR 618 LIPIDS & LIPID METABOLISM The, LDL Uptake, and the Free Cholesterol Pool I. Michael Brown and Joseph Goldstein A. Studied families with familial hypercholesterolemia. B. Defined the relationship

More information

BIOLOGICAL MOLECULES REVIEW-UNIT 1 1. The factor being tested in an experiment is the A. data. B. variable. C. conclusion. D. observation. 2.

BIOLOGICAL MOLECULES REVIEW-UNIT 1 1. The factor being tested in an experiment is the A. data. B. variable. C. conclusion. D. observation. 2. BIOLOGICAL MOLECULES REVIEW-UNIT 1 1. The factor being tested in an experiment is the A. data. B. variable. C. conclusion. D. observation. 2. A possible explanation for an event that occurs in nature is

More information

Biomolecules. Unit 3

Biomolecules. Unit 3 Biomolecules Unit 3 Atoms Elements Compounds Periodic Table What are biomolecules? Monomers vs Polymers Carbohydrates Lipids Proteins Nucleic Acids Minerals Vitamins Enzymes Triglycerides Chemical Reactions

More information

Ecosanoids: Prostaglandins and related compounds

Ecosanoids: Prostaglandins and related compounds Ecosanoids: Prostaglandins and related compounds Presented by The requirements for the Pharmaceutical Biochemistry II Philadelphia University Faculty of pharmacy Ecosanoids: Prostaglandins and related

More information

Biochemistry: A Short Course

Biochemistry: A Short Course Tymoczko Berg Stryer Biochemistry: A Short Course Second Edition CHAPTER 27 Fatty Acid Degradation Dietary Lipid (Triacylglycerol) Metabolism - In the small intestine, fat particles are coated with bile

More information

Metabolic Syndrome. DOPE amines COGS 163

Metabolic Syndrome. DOPE amines COGS 163 Metabolic Syndrome DOPE amines COGS 163 Overview - M etabolic Syndrome - General definition and criteria - Importance of diagnosis - Glucose Homeostasis - Type 2 Diabetes Mellitus - Insulin Resistance

More information

Steroid Hormones Synthesis

Steroid Hormones Synthesis *I ll try my best to incorporate the Slides in this Sheet; you don t need to study the slides if you study this sheet. Steroid Hormones Synthesis - The figure to the right is the Steroid nucleus, it has

More information

Antihyperlipidemic Drugs

Antihyperlipidemic Drugs Antihyperlipidemic Drugs Hyperlipidemias. Hyperlipoproteinemias. Hyperlipemia. Hypercholestrolemia. Direct relationship with acute pancreatitis and atherosclerosis Structure Lipoprotein Particles Types

More information