Adrenal Zona Glomerulosa Targeting in Transgenic Mice

By

Jeniel Parmar

Submitted to the Faculty of the School of Graduate Studies

of the Medical College of Georgia

in partial fulfillment of the Requirements of the

Degree of Doctor Philosophy

December, 2009

V Adrenal Zona Glomerulosa Targeting in Transgenic Mice

This dissertation is submitted by Jeniel Parmar and has been examined and approved by an appointed committee of faculty of the School of Graduate Studies of the Medical College of Georgia.

The signatures which appear below verify the fact that all the required changes have been incorporated and that the thesis/dissertation has received final approval with reference to content, form, and accuracy of presentation.

This dissertation is therefore in partial fulfillment of the requirement for the degree of Doctor of Philosophy.

Date Major Advisor - William E. Rainey, 8

Dept. Chair - R Clinton Webb, PhD

Dean, School of Gr te Studies - Gretchen Caughman, PhD

VI JENIEL PARMAR Adrenal Zona Glomerulosa Targeting in Transgenic Mice (Under the guidance of WILLIAM E. RAINEY II~ Ph.D.)

The final step in the production of aldosterone is performed by the enzyme aldosterone synthase (CYP11 B2)-. CYP11 B2 is primarily expressed in the zona glomerulosa (ZG). of the adrenal cortex. Adrenocortical, expression of CYP11 B2 is primarily regulated by circulating levels of angiotensin II (Ang II) and K+, but the molecular mechanisms that control its ZG-specific expression are not clearly defined. Con$iderable in vitro analyses have been performed towards defining the mechanisms that control CYP11 B2 expression. Previous studies from our laboratory and others have identified ·several cis-regulatory elements on the 5' · flanking promoter region (at -71/64, -129/114, -351/343 and -773/766) that regulate basal expression as well as maximal stimulation of CYP11 B2 transcription. Moreover, key· transcription factors that bind these cis-regulatory regions including NGFIB, NURR1, SF-1 and COUP-TF have also been identified.

Hence, through several in vitro analyses, a considerable evidence exists supporting the contention that these regulatory elements found within the 5' flanking promoter region may control ZG-specific expression of CYP 11 B2 gene.

However, thus far, all evidence is based on in. vitro analyses of transcriptional regulation, which does not always depict in vivo_occurrences.

I To initiate our in vivo assessment of CYP1182 promoter, we began by comparing the DNA sequences between human, mouse, and rat CYP1182 , which interestingly revealed high sequence similarity in the ·5' flanking promoter region of the CYP1182 gene. This result suggested that the cis­ regulatory regions identified by in vitro analyses likely plays an important role in

CYP1182 ZG-specific gene expression. Therefore, we generated transgenic mouse lines by pronuclear injection of a Transgenic (Tg) DNA construct containing 985 base pairs (bp) of the mouse Cyp11 b2 promoter driving expression of a Lacz reporter gene.· Importantly, 4 founder Tg mouse lines revealed Lacz expression exclusively in the adrenal ZG. Mice fed a normal sodium diet (0.3 %) and a low sodium diet (0.03 %) showed Lacz mRNA expression exclusively in adrenal tissue. Furthermore, (3-galactosidase protein

(the product of LacZ) was localized solely in the ZG of the Tg mice. Hence, the role of the proximal promoter region of the Cyp11 b2 gene was confirmed, in vivo, as this region allowed induction of Lacz exclusively in the adrenal ZG of Tg mice.

Moreover, with the expression of Lacz properly restricted to adrenal ZG, we concluded that regions required for Cyp11 b2 gene repression in the adjacent inner two zones of the adrenal cortex were also confined within the 985 bp promoter. This regulatory fragment. will be an invaluable tool for adrenal ZG targeting of genes believed to play a role in adrenocortical diseases and aldosterone dysregulation.

II While developing Tg mice, we also focused on characterization and development

of novel adrenocortical cell lines. As aforementioned, in vitro culture models have

allowed a multitude of studies that have broadened our understanding of normal

adrenocortical endocrine function. Primary cultures of adrenocortical cells have

been an excellent source for in vitro studies. However, the eventual onset of

senescence in primary cultures of cells creates a recurring need for the costly

· and difficult isolations of fresh adrenocortical cells. Hence, the use of primary

cultures has been increasingly supplemented by immortalized cell lines. We

utilized an adrenocortical carcinoma to develop a human adrenocortical cell line.

We entitled it the human adrenocortical carcinoma cell line clone 15 (HAC15).

HAC15 represents only the second human adrenocortical cell line available that

exhibits physiological hormonal responses, steroid.ogenesis, and expression of

steroid-metabolizing enzymes. The ability of HAC15 to respond to Ang II, K+, and

ACTH makes it the first adrenal cell line capable of responding to the three main

physiologic regulators of the adrenal cortex.

III INDEX WORDS: Adrenal cortex, aldosterone, aldosterone synthase, transgenic mice, Lacz, cell lines, in vitro model systems.

NOMENCLATURE GUIDLINES: Throughout this dissertation, names of all human genes and proteins will be fully capitalized (e.g., CYP11 B2, CYP11 B1, and CYP17)-. The names of all rodent genes and proteins will be depicted with only the first letter capitalized (e.g., Cyp11 b2, Cyp11 b1, and Cyp17).

IV TABLE OF CONTENTS

Page

ACKNOWLEDGEMENTS ...... IX

LIST OF FIGURES ...... X

LIST OF TABLES ...... XI I

LIST OF ABBREVIATIONS ...... ■- •• ___ •••••••••••••• XI U

I. INTRODUCTION

A. Statement of the Problem: CYP1182 Gene Regulation ...... _.. 1

1) Specific Aim I ...... 3

2) Review of Literature and Rationale ...... 4 ·

B. Statement of the Problem: A Paucity of Adrenal Cell Lines .... 20

1) Specific Aim 11 •••••••••••••••••••••••••••••••••••••••••••••••••••••••••••••• 22

2) Review of Literature and Rationale ...... 23

II. MANUSCRIPTS·

A. Peer Reviewed Articles ...... 30

1) "Gene targeting to the mouse adrenal zona glomerulosa"

VII 2) "Development of an ACTH Responsive Human Adrenocortical Carcinoma Cell Line"

8. Invited Review Article ...... 76

"Comparisons of Adrenocortical Cell Lines as in vitro Test

Systems"

C. Book Chapter ...... 107

"Adrenocortical Cell Lines"

Ill. DISCUSSION

A. Specific Aim I: CYP1182 Gene Regulation ...... 146

B. Specific Aim II: A Paucity of Adrenal Cell Lines ...... 155

IV. BIBLIOGRAPHY ...... ~ ...... 162

VIII ACKNOWLEDGEMENTS

I wish to express my sincerest gratitude to Dr. William E. Rainey for taking me under his wing and guiding me throughout my graduate career. Seldom does one have the opportunity to learn from a true advisor. I deeply value the wisdom and scientific expertise that he bestowed upon me. I will miss his enthusiasm and ceaseless support. The experience will surely never be forgotten.

I am indebted to Dr. Tsugio Seki for his kind technical as well as advisory support.

I would also like to thank my committee members Dr. Edward Inscho for never letting me forget about the kidneys, Dr. Wendy Bollag for regularly reminding me about the importance of primary cell cultures, and Dr. Lawrence Layman for teaching me the value of statistical analyses. My appreciation is extended to the readers of my thesis Dr. Michael Edwards and Dr. Celso Gomez-Sanchez for willingly provide selfless support.

I would like to thank the faculty of the department of Physiology at the Medical College of Georgia for providing me with an exceptional education. I would also like to thank the past and present Rainey lab members and our department management including Anthony, Jennifer, Daniel, Claudia, Rebecca, Nick, Bobbie, Kathy, Faye, Valerie, and Brenda, who have all been a distinguished part of my life in the past several years.

Finally, a special word of appreciation must be extended to my family. I would like to thank my parents for their seemingly inexhaustible faith and understanding· during my education. I would like to thank my best friend and brother, Hiren, and his wife, Jigna, for their eternal support and their cheerful and enthusiastic endorsements. I would also like to thank my friend, companion and wife, Ami, for her love and compassion.

IX LIST OF FIGURES

Figure Page

1. Phylogenetic an.cestral lineages ...... 5

2. H&E stained mouse adrenocortical tissue ...... 10

3. Major adrenocortical steroidogenic pathways ...... 12

4. Glucocorticoid suppressible hyperaldosteronism model ...... - ...... 16

5. Cyp11 b2 evolutionary conserved regions (ECR) ...... 18

6. Cyp11 b2 ECR analysis and Tg construct (manuscript A 1) ...... 53

7. Cyp11 b2 transcript quantity (manuscript A1 } ...... · ...... •...... 54

8. Cyp11 b2 localization in adrenal cortex (manuscript A 1) ...... 55

9. X-gal stained Tg adrenals under LS diet (manuscriptA1) ...... ' ...... 56

10.Co-localization of Cyp11b2 and X-gal in Tg mice (manuscript A1} ...... 57

11.Amplification curve of Cyp11b2 (manuscriptA) ...... : ...... 58

12.HAC15 and H295R cell comparison (manuscript A2) ...... ~ ...... - .... 74

13. HAC15 cell line enzymatic expression assessment (manuscript A2) ...... ~ .. 75

14. Human a_drenal steroid biosynthetic pathways (manuscrip~ _,B) ...... 104

15. Human adrenal cell signaling pathways (manuscript B) ...... · ...... 105

16. Rodent adrenal steroid biosynthetic (manuscript 8) ...... ~ ...... 106

17. Intracellular adrenocortical cell signaling (manuscript C) ...... 140

18. Phase contrast images of H295R and HAC cells (manuscript C) ...... 141

X 19. Human adrenocortical enzymatic pathway (manuscript C)-...... 142

20. Development and isolation of HAC clones (manuscript C) ...... 143

21. Cytogenetic profiles depicted through karyotypes (m~nuscript C}...... 144

22. Murine adrenocortical enzymatic pathway (manuscript C)...... 145

XI ·LIST OF TABLES

Table Page

I. Plasma aldosterone concentrations in WT mice (manuscript A 1) ...... 49

II. Plasma aldosterone concentrations in Tg mice (manuscript A 1) ...... 49

m. Relative transgene mRNA expression (manuscript A 1) ...... 49

N. Rodent and bovine adr~na~ cell lines (manuscript B)...... 101

V. Human adrenal cell lines (manuscript B)...... 102

VI. All adrenocortical cell lines (manuscript C)...... 136

VII. Cytogenetic analyses of HAC15 and H295R cells (manuscript C)...... 138

XII LIST OF ABBREVIATIONS

• ACTH: adrenocorticotropin • NSAID: non-Steroidal anti-inflammatory

• ANOVA: analysis of variance drugs

• Ang 11: angiotensin 11 • PAGE: polyacrylamide gel

• ATCC: american Type Culture electrophoresis

Collection • PCR: polymerase chain reaction

• cAMP: cyclic AMP • RAS: rennin-angiontensin-system

• cDNA: complementary DNA • RIA: radioimmunoas_say

• CRH: corticotropin~releasing hormone • RT-PGR : reverse transcription-PCR

• CYP 11 B 1 : 11 ~ -hyd roxylase • mRNA: messenger RNA

• CYP1182: aldosterone synthase • rRNA: ribosomal RNA

• DNAase: deoxyribonuclease . • RNase: ribonuclease

• DMSO: dimethylsulfoxide • SE: standard error

• EDTA: ethylenediaminetetraacetic acid • T g: transgenic

• ELISA: enzyme-linked immunosorbent · • qPCR: quantitative PCR

assay • ZF: zona fasciculata

• EMSA: elecrophoretic mobility shift • ZG: zona glomerulosa

assay · • ZR: zona reticularis

• HAC: human adrenocortical carcinoma

• K+: potassium

XIIl I. INTRODUCTION

A. Statement of the problem: CVP11 B2 gene regulation

In humans, the biosynthesis of cortisol and aldosterone is dependent on two key en:zymes: 11 ~-hydroxylase (CYP11 B1) and aldosterone synthase (CYP11 B2}. In humans the coding sequences of these two genes are 95 % identical, their non­ coding intronic sequences are 90 % identical, and their encoded amino acid

5 8 sequences are 93 % identical - • Due to the high shared by the two isozymes, only mouse and rat aldosterone synthase antibodies have

9 been developed, to date, which can exclusively detect the respective protein •

Interestingly, despite this similarity, CYP11 B1 is exclusively expressed in the zona fasciculata (ZF), while CYP11 B2 is exclusively expressed in the zona glomerulosa (ZG). The molecular mechanisms that cause tissue-specific expression and repression of CYP11 B2 are unknown. However, it is clear that the sequence similarity between the 5' flanking promoter regions of CYP11 B2

10 11 and CYP11 B1 genes is significantly reduced • • Perhaps the promoter regions of the two genes harbor discrete transcription factor binding sites that allow for the differential regulation of transcription. Therefore, we hypothesized that this dissimilarity accounts for the differential zone-specific distribution of these two

12 14 genes within the adrenal cortex - • Defining the mechanisms that allow zone­ specific expression and repression of CYP 11 B2 should provide important

1 information with regard to its aberrant expression in human adrenal diseases.

Dys regulation of CYP 11 B2 expression plays a key role in aldosterone-producing·

15 16 _adenomas (APA} and glucocorticoid suppressible hyperaldosteronism (GSH} , both of which lead to primary aJdosteronism (PA), the most common cause of endocrine hypertension.

A DNA sequence comparison between mouse and rat CYP11 B2 genes revealed high sequence similarity in the 5' flanking promoter region of the

CYP11 B2 gene. Aforementioned, the greatest dissimilarity between the highly conserved CYP11 B1 and CYP11 B2 genes lies within the 5' flanking promoter region; concurrenUy, the same promoter region of CYP11 B2 in a number _of species is also highly similar. Collectively, this suggests that as CYP11 B1 and

CYP11 B2 evolutionarily diverged into two distinct enzymes, the zone-specific regulation of the two isozymes must have been established and maintained in closely related species. One aim has been proposed to determine the CYP11 B2 promoter regions that are required for its tissue-specific expression, as well as repression, within the adrenocortical zones. These studies should broaden our knowledge of adrenocortical zonation under physiological conditions and provide insights into human adrenal diseases.

-2 1) Specific aim I

As in humans, Cyp11 b2 in mice is solely expressed in the ZG and its expression

is repressed in the adjacent ZF and zona reticularis (ZR), of the adrenal cortex.

· Herein, an in vivo model to define the regions responsible for the ZG-specific

expression of Cyp11 b2 gene was sought.

Hypothesis: The regulatory regions within the 5' flanking promoter of Gyp 11 b2

are sufficient for restricted adrenal ZG-specific expression.

a. We demonstrated that the 985 bp Cyp11 b2 promoter region was sufficient

for ZG-specific targeting. The 985 bp Cyp11 b2 promoter-driven Lacz

reporter gene was detected solely in_ the ZG of the adrenal cortex. J3-

galactosidase (x-gal) staining analysis was confirmed by quantitative real

time PCR (qPCR}.

b. The 985 bp Cyp11 b2 promoter region was also sufficient for ZF- and ZR­

specific repression. Lacz was -not expressed in ZF and ZR. Upon qPCR

examination of 16 key mouse tissues, no aberrant extra-adrenal Lacz

transcript was found.

3 2) Brief literature review and rationale: CVP11 B2 gene regulation

The adrenal gland, an integral part of the endocrine system, evolved to regulate essential physiological functions. According· to the earliest descriptions of adrenal function, Agnathans (lampreys) were among the first animals to have developed adrenal steroid hormone receptors-the ancestral

17 18 corticosteroid receptor (CR)-about 470 million years ago (mya} • • The ligand for this ancestral CR is yet to be elucidated; consequently, the function of CR remains to·- be established in lampreys. Subsequently, a whole genome wide duplication event is believed to have occurred in the CR lineage giving rise to

.. glucocorticoid receptor (GR) and mineralocorticoid receptor (MR)- homologues in the Gnathostomatas (sharks and skates) 440 mya 19 (Fig._ 1A). Both GR and MR

3 have been cloned and characterized in the ancestral skate, Raja erinacea •

Moreover, a number of studies with sharks and skates indicate the formation of

20 21 corticosterone and 1a-OH corticosterone, capable of binding GR and MR • •

Hence corticosterone is believed to have emerged as a ligand for MR _and GR; importantly, these results suggest the apparent presence of a key enzyme capable of 11 (3-hyroxylase activity since 440 mya. This · ancestral enzyme capable of 11 (3-hydroxylation was designated to ~he cytochrome P450, family 11, and subfamily B (CYP11 B). A distinct role for the CYP11 B enzyme expands with

22 23 the appearance of cortisol in Osteichthyes (bony fish) 420 mya • • Presumably as evolution progressed, stress due to a vast array of extreme environmental and physiological changes necessitated the emergence of cortisol for assistance in .

4 A GR 300 F, Aldo CYPH 81 4¥" MR mya ·Doc CYP11B2 Amniotes · Reptiles; birds, and mammals

GR 350 F, Aldo CYP11 B Tetrapods Amphibians MR mya- DOC

GR 400 F, Aldo CYP11 B Sarcopterygii _Lung fish ,, ' MR mya DOC GR 420 F CYP11 B Osteichthyes Bony fish ,.. MR mya Doc

_,_. GR 440 1a-OH CYP B 11 MR mya B, B Gnathostomatas Cartilaginous fish-Sharks and Skates

41Q ( 470· CR ??? ??? . Agnathans · Jawless fish-Lampreys and Hagfish mya

B Frog CYP118 Bovine CYP11 B

CYP11B Human CYP11 B1

Human CYP11B2

Mouse CYP11 B1 Rat CYP11B1 Rat CYP11B3 Mouse CYP11 B2

Rat CYP11 B2 ·

50 40 30 20 10 0 Time (million years) Present day _

Fig. 1: Phylogenetic ancestral lineages. Panel A: evolutionarily · led major transitions in adrenal steroid· hormone receptors,. adrenal steroid hormones, and 1 3 adrenocortical enzymes · • Functional role of the adrenal gland increases with growth in complexity of vertebrate evolution. Abbreviations: · corticosteroid receptors (CR)-, glucocorticoid receptors (GR), mineralocorticoid receptors (MR), cortisol (F), aldosterone (Aldo), deoxycorticosterone (DOC), and corticosterone 4 (B). Panel B: mammalian phylogenetic tree of the CYP11 B subfamily •

. - 5 coping with perpetual stress. In terms of migration of aquatic vertebrates from

water to land roughly 400 mya, the need-- for mineral and water homeostasis at

the systemic and cellular levels must have risen, which necessitated- a salt- and

water-retaining hormone. In both African and Australian Sarcopterygii (lungfish),

aldosterone, cortisol, and DOC are detected in plasma by double-isotope

24 25 derivative assay (DIDA) and radio-immunoassay (RIA) • • At the outset, the

mineralocorticoid effect of aldosterone exerted at the levels of gills in these

lungfish, where it controls the chloride ion exchange and Na+/K+ ATPase activity

26 • Studies also indicate the ·control of aldosterone secretion by a functioning . ~

renin-angiotensin-system (RAS) as both angiotensin I (Ang I). and Ang 11

24 27 stimulate the release of aldosterone in lungfish • • Intriguingly, this suggests

that the RAS likely played an important role in the aquatic to terrestrial transition

) of vertebrate evolution. It would seem reasonable to- presume that aldosterone

provided a selective sodium- and water-sparing advantage to the first aquatic

invaders venturing into the terrestrial habitats, where both were fluctuating

resources. A comparative genomic approach supports the hypothesis that

phylogeny subsequent to lungfish paved the way 400 mya and gave rise to

28 29 Tetrapods (amphibians) and Amniotes (reptiles, birds, and mammals) • • These

tremendous biological changes enforced the distribution of MR and GR to

various anatomical locations in an array of species according to their habitation.

Aldosterone and corticosterone promote transepithelial sodium and water re­

absorption from the skin and urinary bladders of Anurans (frogs and toads},

31 which do not drink water3°· • Reptiles have nasal salt secreting gland in which

6 corticosterone increases sodium secretion while aldosterone decreases it to

2 stimulate sodium- and water-reabsorption through kidney and urinary bladder3 •

Birds primarily utilize both aldosterone and corticosterone' to· induce sodium

33 secretion and excretion from the nasal glands as they lack urinary bladders •

Hence, corticosteroids may target various osmoregulatory organs and have variable actions in fish, amphibians, reptiles, and birds as compared to mammals.

Common features between mammalian and fish adrenocortical function are likely to predate our understanding of corticosteroid function back 400 mya, · when mammals and fish last shared a common ancestor. Nevertheless, most

(but not all) mammalian species, including humans, have resumed the adrenal synthesis of cortisol as the primary glucocorticoid and aldosterone as the primary mineralocorticoid, since earliest seen in lungfish. Synthesis of corticosteroids in lungfish is made possible by a single CYP11 B enzyme. Presumably, as the role of aldosterone as the salt- and water-sparing . hormone became critical for survival in the terrestrial mammals, in some species, the CYP11 B gene duplicated to originate the CYP11 B 1 and CYP1182 genes (Fig. 1B). The complexity of this system is increased even more in rat where a duplication has

34 led to a third gene, CYP1183 (Fig. 18) • The enzyme CYP1181, incapable of synthesizing aldosterone, was labeled 11 J3-hydroxylase as it 11 J3-hydroxylates

11-deoxycorticoids to yield cortisol and corticosterone. The enzyme CYP1182 was labeled aldosterone synthase as ~t performed 11 J3-hydroxylation, 18- hydroxlation and 18-oxidation to yield aldosterone. It is unclear why the ancestral

7 CYP11 B enzyme, capable of both aldosterone and cortisol synthesis, is still

expressed in some mammals such as cattle, sheep etc. Moreover, . some

mammalian species such as mice and rats have diverged about 100 mya,

utilizing corticosterone as their primary glucocorticoid instead of cortisol. Hence

the phylogenetic tree of the CYP 11 B gene subfamily remains elusive (Fig. 1B).

It is noteworthy that although terrestrial mammals rely on aldosterone for

sodium and water retention, marine mammals have an opposite osmoregulatory

role as they need to excrete the surplus of sodium. Immunoassay measurements

of Antarctic seals and whales show 20-100 times lower aldosterone secretion

35 36 than in terrestrial mammals • •

As the evolutionary tree expanded becoming more complex so did the

1 function, anatomical location, and zonation of the adrenal gland • Hardisty

et al. have shown in lampreys that putative adrenocortical cells are located as

37 loose pockets around the great dorsal vessels , representing the least

specialized system in terms of structural organization of the adrenal gland among

_all vertebrates. These adrenocortical cells have an ultra-structure of typical

steroid-secreting cells with abundant lipid droplets, extensive smooth

endoplasmic reticulum, and they are rich in tubular and transverse mitochondrial

18 cristae . Sharks and skates have islets of adrenocortical cells located between

38 the two kidneys, referred as the interrenal gland • Bony fish have patches of

dispersed adrenocortical cells located within their nephritic and hemopoatic

20 tissues • In African as well as Australian lungfish, adrenocortical cells appear in

8 gross· dissection as small, yellow-pigmented regions within the kidney, referred

39 as . the intrarenal glands • Histologic preparations demonstrate that these pigmented regions have irregularly organized clusters of lipid-contain1_ng. cells

40 41 and that these cells are immuno-reactive to adrenal-specific enzymes • • A positive enzymatic reaction during incubation of frozen sections is considered to be the strongest proof of potential for steroidogenesis.

In terms of location of adrenocortical cells, all possible anatomical locations discussed above are represented by different species of amphibians,

42 intrarenal and interrenal glands • Reptiles and birds represent the first vertebrates to have developed separate adrenal glands atop of the kidneys, however, their chromaffin cells are not encapsulated by the cortical cells, rather the . adrenal is a mixture of the two: In all mammals, the adrenal gland is subdivided into two different compartments-the outermost cortex and the innermost medulla. Consequently, only the mammals contain a well-defined adrenal cortex that surrounds the adrenal medulla, while all the lower vertebrates have mixed cortical and chromaffin cells.

The histological aspects of the adrenal cortex vary to some extent among different mammalian species. However, in general, it has been known since the end of the 19th century that the adrenal cortex is divided into three concentric

43 zones; ZG, ZF, and ZR (from_ outer- to inner-most} • Each zone has been histologically separated and characterized. The ZG appears to be irregularly structured with groups of rounded and columnar shaped cells (Fig. 2). The ZG . cells also appear to have round, dark nuclei and a much smaller cytoplasm as

9 compared to the other two zones. Cells in the ZF are polyhedral with a foamy appearance due to the abundance of lipid droplets (Fig. 2}. The ZF appears, to be radially arranged into cords of cells _separated by fenestrated sinusoidal

_capillaries. Cells of the ZR

appear to be smaller than

ZF and uneven~

arranged. The Nuclei of

cells in the ZR appear

well-rounded and due to

the small cell size there is

a larger nuclear to

cytoplasmic ratio (Fig. 2).

Including histological divisions, each zone clearly has biochemical differences that are responsible for their distinctive functions_~ Each zone is responsible for the production and biosynthesis of unique steroid hormones. In the ZG, cholesterol is used to synthesize mineralocorticoids (e.g., aldosterone) in response to Ang II and K+ stimulation. In the ZF,, cholesterol is metabolized into glucocorticoids (e.g., cortisol in humans and corticosterone in mice) _under the regulation of adrenocorticotropin hormone (ACTH). In humans, the ZR utilizes cholesterol to synthesize the adrenal androgens (e.g., DHEA and DHEA-sulfate), regulated by

ACTH and other unknown factors. While histological and biochemical distinctions have been evident for quite some time, the molecular mechanisms that are

10 responsible for these differences remain unclear. Thus, the question remains as

to how the zones are able to regulate the exclusive production of specific steroid

hormones without a physical barrier separating each zone.

Functional zonation relies, in part, on the zone-specific expression of the

unique adrenocortical enzymes. Zonation functionally relates to production of

unique steroids, which result from the differential expression of steroid­

metabolizing enzymes. Common to all adrenocortical pathways is the rate­

limiting step; the translocation of cholesterol from the outer mitochondrial

membrane into the mitochondrion for side-chain cleavage by the cholesterol

side-chain cleavage enzyme (CYP11A1) (expressed throughout the adrenal

44 cortex) • On the contrary, CYP1182 is solely expressed in the_ ZG of the adrenal

cortex, and it represents a key regulatory enzyme in the aldosterone biosynthetic

11 45 pattiway ' • As shown in Figure 3, the three main adrenocortical steroid

hormones share certain enzymes within the biosynthetic pathway. In other

words, many of the intermediate enzymes needed to produce a particular steroid

· hormone are common to all of the zones. Taking this fact into consideration, if

there were even small ectopic expression of CYP1182 in the much larger ZF

zone, it would result in an excess of circulating aldosterone. As with CYP1182, if

CYP17 were expressed in the ZG, cortisol would be produced in the ZG. In

humans, the isozyme of CYP1182, CYP1181, is modestly expressed in ZG

46 cells , where it catalyzes 18-hydroxylation poorly and is unable to perform 18-

oxidation; therefore, CYP1181 is not able to produce aldosterone. The ZG of

11 47 mice and rats are .devoid of CYP1181 enzyme activity • Due to its role in glucocorticoid production, CYP1181 expression· is significantly elevated in the

8 ZF • Therefore, the spatial segregation of biosynthesis pathways through zonation of the adrenal is one way to control· the relative production rates of mineralocorticoids and glucocorticoids. The molecular mechanisms that cause

Cholesterol

• 11-DO~ Aldosterone

17a-hydroxy 17a-hydroxy 11-deoxycortis.ortisol Pregnenolone Progesterone

Location of enzymes: CHEA Mitochondrial

Smooth endoplasmic reticulum

Fig. 3: Major human adrenocortical steroidogenic pathways. Nomenclature for the steroidogenic enzymes are as follows: CYP11 A 1, cholesterol side-chain cleavage; CYP17, 17a-hydroxylase- 17, 20-lyase; HSD382, 3(3-hydroxysteroid dehydrogenase type II; CYP21, 21-hydroxylase; CYP1181, 11 (3-hydroxylase; CYP1182, aldosterone synthase. the zone-specific expression pattern of the adrenocortical enzymes are yet to be_ resolved. It has been demonstrated in vitro that the regulatory regions within the

5' flanking regions control the relative expression of these genes required for

6 11 steroidogenesis in each of the zones • •

12 Regulatory regions within the human CYP1182 promoter have been

11 12 studied and several elements have been identified ' • The earliest_ work on defining the regulatory regions of the adrenocorticaJ steroidogenic. enzymes was performed utilizing bovine ~nd porcine CYP11 B genes. Unlike humans and rodents, who express CYP11 B1 and CYP11 B2 genes, cattle, ·sheep, and pigs

4 48 only have one CYP11 B gene that is expressed in both ZG and ZF • . The same

CYP11 B gene product is responsible for the production of mineralocorticoids in the ZG and glucocorticoids in the ZF through mechanisms that are not yet clear.

However, considerable progress has been made towards understanding the regulation of CYP11 B gene transcription in the· adrenal cortex. Omura and colleagues have assessed the 5' flanking· CYP11 B gene promoter and identified

49 52 six different cis-regulatory elements, termed adrenal (Ad) 1-6 - • Through several in vitro deletion and mutation analyses of the .5' flanking region of the

CYP11 B promoter, Ad1 and Ad4 elements have been identified to be the most important regulators of gene transcription.

Consequently, as with the CYP11 B gene promoter, the 5-' flanking region of the CYP11 B1 and CYP11 B2 gene promoters have ·also been analyzed in

47 53 54 55 56 detail for rodents • • and humans • • Clyne and colleagu·es have utilized

H295R-a human adrenocortical carcinoma cell line-to assess the role .of the promoter region of the CYP11 B2 gene. H295R cells, transiently transfected and stimulated with Ang II or· K+, increase the expression of luciferase reporter constructs prepared with variable lengths of the CYP11 B2 promoter; similar

13 2 57 58 increases are ·observed with cAMP · and, Ca + stimulation • • Reporter. constructs of the CYP11 B2 promoter with serial deletions and mutations have been tested through electrophoretic mobility shift assays (EMSA), and DNase I footprinting experiments. Mutation of the Ad1 element at position -71 bp leads to a decreased induction by cAMP, Ang II, and K+, suggesting the importance of this element for both Ca2+- and cAMP-induced CYP11 B2 transcription. However, deletion of the Ad1 element does not terminate basal activity, suggesting requirements of other elements. Deletion or mutation of the Ad5 element at -129 bp in the CYP11 B2 promoter completely abolishes the basal and agonist­ stimulated transcription. DNA-protein interaction analyses· show that the Ad1 site resembles a cAMP response element (CRE) and binds CRE binding proteins

(CREB, ATF1 and ATF2), while the Ad4 sites bind steroidogenic factor-1 (SF-1) and Ad5 sites bind chicken ovalbumin upstream promoter transcription factor

54 56 59 61 (COUP-TF), among other transcription factors - • - •. Other Ad elements have been identified that bind transcription factors on the CYP11 B2 promoter; however, in vitro studies thus far do not support the role of these elements in

CYP11 B2 transcription. However, subsequent studies using the human

CYP11 B2 promoter have identified an additional cis-regulatory element that

55 62 binds the nuclear hormone receptor NURR1 • • In sum, three distinct cis­ regulatory elements on the CYP11 B2 promoter,· Ad1 (at -71/64), Ad5 (at -

129/114), and NBRE (at -766 /-759} are necessary for basal transcription; Ad5 site is also required for cAMP- and Ca2+-dependent signaling pathway induction.

14 · Analogous elements are present on the CYP1181 promoter; the Ad1 and

Ad4 sites are required for basal, as well as cAMP-ihduced, transcription of the

56 CYP1181 gene . The Ad1 site of the CYP11 B1 and CYP1182 promoters is nearly identical, differing in only two bp·, while the Ad4 site is exactly same between the two isozymes. As with CYP1182, CYP1181 Ad4 site binds SF-1; however, SF-1 plays a rather contrasting role in regulation of the two genes, as it promotes the transcription of CYP1181 and inhibits the transcription of CYP1182

54 61 through mechanisms that are still unknown • Collectively, the 5' flanking

promoter region of the CYP1181 and CYP1182 genes has evolutionarily

diverged just enough to control their zone-specific expression. Both genes rely

on the Ad1 site for basal transcription. The Ad5 site plays a major role in_

CYP1182 and not in CYP1181 transcription, while the Ad4 site plays a significant role in CYP1181 and not CYP1182 transcription. The NBRE and

NURR1 regulate CYP1182 but not CYP1181. Such differences in the cis­

regulatory elements on the 5' flanking regions of the two genes are likely to play

an important role in differential regulation of these isozymes within the adrenal cortex.

Consequently, the significance of the differences between the two genes

is affirmed by a genetic disease, glucocorticoid supressable hyperaldosteronism

(GSH), a rare but well understood cause of primary aldosteronism (PA).

Normally, the two CYP11 B genes (CYP11 B1 ahd CYP1182) are 10 kilo bp (kbp)

63 apart on 8 ; interestingly, in GSH, an unequal meiotic crossover occurs that produces carrying three CYP11 B genes: a wildtype

15 CYP11 B2, a wildtype CYP11 B1, and a chimeric CYP11 B2 gene that is under-the

14 64 control of CYP1181 5' flanking promoter region • (Fig.- 4}. Although rare, such

unequa~ meiotic crossovers occur among other genes leading. to several human

diseases. For example, a crossover on human chromosome 6 between 21-

C_YP1182 40-kb CYP11B1 l Chromosome Unequal meiotic 8 locus q24.3 :>; crossover l Chimeric gene CYP1182 CYP1181

Fig. 4: Glucocorticoid suppressible hyperaldosteronism (GSH) model. Crossover between CYP11 B2 and CYP11 B1 occurs during meiosis, resulting in one gamete that has a a chimeric gene with 5' flanking promoter sequence of CYP11 B1 and 3' seauence of CYP11 B2. Presence of a sinale chimeric aene could lead to GSH.

hydroxylase (CYP21) and a human leukocyte antigen family gene causes

65 congenital adrenal hyperplasia due to 21-hydroxylase deficiency • The unequal

crossover between CYP11 B1 and CYP11 B2 that leads to GSH produces a

chimeric gene that encodes an enzyme with aldosterone synthase activity, but

the gene is expressed in the ZF under the control of ACTH and not its typical

agonist (Ang 11)-. ACTH activates expression of the c~imeric enzyme increasing

the production of aldosterone, which can be inhibited upon administration of

glucocorticoids to suppr~ss circulating- ACTH levels. Importantly, in 1992, White

and colleagues showed that- the DNA sequence of this chimeric gene contains 5'

16 flanking regions of the CYP1181 gene _promoter, while the majority of the coding

6 sequence belongs to.the CYP1182 gene 1 • Since the chimeric gene is expressed-

. in the ZF under the control of ACTH while encoding. an enzyme with aldosterone

synthase activity, this strongly suggests that DNA sequences within the 5'

flanking promoter region of CYP11 B1 can _mediate its zone-specific expression.

One of my goals was to determine if a similar scenario is true for CYP1182.

As with GSH, another alternative mechanism leading to ectopic zonal

expression of CYP1182 has been reported in TASK1 (KCNK3; acid-sensitive K+

9 66 channel} deficient mice • • In these mice, CYP1182 is absent from the ZG and

expressed in the ZF of the female transgenic (Tg): mice, but not in male mice. In

addition, similar to GSH, these mice have ZF expression of both CYP11 B1 and

CYP1182 under the control of ACTH. This demonstrates another mechanism

leading to ectopic CYP1182 expression.

Recent literature reports inexplicable aldosterone increases in patients

67 69 with hypertension - ; which may in part be due to ·cYP1182 gene

70 74 dysregulation - • The exact mechanisms that cause low-renin expression of

adrenal CYP1182 in these patients have not been defined. Thus far, through in

vitro analyses, a wealth of knowledge has been attained regarding the normal

physiologic regulation· of the CYP1182 gene. However, progress towards

understanding the role of CYP1182 in human diseases continues to be slowed

by the lack of optimal in vivo models. As discussed in the subsequent pages, the

development of a better model to study CYP1182 was the goal of Aim two of my

research program.

17 Tg mice are an appropriate in vivo model to study CYP1182 gene

regulation. CYP1182 gene is expressed exclusively in the ZG of both humans

and mice, and its enzymatic regulation and activity are also highly similar

Fig. 5: Mouse Cyp11 b2 ECR analysis. A. Schematic representation of mouse Cyp11 b2 genomic structure. Boxes indicate positions of exons. The first exon contains translation start site, which is marked by an arrow from the box. 8 . The mouse-human (top) and mouse-rat (bottom) ECR analyses of the Cyp11 b2 promoter region obtained from the ECR Browser, which are presented in percent identity plots (pip-plots). The height of the colored areas corresponds to the percent identity of alignment (50 - 100 %), whereas the length corresponds to the length of alignments. Color indicates the nature of the ECRs (see legend).

between the two species. Previously, it was shown in Tg mice that 6.4 kbp of the

5' flanking promoter region of the mouse Cyp21 gene is sufficient to direct

75 expression of a reporter construct exclusively in the adrenal cortex • This

18 suggests that since the CYP21 promoter contains adrenocortical-specific locus control regions, such a ,region may also lie inside of the CYP 1182 promoter,

directing its ZG-specific adrenocortica~ expression. Targeting ZG utilizing the

CYP1182 gene has been attempted previously a bacterial artificial chromosome

(BAC) containing the entire CYP1182 gene or a large segment (>5 kbp) of the

76 gene promoter • However, the previous studies were not successful, possibly

due to the large amount of DNA that was utilized. Our analysis of the

evolutionarily conserved regions (ECR) between mouse and human CYP1182

promoters revealed homology within the -1 to -362 bp region, while greater

homology between the two rodents continued from -1 to -891 bp before reaching

a 104 bp gap and repetitive sequences (Fig. 5}. Therefore, Tg mice were utilized

in the specific aim of my project directed toward a better understanding of

CYP1182 gene regulation and the development of targeting strategies for the

mouse ZG. Our in vivo study is, in part, based on the 5' flanking cis-regulatory

regions of the CYP1182 promoter that have been identified through decades of

in vitro analyses. We presume that these key elements on the promoter region

should be important to physiologic regulation and ZG expression in Tg mice. We

believe that the adrenal ZG-specific expression of CYP1182 is -controlled· by the

promoter, which leads to its tightly regulated adrenocortical expression. This

study is of potential clinical importance, as it may become the first in vivo model

that advances our understanding of CYP1182 gene regulation.

19 B. Statement of the problem: A paucity of adrenal cell lines

Explorations into the physiology of the adrenal cortex have been hampered, in part, by the lack of available adrenal tissue ·samples and cell cultures. Primary adrenocortical cultures and adrenal cell lines allow critical in vitro analyses that could lead to- in vivo studies. Primary cultures are useful, as they are sufficient for transient in vitro analyses; however, handling and management of primary cultures becomes progressively problematic. Concerns regarding ·cell growth, response to agonists, and maintenance of steroidogenic capacity in primary adrenocortical cultures become more apparent with time. The ability of adrenal primary cultures to produce steroids or to respond to physiologic agonists, such as Ang II, ACTH, and K+, reduces over time. Expression of the enzymes involved in steroid hormone biosynthesis is reduced for most primary cultures. As a result,­ the steroids synthesized and released by primary cultures no longer resemble in vivo occurrences. Importantly, due to the above, there is a constant requirement for freshly acquired adrenal tissue for laboratories relying on primary cultures.

Collectively, the limitations of primary cultures have forced many laboratories to develop adrenocortical cell lines or use currently available cell lines.

Cell lines continuously allow provision of large numbers of viable cells without the need for animal sacrifice or acquisition of human tissue. Considerable progress in defining the basic molecular mechanisms involved in adrenocortical function has been made through the use of cell lines. Adrenocortical cell lines investigated thus far were generated from different sources, including cell

20 77 79 0O 84 · suspensions from adrenal tumors, • established _cell lines from tumors · ,

isolated immortalized cells following oncogenesis and cells harvested from Tg­

animals85-88. Consequently, the difficulties associated· with development and

characterization of viable cell lines should not be overlooked. As with primary

cultures, the ability of cell lines to produce steroids or to respond to- agonists can

change over time; therefore, steroid synthesis and cellular responses to agonists

must be monitered on a regular basis. The expression of the enzymes involved in

steroid hormone biosynthesis can also change under culture conditions. Due to

such stringent criterion, it is difficult to develop and characterize viable cell lines

to study the adrenal. Hence, the rarity in available human adrenocortical cell lines

to study the physiology and pathophysiology of the human adrenal cortex is re~I.

Herein, we attempted the development and characterization of a human

adrenocortical cell line. One aim has been proposed to determine the CYP11 B2

promoter regions that are required for its tissue-specific expression, as well as

repression within the adrenocortical zones. These studies should broaden our

knowledge of adrenocortical zonation under physiological conditions and provide

insights into human adrenal diseases.

21 1) Specific aim II

An appropriate, well characterized human adrenocortical cell model system· would be critical for studies focused towards defining the molecular mechanisms that lead to zonation of the adrenal ·cortex. Development and utilization of a human adrenocortical cell model is sought to further our understanding of ·the adrenocortical function.

Hypothesis: Adrenocortical carcinomas are an appropriate source of viable cell lines.

a. We developed several monoclonal populations of cells from an

adrenocortical carcinoma. Among the 47 different clones developed,

human adrenocortical carcinoma clone 15 (HAC15) expressed ·all of the

genes involved in the adrenocortical pathway, including StAR, CYP11 A 1,

CYP17, CYP21, HSD3B2, CYP11B1, CYP1182, and SU_LT2A1. Upon

stimulation with Ang· 11 and K+, an increase in aldosterone levels, as well

as the expression level.s for all of the enzymes involved in its production

was obsetved. ACTH caused a modest increase in cortisol and DHEA,

along with increased expression of all of the enzymes in their respective

biosynthetic pathways. HAC15 emerged as an appropriate in vitro model

to study the regulation of adrenocortical function.

22 2) Brief literature review and rationale: A paucity of adrenal cell lines

The historical methods of developing cell lines and maintaining cell cultures. Discovered in the early 20th century, the concept of utilizing

89 immortalized cell lines is quite complex • Most cell lines are grown and maintained under a tightly controlled environment, with optimal temperature and gas mixture. Culture conditions vary widely for each cell line, and slight variations in conditions could result in different phenotypes being expressed. One of the key factors in cell culture systems is growth media, as most cell lines require a

9 specific combination of components that make up growth media ~. Growth media vary in pH, mineral content, amount of growth factors, and presence of a variety of nutrients. Improper growth media could lead to suboptimal cell growth and reduction in efficiency of in vitro analyses. The most important constituent to consider before beginning in vitro analyses is the origin and development of a cell line. There are several innovative techniques of developing cell lines; proper procedures need to be followed in order to complete the characterization and development process.

Oncogenes such as Kirsten murine scarcoma virus (KMSV) have proven

91 useful in establishing virally infected immortalized cell lines • Following infection of a host cell, KMSV releases its single-stranded RNA genome into the host cell.

Once inside, such an oncogene is reverse-transcribed into double-stranded (pro­ viral) DNA, which then becomes covalently integrated into the host cellular DNA.

23 The proteins encoded by oncogenes are capable of transforming cells by stimulating- uncontrolled mitosis and hence providing a permanently growing population of cetls. Auersperg et al. developed rat adrenal cell lines by infection with KMSV. Primary cultures of rat adrenal cells in early passage were infected

92 94 with virus and allowed to overgrow the normal cell population - • Ragazzon et al. recently established mouse adrenocortical tumor cell lines using similar genetically targeted oncogenesis. Two cell lines, namely ATC1 and ATC7-L,

95 were developed and exhibit a ZF-specific phenotype •

Genetically targeted tumorigenesis in. Tg mice to establish adrenal cell

87 88 96 98 lines has been utilized by several investigators • • - • Mellon et al. used a human CYP11 A promoter to target expression of Simian Virus 40 T-antigen

(SV40) as a transgene in mice, which resulted in adrenal tumors. Moreover,

87 96 these generated adrenal tumors were used to establish cell lines • •

Correspondingly, mouse inhibin a-subunit promoter has ·also been utilized to drive SV4O expression in mice to generate adrenal tumors. Several adrenocortical cell lines were also generated from mice bearing a trarisgene for a

88 temperature-sensitive form of SV40 • At permissive temperatures, the protein encoded by the SV40 gene is activated, and the host cells are transformed. This process is reversible once the optimal temperature is shifted. However, cell lines developed from oncogenesis and -tumorigenesis have, thus .far, .yielded suboptimal cell lines with limited steroidogenic responses and modest enzymatic stimulation.

24 Consequently, naturally occurring adrenocortical tumors are the best source for the development of viable adrenal cell lines. Adrenocortical tumors provide a large number of viable steroidogenic populations of rapidly dividing. cells. A human adrenocortical ·cell line, NCI-H295, was established from a woman diagnosed with an adrenocorticat carcinoma84; the excised tumor was 14 x 13 x 11 cm and was used to establish this cell line. Tumor tissue was finely minced and the resulting population of cells has been maintained in various growth media.

Human adrenocortical cell lines have provided a wealth of knowledge towards understanding the mech_anisms that control . adrenal

_steroidogenesis. To date, of the several cell lines that have been developed, the human adrenocortical cell line, NCI-H295, and its strains have proven to be the most valuable. Initially, gas chromatography/mass spectrometry (GC/MS) and radioimmunoassays were utilized to identify steroids in the NCI-H295; 30 different steroids were discovered. Based on secreted steroids, these cells appear to contain all of the adrenocortical enzymes. After several analyses, NCI­

H295 adrenocortical cells express all of _the enzymes participating in normal human adrenal steroidogenesis, including CYP11A, HSD3B2, CYP11 B1, CYP21,

99 1 1 ·cYP17, CYP11 B2, and HSD3B2 - ~ • The NCI-H295 cell line is presently available from the American Type Culture Collection as ATCC CRL-10296 and wiH grow in suspension.

25 A population of cells, namely H295R, was screened and developed from

102 the parental NCI-H295 • In comparison to the p·arent NCI-H295 cell line, H295R cells grow as an adherent monolayer, and population doubling time is reduced from five to two days. In vivo Ang II acts on the adrenal ZG cells to increase production of aidosterone through type 1 Ang II (AT1)- receptors. Studies utilizing

H295R with radiolabeld Ang 11 in the presence of antagonists to the AT1 and AT2

· receptors established that the H295R cells utilize AT1 receptors almost

103 105 exclusively in aldosterone biosynthesis - • Another major physiologic regulator of adrenal aldosterone production is extracellular K+. Experiments utilizing

2 H295R cells have shown that K+ causes an increase in intracellular Ca + level leading to aldosterone biosynthesis and secretion. The H295R cells are currently being used as a model to better define the mechanisms of K+ regulation of

106 111 adrenal steroid production - . ACTH is the primary hormonal regulator of adrenal cortisol production. The H295R cell line is only mildly responsive to

ACTH, and most other strains and subpopulations of NCI-H295 are completely

82 unresponsive . H295R cells treated with ACTH respond acutely with an increase in adrenocortical steroid biosynthesis though the cell line lacks long-term

83 responsiveness .

These cells are useful in defining the role of enzymes controlling adrenocortical steroidogenesis. mRNA transcripts encoding the StAR gene, as well as the five forms of cytoch~ome P450 known to be involved in normal adrenal steroidogenesis (CYP11A1, CYP17, CYP21, CYP1182, and CYP1181),

112 116 are detectable in the H295R cells • • Experiments using the 5' flanking DNA

26 regions from CYP1181, .CYP1182, CYP17, and HSD382 have been performed .. 1 126 using these cells and several key transcription factors have been identified n• •

Taking adrenocortical steroidogenesis into consideration, the H295R cell line produces ·an steroid hormones under basal conditions, as well ~s upon

105 127 stimulation by Ang II, K+, and ACTH • • These cells ·have the ability to produce steroids that originate from all three zones of the adrenal cortex, including aldosterone (ZG), cortisol (ZF), DHEA (ZR), and DHEA-S (ZR). Hence, the H295R cell line is a potential model for studying steroid hormone

61 128 135 biosynthesis in all adrenocortical zones • • •

Subsequently, another strain of cells has been developed from the original

NCI-H295, designated H295A. Unlike the NCI-H295 cell line that grows in

1 suspension, H295A cells have been described to grow as a· monolayer8 • The method of isolation of the H295A strain was similar to that of the H295R, relying on the removal of non-attached cells with successive media changes. H295A cells lack Ang II, K+, and ACTH-stimulated steroidogenic responses and produce significantly less amount of steroids than the H295R cells. The AT1 receptor is

136 also highly expressed _in the H295R strain compared to H295A • Comparing the steroidogenic response to agonist and basal steroidogenesis, the H295R strain seems to be superior to the H295A strain.

The original NCI-H295 and its strains are useful models in defining the mechanisms controlling adrenocortical steroidogenesis, and they will continue to provide valuable information regarding the regulation of genes encoding steroidogenic enzymes. The regulation of genes in the steroidogenic pathway

27 has been demonstrated by a number of transcription factors identified in NCI­

H295 and H295R119-12a, 136-141 _

Another human adrenal carcinoma-derived cell line, SW13, has been developed. Although adrenal in origin, SW13 cells produce no steroids; perhaps this cell line was derived from non-steroidogenic cells of the original carcinoma 142. Due to the lack of steroidogenesis, its usefulness as an adrenocortical model system is none. These cells are also available from the

American Type Culture Collection (CCL-105).

Rodent and bovine adrenocortical cell lines have also contributed to our understanding of the adrenal gland. Its is presumed that following exposure to an atomic blast, an adult LAF1 (C57L x A/HeJ) mouse developed an adrenal tumor143. Sato and colleagues captured the transplantable tumor and allowed it to grow in vitro and developed the Y1 mouse adrenocortical cell line 144. Although the major steroid pathways present in Y1 cells are unlike the normal physiologic mous~ adrenocortical steroid pathways, a wealth of information has been gained through the utilization of these cells. Y1 cells have proved to be very useful in assessing the roles of cAMP, protein kinase A, adenylyl cyclases, ACTH, and

ACTH receptor in the adrenal cortex145-151 . Y1 cells do not respond to Ang II, although transgene expression of the Ang 11 receptor has been described in the

Y1 cells 152. Other mouse and rat adrenocortical cell lines have been developed through various aforementioned techn·iques; however, due to the lack of

28 hormonal responsiveness and - in_significant steroidogenesis, they remain unimportant.

Due to the relative ease in obtaining fresh bovine adrenals, the need for development - of bovine adrenal cell lines has been limited. Hornsby and

53 colleagues utilized SV40 to create a series of clonal bovine adrenal cell lines 1 •

The addition of SV40 to the cells greatly enhanced the cell culture lifespan of the cells; however, c·ellular expression of adrenocortical enzymes was found to

154 require specialized growth conditions • Consequently, this area of research remains largely unexplored.

The aforementioned human and rodent cell lines have allowed a multitude of studies that has broadened our understanding of normal adrenocortical endocrine function, as well as the properties of adrenocortical tumors. However, most of the cell lines have several limitations. The mechanisms involving hormonal regulation of adrenocortical cell growth, morphology, arid steroidogenesis remains a topic of active research; therefore, additional viable adrenocortical cell lines are sought.

29 n. MANUSCRIPTS

A. Peer Reviewed Articles

1) Gene targeting to the mouse adrenal zona glomerulosa

1 1 2 Jeniel Parmar , Anthony M. Cohen , Celso E. Gomez-Sanchez , William E. Rainey1 and Tsugio Seki1

1 Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912 2Division of Endocrinology, G. V. Montgomery VA Medical Centerand University of Mississippi Medical Center, Jackson, MS 3911 O

Abbreviated Title: Adrenal glomerulosa targeting

Key words: Cyp11 b2, adrenal cortex, zona glomerulosa, in vivo targeting

Name/Address for Correspondence: Jeniel Parmar Department of Physio.logy Medical College of Georgia Augusta, Georgia 30912

Telephone: 1-706-721-8363 Fax: 1-706-721-8360( •E-mail: [email protected]

30 Abstract

Adrenal zona glomerulosa (ZG) cells synthesize and secrete the majority of

circulating aldosterone, which is one of the key players of the· renin-angiotensin­

aldosterone-system (RAAS). Aldosterone plays a vital role in maintaining sodium

homeostasis, and its secretion is regulated, in large part, at the transcription level

of the aldosterone synthase (Gyp11 b2) gene in ZG cells. Although several

adrenocortical cell lines are available to study the mechanism regulating adrenal

gene transcription, lack of in vivo targeting strategies has made it difficult to apply

the in vitro findings to animal models. In this study, we isolated a 985 bp

Cyp11 b2 promoter fragment and successfully targeted adrenal ZG cells in vivo

using a reporter transgenic mouse model. In this Tg (Cyp11 b2-lacZ) mouse, the

lacZ reporter gene was detected only in the ZG cells and RAAS activation by low

sodium diet significantly upregulated ZG expression of lacZ. Moreover, the

transgene expression was not detectable in any of the 15 non-adrenal organs

assessed using quantitative PCR, indicating that this fragment exclusively targets

. adrenal ZG cells. This regulatory fragment will be an invaluable tool to study in

vivo the functional role of genes believed to play a pivotal role in adrenal

physiology and diseases.

31 Introduction

Aldosterone · is ·the principal mineralocorticoid synthesized in the zona glomerulosa (ZG) of the adrenal cortex. In humans and mice, aldosterone functions primari_ly to preserve cardiovascular homeostasis by regulating fluid

155 156 and electrolyte balance • . The principal glucocorticoid-cortisol in humans and corticosterone in mice-is synthesized in the zona fasciculata (ZF) of the

8 47 adrenal cortex • • The Cyp11 b2 and Cyp11 b1 genes share a high level of sequence similarity and the enzymes they encode, aldosterone synthase and

1113-hydroxylase, respectively, also possess high level of amino acid sequence

54 157 160 similarity • - • Despite these similarities, Cyp11 b2 is solely responsible for the

10 final step conversion of 11-deoxycorticosterone (DOC) to aldosterone , while

Cyp11 b1 converts DOC and 11-deoxycortisol to corticosterone (mice) and

56 cortisol (humans) respectively .

Studies utilizing transgenic mice have demonstrated that regulatory elements within the gene or in proximity of the gene can control tissue- and cell-specific

161 171 expression of certain genes - • Several genetically alte'red mouse studies have demonstrated that adrenal cortex targeted transgene expression is possible

163 172 177 through steroidogenic or non-steroidogenic gene promoters • - • Although the adrenal cortex has been targeted previously, these transgenic studies do not permit further adrenal cortex specific applications as most of these transgenic constructs have extra-adrenal targets. The exception is 21-hydroxylase driven

32 adrenocortical targeting by Morley et al (1996), however, this transgenic construct exhibited scattered-inconsistent targeting throughout the adrenal cortex and hence no further analyses have been conducted using this targeting

75 strategy • Attempts to develop a ZG-specific targeting strategy using a 5 kb promoter of mouse Cyp-11_ b2 gene have also been conducted, however this

177 approach essentially failed •

Herein, we have developed a new transgenic mouse mo~el using a 985 bp_

Cyp11 b2 promoter that specifically targets ZG. This DNA region was selected based on evolutionarily conserved region (ECR) analysis of the mouse, rat and human CYP11 B2 promoter sequences, which allowed us to reduce the length of the promoter from the previous study that attempted Cyp11b2 driven ZG targeting. We also utilized a lacZ reporter construct with enhanced stability in order to maximize· sensitivity of the -reporter gene expression in the mouse genome. Our results demonstrated that 985 bp Cyp11 b2 promoter-lacZ

(Cyp11 b2-lacZ) construct caused reporter gene expression solely in a ZG­ specific manner in transgenic mice. Furthermore, as with the endogenous

Cyp11 b2 gene, the expression of reporter lacZ was under the control of renin­ angiotensin-aldosterone system (RAAS).

33 Materials and Methods

Generation of transgenic mice

All animal procedures carried out in this study were reviewed and approved by

the Institutional Animal Care and Use Committee (IACUC) in the Medical College

of Georgia (MCG). All animals were housed in specific pathogen free (SPF)

facility.

An alignment of human, mouse, and rat aldosterone synthase (CYP11 B2)

genomic DNA sequences was obtained from the ECR Browser

178 . (http://ecrbrowser.dcode.org) • Based on the alignment analysis, a 985 bp

mouse Cyp11 b2 regulatory fragment was amplified from the RP23-323C12

bacterial artificial chromosome (Children's Hospital Oakland Research Institute)

using PCR. In order to generate transgenic (Tg} construct, the amplified fragment

was ligated to the SIB reporter cassette, which contains the SV40 splicing

donor/acceptor signals (SD/SA}, internal ribosomal entry sequence (I RES),

179 bacterial lacZ coding sequence,_ and SV40 late poly A signal • The Tg construct

was injected into B6C3 fertilized eggs by the MCG Mouse Embryonic Stem Cell

and Transgenesis Core Laboratories. Screening of founder lines were performed

by following the published protocol and the Tg lines were maintained as

hemizygous for transgene using C57BU6 mice (Harlan Laboratories) as

179 bleeders • Out of the seven Tg(Cyp11 b2-lacZ) founders that transmitted

34 transgene to the F1 generation, lines 5 and 6 that be·came available first were expanded for detailed analyses presented in this report. Later, additional lines 7 and 16 were analyzed for their lacZ reporter expression and found it essentially identical to the lines 5 and 6. None of the lines showed any detectable ectopic lacZ expression.

Of note, following the convention of the adrenal research community, mouse aldosterone synthase gene is designated as Cyp 11 b2 with italicization and its protein product, .aldosterone synthase, is designated as Cyp11 b2 without italicization.

Diet regimen and tissue sample collection

Only male mice were used in this study. All mice were separated from mothers at weaning age (3 weeks) and fed either normal sodium (NS, 0.3 % NaCl) or low sodium (LS, 0.03 % NaCl)- diets (Harlan Laboratories) ad libitum with unrestricted access to drinking water for 7 days. At the end of diet treatment, mice were instantly decapitated using a guillotine and trunk blood was collected into EDTA­ coated tubes for plasma aldosterone assay, followed by collection of sixteen major organs for immunohistochemical and molecular biological assays. These organs include adrenal glands, thoracic aorta, brain, eye, fat (visceral fat adjacent to spleen), heart, large intestine, small intestine, kidney, liver, lung, pancreas, skeletal muscle, spleen, testis, and thymus.

35 Plasma aldosterone measurement

Plasma aldosterone levels in mice were determined using the Diagnostic

180 Systems Laboratories RIA kit as previously described • Plasmas from mice fed­

NS diet (NS mice) were diluted 1 : 2 or 1 : 3 with charcoal stripped mouse plasma (Bioreclamation)- to obtain an appropriate volume for assay. Plasmas from LS mice were diluted 1 : 20 with the same stripped plasma so that aldosterone concentrations fall within the kit's detection range. Samples for the standard curve were prepared by serially diluting the supplied control aldosterone with the same plasma.

Total RNA and cDNA preparation

After the diet treatment, tissues samples were homogenized using the FastPrep homogenizer (Thermo Fisher Scientific}, and total RNAs were extracted using the

RNeasy Mini plus kit (QIAGEN). Two µg each of total RNAs was reverse­

83 transcribed using the High Capacity cDNA RT kit (Applied Biosystems) ..

Absolute quantif-ication of Cyp11 b2 transcript in male C57BU6 organs

Due to significant sequence homology between Cyp11 b2 and closely related steroid 11 ~-hydroxylase (Cyp11 b-1) genes, determining transcript levels of these two genes independently was challenging. In fact, one of the Applied Biosystems

Pre-developed TaqMan Gene Expression Assays for Cyp11b2, Mm0·1200453- g1, showed cross reactivity to Cyp11 b1 cDNA (data not shown}. Therefore, a

36 new Taqman assay for Cyp11 b2 was established. This assay showed a linear detection range over 7 logs of Cyp11 b2 cDNA inputs with a minimal cross­ reactivity to Cyp11 b1 cDNA (Supplemental Fig. 11 ). Detailed information and protocols for the Cyp11 b2 Taqman assay, real-time quantitative PCR (qPCR), nor~alization to 18S rRNA quantity, and data exclusion criteria are described in the Supplemental Materials and Methods.

Whole mount X-gal staining

Targeting ability of the Cyp11 b2 promoter fragment was visualized by X-gal staining. On application of X-gal staining solution, the lacZ gene product, 13- galactosidase, hydrolyzes X-gal to produce an insoluble blue dye. Hence, blue staining in cells indicates active transcription from the promoter. Whole mount

162 organ X-gal staining was performed as previously described . Harvested organs were sectioned into 1-2 mm thickness and stained for at least 8 h at 37

°C with constant rocking. Organ samples from wild type littermates were used as a negative control. The stained organs were photographed and then fixed with formalin overnight, followed by paraffin embedding and sectioning.

Histology and Cyp11 b2 immunohistochemistry

In order to localize Cyp11 b2 protein in adrenal sections, a novel polyclonal anti­

"rabbit Cyp11 b2 (mouse} antibody was developed by immunizing rabbit with human CYP1182 peptide. The VECTASTAIN Elite ABC Kit (Vector Laboratories)

37 was utilized to stain mouse tissue sections. First, antigen retrieval was performed by submerging the slides in sodium citrate buffer (2· mM citric acid and 9 mM trisodium citrate dehydrate, pH 6.0) and heating the container using a 1450 W microwave at 30 % power level for 12 minutes. The sections were then blocked with 2.5 % horse serum, followed by· application of the primary antibody, which was diluted 1 : 75 with PBS containing 0.5 % BSA. After the secondary antibody application, localization of Cyp11 b2 protein was visualized by application of 3.3'­ diaminobenzidine (DAB), and the sections were counterstained with nuclear fast red (NFR) or methyl green (Vector Laboratories). A negative control with no primary antibody treatment showed no detectable background staining in adrenal sections.

Relative quantification of transgene

In order to determine the transgene expression level in Tg_ mice, a transgene specific Taqman assay with a sense primer (5'-aaaaggctgtgaactgaagacaga), anti-sense primer (5'-tccgtacaggccagttaactttc), and probe (5'-ccccctcgaggaac) was developed using the Primer Express 3.0 program (Applied Biosystems). This. assay was designed to encompass the exon boundary in SD/SA sequence so that the Tg construct in genome will not be amplified. The following PCR program were used for qPCR with the ABI 7500 Fast Real-Time PCR System (Applied

Biosystems): the initial denature at 50 °C for 2 min and 95 ~C for 10 min, followed by 40 cycle amplification at 95 °C for 15 s, 50 °C for 1 min, and 72 °C for 1 min.

Using the relative qPCR method with standard curve, the results were -first

38 normalized to 18S rRNA, and then fold changes relative to average NS adrenal value were calculated.

Statistical analysis

Statistical comparisons were assessed between the NS and LS animals by

utilizing non-parametric unpaired Mann Whitney U-test. Significance was

accepted at the 0.05 level of probability. Data are expressed as mean± SE.

39,. Results

Since ECRs in promoter sequences are most likely to possess gene regulatory

178 functions , the essential regulatory region for the mouse Cyp11 b2 gene (Fig:.

6A) was predicted using mouse, rat, and human CYP11 B2 genomic sequences.

Counting from the translation start site, sequence homology between mouse and

human CYP11 B2 promoters was observed in -1 to -362 bp region, while higher

homology between the two rodents continued from -1 to -891 bp before reaching

a 104 bp gap and repetitive sequences (Fig .. 6B). Utilizing the endogenous Xbal

site at -989 bp position, we isolated a 985 _bp mouse Cyp11b2 regulatory

fragment covering- -989 to -5 bp sequence and then connected to the lacZ

reporter cassette (Fig. 6C}. The construct was injected to fertilized mouse eggs

and several Tg(Cyp11 b2-lacZ) mouse lines were established.

In advance characterizing this novel Tg mouse model, we first determined

· endogenous Cyp11 b2 gene expression patterns at RNA and protein levels using

male C57BU6 mice. Since LS diet significantly induced the Cyp11 b2 gene and

hence aldosterone production (Table I) via activation of renin-angiotensin­

system (HAAS}, organs from the mice fed normal sodium (NS) and low sodium

(LS) diets were collected and their Cyp11 b2 transcript contents were quantified.

In NS mice, the highest expression was observed in adrenal glands (3.35 x 106 ±

1. 71 x 106 copies/µg 18S rRNA}, which showed 260 times higher level than the

4 3 average of the other 15 organs (1.29 x 10 ± 3.49 x 10 ), indicating that adrenal

40 glands are the major organ for Cyp11 b2 expression in mice (Fig. 7 A). In LS mice,

9 8 the highest expression Was also obseived in adrenals (2.50 x 10 ± 8,.04 x 10 ), which was 42,744 times higher than the average of the 15 organs (5.83 x 104 ±

4 2.00· x 10 ), again indicating that the Cyp11 b2 is almost exclusively expressed in the adrenals (Fig. 7B). Compared to NS diet, LS diet induced 745 fold higher

Cyp11 b2 transcripts in adrenal glands· (p < 0.01 ). In addition·, thoracic aorta (20-­ fold, 1.52 x 104 ± 7.91 x 103 in NS vs 3.11 x 105 ± 2.66 x 105 in LS}, thymus {3.9 fold, 1.53 x 104 ± 3. 73 x 103 vs 5.98 x 104 ± 1.35 x 104), and kidney (2.9 fold, 4.01

3 2 4 3 x 10 ± 6.09 x 10 vs 1.16 x 10 ± 2.23 x 10 ) showed statistically significant

Cyp11 b2 induction by LS diet (p < 0.05). It is noteworthy that, although the amount of Cyp11 b2 transcript in LS aorta is the second highest among the 16 LS organs, it is about 1O times less than that of NS adrenal glands. These results suggest that our 985 bp Cyp11 b2 promoter fragment should target primarily the adrenal glands~

Furthermore, immunohistochemical staining of C57BU6 adrenal glands showed zona glomerulosa (ZG) specific localization of Cyp11 b2 (Fig. 8). As it has been reported 181 one week of LS diet stimulated ZG layer prqliferation/expansion (Fig.

8A and 8D) and significant accumulation of Cyp11 b2 protein in the ZG layer (Fig.

88 and BE). These qPCR and_ immunohistochemical analyses led us to characterize our Tg(Cyp11 b2-lacZ) mice by first investigating transgene expression in adrenal glands under LS diet.

41 Intriguingly, whole mount X-gal staining of Tg(Cyp11 b2-lacZ) adrenals revealed that, in all Tg lines 5, 6, 7, and 16, th~ reporter lacZ gene was highly expressed· in the outer layer of adrenal glands under LS diet (Fig .. 9). Littermate wild type controls showed little background staining after 8 h staining-, indicating that this staining was specifically induced by the transgene. The Tg mice fed NS diet also showed _very faint X-gal staining, which was hardly detectable under dissection microscopy (data not shown). Tg mice under NS and LS diets produced plasma aldosterone at comparable levels to the wild type C57BU6 mice {Table II), indicating that these Tg adrenals are functionally normal.

The reporter lacZ localization in adrenal glands was further characterized by

Cyp11 b2-immunohistochemistry and X-gal double staining. In consistent with the endogenous Cyp11 b2, under NS diet the blue staining was found only in the ZG layer and a few of these cells were expressing detectable levels of Cyp11 b2 protein (Fig. 1OA-C). Furthermore, LS diet dramatically upregulated the X-gal staining intensity and area in parallel to the endogenous Cyp11 b2 (Fig. 1OD-F).

Intriguingly, the X-gal staining was darker in more outer layer of ZG while the immunohistochemistry staining was more prominent in the inner layer of ZG. No detectable X-gal staining was observed in the adrenal capsule cells. These results indicated that 985 bp Cyp11 b2 promoter fragment was sufficient to target

ZG layer in adrenal glands and held a capability to respond RAAS activation.

42 Lastly, the upregulation of transgene in LS mice and its adrenal specificity was confirmed at RNA level by qPCR using Tg-lines 5 and 6-. ·Although transgene expression in some of the NS adrenals was not detectable due to a somewhat

low sensitivity of this assay, transgene was significantly induced by LS diet

(Table Ill}. Relative quantification of transgene showed 4,908 times higher transgene transcript in LS adrenal than in NS adrenals (p<0.01 }, indicating that the Cyp11 b2 promoter fragment is highly inducible by RAAS activation. There was no significant difference between Tg lines 5 and 6 in transgene expression

levels. Furthermore, the same qPCR assay revealed that none of the 15 non­

adrenal organs listed in the Fig. 7 showed detectable transgene expression. This

observation was confirmed by the X-gal staining, which demonstrated no specific staining in any of these major organs (data not shown). Therefore, we concluded that the 985 bp Cyp11 b2 promoter fragment contains essential regulatory elements for Cyp11 b2 gene transcription, it is sufficient to recapitulate the

endogenous promoter activity, and it drives transgene expression exclusively in

ZG cells in vivo.

41 . Discussion

Targeting adrenal glands in transgenic mice has been explored by utilizing

promoter fragments from various genes including the Cyp11 b2. Previously,

Morley et al have generate.d 7 independent Tg lines with a construct harboring

5.4 kb of mouse Cyp11 b2 5' flanking region and the lacZ reporter gene, which

177 resulted in no detectable adrenal lacZ expression in any of the lines • In

contrast, we used a much shorter (less than 1 kb) region from the same gene to

.successfully target the adrenal ZG layer. These results may suggest that the

additional 4.4 kb region contains further regulatory element(s) that have silenced

the lacZ reporter expression in .the 5.4 kb Tg mice. This additional 4.4 kb

sequence, however, does not show any homology to human corresponding

regions and in fact about three quarters of this sequence is filled with various

types of repetitive sequences. Alternatively, it is possible that the 5.4 kb Tg mice

may also have expressed the reporter gene if LS diet had been administered .

. The NS adrenals from the current Tg mice showed only very faint X-gal staining

and it could have been easily missed if we did not pay extra attention. We may

owe our success to our use of the LS diet that activated the RAAS .

. In addition, in the same report Morley et al have described that a 5.0 kb Cyp11 b1

regulatory fragment, which is not under the control of RAAS, failed to target

177 adrenal glands • In another report, however, the same group successfully

targeted adrenal cortex with a 6.4 kb regulatory fragment of the steroid 21-

44 hydroxylase gene (Cyp21a1}. In this Tg(Cyp21a1-lacZ) mouse model, the lacZ expression was observed exclusively in adrenal cortex, though it was in a

182 variegated pattern throughout adrenal cortex and was not layer specific • Other groups have also reported adrenal targeting by using regulatory regions from

173 174 various genes, •including aldo-keto reductase 187 (Akr1 b7) . • , steroidogenic

163 172 183 184 factor 1 (SF-1, Nr~a1) • • , and inhibin a (lnha) • These regulatory fragments, however, also lead to transgene expression in non-adrenal organs.

The current 985 bp Cyp11 b2 sequence is unique and distinct from these fragments since it induces transgene expression selectively and exclusively in the adrenal ZG layer.

Although both endogenous Cyp11 b2 protein and the lacZ reporter gene were induced in ZG layer in LS adrenals, there was a slight difference in their staining

185 186 patterns. Consistent with the previous reports • , both wild-type C57BU6 and

Tg mice showed ZG cells to proliferate from 3-4 layers to a 12-15 layered zone.

Within this thickened ZG layer, the X-gal staining was more prominent on the outer· (capsule} side, whereas the Cyp11 b2 immunostaining was more intense on the inner (medulla) side (Fig. 8 and 1O}. This difference may be explained by the current hypotheses that adrenocortical stem cells in capsule or sub-capsular layer proliferate, differentiate, and move inward to generate adrenal cortical

175 cells • Upon LS diet administration, the ZG cells start both inward proliferation and activation of Cyp11 b2 promoter. This will lead to Cyp11 b2 and Lacz protein production in the outer ZG layer in Tg adrenals, which will result in aldosterone

45 production and intense X-gal staining, respectively. Since the inward blood flow

in adrenai cortex transports aldosterone toward the inside, inner ZG celts will sense a high leveJ of aldosterone, which should results in down-regulation of

Cyp11 b2 promoter activity in these cells and hence weaker X-gal staining-. The

Cyp11 b2 protein, however, may have been stabilized under the LS diet condition

and thus have accumulated in the inner ZG layer.

In addition to adrenal glands, Cyp11 b2 expression has been described in non­

adrenat organs, including heart, brain and vasculature, under physiological

187 195 and/or pathological conditions - • In our study, we have dete~ted Cyp11b2 transcript in all organs tested (Fig. 7), whereas the levels were much lower in

non-adrenal organs. In addition, LS diet induced Cyp11 b2 upregulation in thoracic aorta, thymus, and kidney, while their fold changes were small. Although

these non-adrenal Cyp11 b2 expressions may have little impact on plasma

aldosterone levels, it may activate a local aldosterone-mineralocorticoid receptor

system, and hence it should not be overlooked. Nonetheless, our Tg mice

showed no detectable reporter gene expression at RNA and X-gal staining levels

in any of the non-adrenal tissues tested, which may be due to the limitations in

our assay sensitivities. The NS adrenal in Tg mice showed minimal transgene

expression at both qPCR and X-gal staining levels (Fig. 1O and Table Ill),

indicating that the sensitivities of our assays were not sufficient to detect non­

adrenal transgene expression, which should be much lower than that of NS

adrenals (Fig. 7). Therefore, it is still unclear whether the current Cyp11 b2

46 promoter fragment contains the essential regulator elements for non-adrenal transgene expression. Further analyses under various pathological conditions

may shed some light on this issue in• the future.

In this study, we identified the Cyp11 b2 regulator fragment that exclusively targets adrenal ZG cells and responds to RAAS activation. This regulatory

fragment can be used in a T g construct .to specifically activate or silence any

gene in ZG. cells in vivo. Nevertheless, this regulatory fragment will be an

invaluable tool for the advancement of research associated with adrenal

development, physiology, and pathology.

47 . Acknowledgement

We appreciate the work of the Medical' College of Georgia Mouse Embryonic

Stem CeJI and Transgenesis Core Laboratories and- the Electron Microscopy &

Histology Core Laboratory.

48 Table I: Plasma aldosterone concentrations in C57BU6,mice (average pg/ml± SE)-

NS (n=5} , LS (n=6} NS vs LS

587.6 ± 38.4 11,367.5-± 4,160.9

Table II: Plasma aldosterone concentrations in Tg mice (average pg/ml± SE)

NS (n=5} LS (n=5} NS vs LS

669.6 ± 167.5 5,688.4 ± 581.4 p<0.01

Table Ill: Relative Cyp11 b2-lacZ mRNA expression in adrenal glands (fold change against NS)

# of # of samples Tg mice in detection Fold change ± Fold change ± Diet line analyzed range SE Line 5 vs 6 SE NS vs LS

Line 5 6 4 (67%) 0.93 ± 0.39 NS p = 0.69 1.00 ± 0.24 Line 6 5 4 (80%) 1.07 ± 0.34 p < 0.001 3,103.26 ± Line 5 7 7 (100%} 736.20 4,908.27 LS p = 0.32 * 1,456.70 6,713.29 ± Line 6 7 7 (100%) 2,750.85 Figure legends:

Fig. 6: Mouse Cyp-11 b2 ECR analysis and Tg construct. A. Schematic representation of mouse Cyp11 b2 genomic structure. Boxes indicate positions of exons. The first exon contains translation start site, which is marked by an arrow from the box. B. The mouse-human (top) and mouse-rat (bottom) ECR analyses

.of the Cyp11 b2 promoter region obtained from the ECR Browser, which are presented in percent identity plots (pip-plots)'. The height of the colored areas corresponds to the percent identity of alignment (50 - 100 % }, whereas the length corresponds to the length of alignments. Color indicates the nature of the ECRs

(see legend). Note that the shaded area corresponds to the promoter fragment in panel C. C. Schematic representation of the Cyp11 b2-lacZ construct that is used to establish Tg(Cyp11 b2-lacZ) mouse lines.

Fig. 7: Cyp11 b2 transcript quantity in 16· major organs in male C57BU6 mice. A.

Cyp11 b2 transcript quantity in mice fed NS diet. Due to a significantly high copy number in adrenals, a graph without adrenal data is re-plotted in the inset. Sk. muscle: skeletal muscle; S. intestine: small intestine; L. intestine: large intestine.

B. Cyp11 b2 transcript quantity in mice fed NS (solid bar) and LS (open bar) diets.

Note that, to aid NS vs LS mouse comparison, the same NS data in A are shown in this graph but in logarithmic scale. Statistically significant difference between

NS and LS mice were seen in the 4 organs (**: p<0-.01 and *: p<0.05). Numbers

50 of mice ·tested for each organ is shown in parentheses. The inset shows fold changes of transcript quantity in LS mice compared to that of NS mi_ce.

Fig. 8: Cyp11 b2 localization in adrenal cortex. Adrenals from male C57BU6 mice fed NS (A-C) and LS (D-F) diets are immunohistochemically stained with a novel rabbit polyclonal antibody. Mouse adrenal sections show histologically distinct 3 cortical layers by hematoxylin and eosin (H&E) staining (A and D) and Cyp11 b2 immunohistochemistry (brown) revealed its localization in the ZG layer (Band E).

Note that LS adrenals show much thicker ZG layer .with significantly intense

Cyp11 b2 immunostaining. Negative control staining performed without the primary antibody show little background (C and F). lmmunohistochemical sections in panels B, C, E, and F are counter stained with methyl green. ZG: zona glomerulosa; ZF: zona fasciculata; ZR: zona reticularis. All panels are in the same magnification. Scale bar in F: 0.5mm.

Fig. 9: X-gal stained Tg adrenals under LS diet. A. Adrenals from Tg and wild type littermate control (wt) were sectioned in half and then X-gal stained. Tg adrenal shows dark X-gal staining at the outer cortex layer. B and C. Magnified views of panel A. Scale bar in A: 0.5mm.

Fig. 10: Colocalization of Cyp11 b2 and X-gal staining in Tg adrenal cortex.

Adrenals from male Tg mice fed NS (A-C) and LS (D..:F) diets were X-gal stained

(A and D), followed by immune-staining for Cyp11 b2 (8, C, E, and F). Sections are counterstained with nuclear fast red. Panels A, B, D, and E are in the same magnification. Panels C and F shows high power view of Band E, respectively.

51 Fig·. 11 (supplemental figure}: Amplification cutve. using· descending pattern of

Cyp11 b2 transcript number as depicted · (right). Cyp11 b2 and Cyp11 b1 primer/probe were tested against Cyp11 b2 transcripts to assess the cross reactivity against the two highly similar genes. Standard cutves were constructed using 9 samples containing varying known quantities of Cyp11b2 transcripts.

During the real-time PCR amplification, the cycle at which these samples attain a particular level of product-the crossing point-is noted and plotted against the starting concentration of standard. The crossing points of samples are then measured and the starting concentration of target mRNA is extrapolated from the standard cutve.

52 Fig. 6:

C ----ti .· so/SA~IR.E~la~ I Cyp11 b2~1acz constru¢f

53 Fig.

7:

54 Fig. 8:

N$

LS

55 Fig. 9:

56 Fig. 10:

57 Fig. 11:

Cyp11 b2 copy numbers and corresponding cycle of threshold (Ct) values

Cyp11b2 Average Ct transcript copy value number

108 13.03 • 107 17.17 • 106 20.21 • 105 23.90 • 104 27.64 I 2 3 • 5 8 T 8 910' 111213U1S1617 1819 2021 22231{25 262728 • 103 31 .24 • 102 35.29

30 1 36.92 :;; • 10 .Q 25 E Undetermined ::, • C 20 QI Cyp11b1 Average Ct 0 transcript copy >. 15 value I.) number ·10 108 35.68 5 +,:=:,~1::;:;:.,...,::,=:-,,=~~:,.-...-~~~ 107 Undetermined 0 ..p,...... _.,...:_.....,,__.....; __ ...... ,.....:..-.::...-"r-' ...... • 0 2 3 4 5 G 7 s 9 • 106 Undetermined Cypft b2 transcript copy number (log scale)

58 II. MANUSCRIPTS

A. Peer Reviewed Articles

2) Development of an ACTH Responsive Human Adrenocortical Carcinoma (HAC) Cell Line

Jeniel Parmar, Rebecca E. Key and William E. Rainey

Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912. -

Abbreviated Title: Development of a human adrenocortical cell line

Key terms: Adrenal, ACTH, angiotensin II, aldosterone, cortisol, model system

Name/Address for Correspondence: Jeniel Parmar Department of Physiology Medical College of Georgia Augusta, Georgia 30912

Telephone: 1-706-721-8363 Fax: 1-706-721-8360 E-mail: [email protected]

59 Abstract

Context: The molecular mechanisms regulating adrenal steroidogenesis continue to be defined. The only current human adrenocortical cell line is the

NCI-H295 and its substrains. One of the strains, the H295R, has retained the ability to respond to angiotensin 11 (Ang 11) however, it lacks ACTH responsiveness. An ACTH responsive human adrenocortical model would add significantly to studies directed at defining the molecular control of corticosteroid biosynthesis.

Objective: Develop a human adrenal cell line that retained both Ang II and

ACTH regulated corticosteroid production.·

Design:· Human adrenocortical carcinoma (HAC) cells were isolated from an adrenal tumor removed from a girl presenting with virilization and hypertension.

Clonal populations of cells were established and characterized. HAC cells were treated with ACTH, Ang II and forskolin, followed by examination of steroidogenic enzyme mRNA expression using quantitative real-time PCR and steroid production.

Results: HAC clone 15 (HAC15) cells responded to treatment with ACTH, Ang II, and forskolin, with increased cortisol and aldosterone production. ACTH, Ang II,

60 and forskolin also increased expression of mRNA, encoding all enzymes needed for cortisol and aldosterone biosynthesis; namely StAR, CYP11A, CYP17,

HSD3B2, CYP21, CYP11 B1, and CYP11 B2. In addition, the cells expressed mRNA for ACTH receptor (MC2R) and Ang II receptor (AT1 R). MC2R protein was also expressed in HAC 15 cells.

Conclusion: The current study describes the development and characterization of an ACTH and Ang II responsive human adrenal cell line. The HAC15 cell line should provide an important model system for · defining the molecular mechanisms regulating aldosterone and cortisol production.

61 Introduction

Defining the molecular mechanisms and cellular signaling pathways that regulate the production of adreriocortical steroids is often limited by the .availability of appropriate model systems. One method of modeling the adrenal is through the

196 use of cell lines that retain adrenocortical differentiated functions • The list of permanent cell lines established from human adrenal tumors. is remarkably short,

84 196 consisting of the NCI-H295 and its substrains • • The NGI-H295, H295R and

H295A retain several differentiated functions, which make them useful for

12 57 84 102 105 110 defining the cellular mechanisms regulating steroid production • • • , , ,

127 196 203 • - • However, the NCI-H295 cells have little or no ACTH .response. Thus, there is currently no human adrenal cell model that responds to a sustained

ACTH stimulation.

In this report, we describe the establishment of a permanent human adrenocortical carcinoma . (HAC) cell line. These cells continue· to secrete glucocorticoids -and mineralocorticoids. Most importantly, these cells have retained responsiveness to·both ACTH and angiotensin II (Ang II).

62 Materials and Methods

HAC Cell Culture Establishment

Primary cultures of HAC- cells were isolated following surgical removal of an

adrenocortical carcinoma from an 11 month old female with. hypertension and

hirsutism. The use of tumor tissue was approved by the Institutional Review

Board of the Medical College of Georgia (Augusta, GA). Primary carcinoma cells

were isolated after the tumor was minced into small pieces and incubated in

DME/F12 (lnvitrogen, Carlsbad, CA) containing 0·.1 % collagenase (Roche,

Indianapolis, IN}. Digestion and mechanical dispersion were carried out for 1.5 h

at 37°C, with a final digestion of the dispersed cells in medium containing 0.01 %

DNase I. Following isolation, cells were frozen in DME/F12 medium, 50 % Nu­

Serum (BD Biosciences, Franklin · Lakes, NJ), and 10 % dimethyl sulfoxide

(DMSO) (Sigma, St. Louis, MO). The frozen aliquots were later suspended in

_growth media consisting of DME/F12 medium supplemented with 10 % Cosmic

Calf Serum (CCS) (HyClone, Logan, UT) antibiotics and 1 % ITS Premix (BD)

and plated at cloning density. After three weeks, clones were isolated for

characterization. The most ACTH responsive steroidogenic clone, HAC clone 15

(HAC15), was used for this study. H295R cells, grown under similar conditions

were used for comparison studies. For experiments, cells were plated onto 12

dishes (400,000 cells per well) in growth medium. Cells were treated in low

63 serum medium (0.1 % CCS) with either Ang II (1 O nM) (Sigma), forskolin (1 O µM)

(Sigma), or ACTH (1 O nM} (Organon, Bedford, OH}.

Analysis of Steroids

Experimental media from HAC15 and H295R cells were analyzed for aldosterone and cortisol levels using specific immunoassays (Siemens Diagnostics,

Tarrytown, NY and Alpco Diagnostics, Salem, NH, respectively). Steroid data· was normalized to mg total cell protein.

Real-Time Reverse-Transcriptase Polymerase Chai'1 Reaction {qPCR)

Total cellular RNA was extracted using RNeasy Mini Kits (Qiagen, Valencia, CA).

Two µg of totai RNA was reverse transcribed using the High C_apacity cDNA,

Archive Kit (Applied Biosystems, Foster City, CA). cDNA of the specific mRNA encoding enzymes and ·receptors were examined using probe sets obtained from

TaqMan Gene Expression Assays (Applied Biosystems). Quantitative normalization of cDNA for each gene was performed using the expression of 18s rRNA.

64 Protein lmmunoblot Analysis

Cells were lysed and 40· µg of lysate was applied to polyacrylamide gel electrophoresis using 12 % bis-tris NuPage gels (lnvitrogen). Membranes were incubated overnight at 4°C, with antibody against MC2R (ACTH receptor},

(Chemicon International, Temecula, CA), at a 1:1000 dilution. Membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies

(Santa Cruz, Santa Cruz, CA). lmmunoreactive bands were visualized using the

ECL plus Western Detection System (GE Healthcare,. Piscataway, NJ) and a

GBOX image analyzer system (Syngene, Frederick, MD}.

Statistical Analysis

All experiments were repeated a minimum of three times, and within each experiment, variables were performed in triplicate. Statistical comparisons were analyzed using one-way ANOVA. Significance was accepted at the 0.05 level of probability.

65 Results

The effects of Ang II, ACTH, and forskolin on HAC15 cell production of aldosterone and cortisol were examined following treatment for 24 h. Treatment with ACTH and Ang II significantly increased HAC15 cortisol production by over

2-fold (Fig. 12A). HAC15 cells also increased aldosterone production by 3-fold in response to ACTH and 8-fold in response ·to Ang 11 (Fig. 12A)-. Bypassing the

ACTH receptor by treating with forskolin (10 mM) significantly increased both aldosterone production (2.5-fold) and cortisol production (3-fold) in HAC15 (Fig.

12A}.

Due to the ACTH response observed for HAC cells, we examined expression· of

MC2R mRNA and protein. Protein immunoblot analysis was used to examine

MC2R in cell lysates of H295R and HAC15 (Fig. 12B). MC2R expression was evident in the HAC15 resulting in an unambiguous band of approximately 38

· kDa. In contrast, MC2R expression was undetectable in the H295R cells. MC2R and AT1 R (type 1 Ang ll} mRNA levels were also compared between the HAC15 and H295R using qPCR. HAC15 mRNA for MC2R was elevated 9.25-fold as compared to the H295R cell line. AT1 R mRNA levels were similar in both cell_ lines (Fig. 12B}. ·

To further assess adrenal differentiated function in HAC15 cells, the expression of mRNA encoding enzymes involved in the production of aldosterone and

66 cortisol were examined (Fig. 13). Treatment with ACTH increased mRNA expression of all the enzymes needed for aldosterone and cortisol production, name~y StAR, CYP11A1, CYP17, CYP21, HSD3B2, CYP11B1·, and CYP11B2 mRNA as compared to untreated cells (Fig. 13}. Treatment with Ang. II and forskolin also caused an increase in mRNA expression of all the enzymes needed for aldosterone and cortisol production.

67 Discussion

We previously developed a human adrenocortical cell model, H295R, which

96 retains Ang U and forskolin responses 1 • These cells have been widely used to study steroid hormone production; however, they are not a good model for ACTH regulated steroidogenesis. Herein, we isolated a clonal pop·ulation of adrenocortical carcinoma cells that retained Ang 11, ACTH and forskolin responsiveness. These cells will provide an alternative to the H295 substrains and should act as a better model for ACTH response.

Adrenal cell lines have proven useful for defining basic mechanisms involved in adrenal function. There have been a number of rodent adrenal cell lines developed -using either adrenal tumors or transgenic mice with adrenal-targeted

196 oncogenes • The Y-1 _mouse adrenal cell line is the most widely used for

196 204 studying acute regulation of steroidogenic· enzymes • • However, the Y-1 cells have _lost the expression of 21 hydroxylase and therefore cannot produce cortisosteroids. Two other mouse adrenocortical cell lines, ATC1 and ATC7-L,

95 were developed using genetically targeted oncogenesis • Both cell lines retain a zona fasciculata phenotype, which is manifested at multiple levels, including· corticosterone secretion, _ACTH responsiveness, and expression . profiles of steroidogenic enzymes .. While each of these cell lines are useful for many adrenal research projects, mouse adrenal cell lines can not replicate the ability of

human adrenal cells to produce cortisol or adrenal androgens. Therefore, human

68 adrenal cell lines are needed for many studies directed at understanding the

human adrenal.

The NCI-H295 and its substrains are currently the only human adrenocortical cell

12 57 84 102 105 110 127 196 203 models available • • • • • • • - • Two strains of., H295 cells,

196 _designated H295R and H295A, have been selected to grow as a monolayer •

These cells. have been used to study steroidogenic enzyme expression and

steroid hormone production. In addition, the cells have been used to examine the

molecular mechanisms regulating transcription of CYP11 A 1, CYP11 B1,

12 57 198 203 CYP1182, CYP17, CYP21, and HSD3B2 • • - • The H295R cells are also

responsive to Ang 11 and potassium through the production of aldosterone and

57 105 the expression of CYP11 B2 • • Thus, thes·e cells have been widely used to

bett.er understand the molecular mechanisms regulating human adrenal cell

steroidogenesis.

The successful development of the original NCI-H295 cells from an

adrenocortical carcinoma led us to use a similar approach to develop the HAC15

cells. Our findings indicate that the HAC15 cells have similar steroidogenic

profiles as compared to the H295R. These findings include the production of all

the corticosteroids. Cortisol was produced at significantly higher levels than

aldosterone. However, this may be the due to low expression of CYP11 B2

(al_dosterone synthase), which requires Ang II treatment to increase . its

expression. The HAC15 cells also consistently produce significant amounts of

the adrenal androgen, dehydroepiandrostenedione (DHEA) (data not shown).

69 Further studies will be needed to determine if chronic hormonal treatment (Ang II or ACTH) can influence the HAC15 phenotype to be either glomerulosa, fascicuJata or reticularis like.

Similar to the NCI-H295 and its substrains, the HAC15 cells expressed mRNA encoding all enzymes needed for production of aldosterone, cortisol and DHEA.

All of these transcripts were responsive to both Ang II and ACTH. HAC15 cells were also responsive to extracellular potassium through an increase in aldosterone production and CYP11 B2 expression (data not shown). The major difference between the HAC15 and H295R appears to b~ their ACTH response.

MC2R is the well documented receptor to which ACTH binds and regulates

205 different cellular responses • While the H295R cells have a number of limitations, one of the most obvious is the lack of a sustained ACTH response.

The lack of responsiveness in H295R cells is not clearly understood. The cells have mRNA expression for MC2R, which is regulated by Ang II and cAMP

206 signaling systems • In addition, the H295R cells respond acutely to ACTH with

207 an activation of the ERK 1/2 phosphorylation • However, our direct comparison of· H295R and HAC15 cells demonstrated significantly higher_ levels of MC2R m RNA and protein expression in the HAC 15 cells. These data suggest that the differences between the HAC15 and H295R cells are at least, in part, due to differences in MC2R expression.

70 In summary, we have developed an adrenal cell line, HAC1~ that exhibits hormonal responses, steroidogenesis, and expression of steroid-metabolizing. enzymes. This cell line represents only the second human adrenocortical cell line

available. The ability of HAC15 cells to respond to Ang II, potassium and ACTH

make them the first adrenal cell line capable of responding to the three main

adrenocortical physiologic regulators.

Acknowledgements

We thank the expert editorial assistance of Ors. J. Ian Mason, Raymond J.

Rogers, and Ora Pescovitz.

71 · Figure Legends ·

Fig. 12: Panel A. Steroidogenic responsiveness of HAC15 and H295R cells to

agonist treatment. Cell lines were treated for 24 h with Ang II (10 nM), ACTH (1 0

nM}, or forskolin (10 µM). Cortisol and aldosterone concentrations were

determined by EIA and RIA, respectively. Values represent the mean ± SE of six

independent experiments. *** P < 0.001 for agonist-stimulated steroidogenesis

vs. basal. Panel B. HAC 15 cell expression of MC2R and AT1 R receptors. Protein

immunoblot analysis was used to examine expression of MC2R in cell lysates of

H295R and HAC15. 40 µg of protein was loaded per lane. Comparison of MC2R

and AT1 R mRNA levels in HAC15 to H295R was done following qPCR with the

H295R cells used as a calibrator. Values represent the mean ± SE of six

independent RNA samples. *** P < 0.001.

Fig. 13: HAC15 cell expression and regulation of genes encoding enzymes

involved in steroid hormone biosynthesis. HAC15 cells were treated with or

without Ang II (10 nM}, ACTH (10 nM), or forskolin (10 µM) for 24 h. mRNA level

for each enzyme was determined using qPGR. Values represent the mean ± SE

of six independent experiments. Nomenclature for the steroidogenic enzymes are

as follows: StAR: steroidogenic acute regulatory protein; CYP11 A 1: cholesterol

side-chain cleavage; CYP17: 17a-hydroxylase-17,20-lyase; HSD382: 3(3-

hydroxysteroid dehydrogenase type II; CYP21: 21-hydroxylase; CYP1181: 11 (3-

72 hydroxylase; CYP11 B2: aldosterone synthase. *** P < 0.001, ** P < 0.0,1, * P <

0.05.

73 Fig .. 1-2:

A 90 18

80 *** • Aldosterone 16 -C: "

O· 0 Basal Ang II ACTH Forskolin

B

12 -a: (kDa) LO H295 HAC1 0) 220 *** -MC2R 11 ~_,_ C\J I 120 10 JJ :i... 60 - AT1R Q) 3 > 9 JJ 0 40 z Q) 8 )> 0) C: a, MC2R 7 --0 ..c: C. (.) 6 (') :::r "'O m 0 5 :::::, (Q -<( 4 CD 0 z < a: 3 CD E -,; 2 I a: I\) C\J c.o (.) 01 ~ JJ 0 -74 H295R HAC15 Fig. 13: Cholesterol

- CYP11A1 liiiilStAH

tt * **

Basal Ang 11 ACTH Forskolin Pregnenolone 17a-Hydroxypregnenolone 40 2.5 .,..... HSD382 CYP17 *** -a -!/) -a 2.0 -!/) CCI 30 CCI .0 .O· Q) Q) > 1.5 ~ 0 -0 20 "O 0 g ::::.. 1.0 C\I ..... CD C") 0:: 0 10 >- en C.) 0.5 J:

0.0 Basal Ang II ACTH Forskolin Basal Ang 11· ACTH Forskolin Progesterone 17a-Hydroxyprogesterone 6..------, CYP21

g"C 3 N 2 Cl. >- C.) 1

0 Basal Ang II ACTH Forskolin 11-Deoxycorticosterone 11-Deoxycortisol

CYP11B1 cti CYP1182 -(/) l - 30321 CCI m .0- :j Q) 10 > ! 28 0 ~ -0 8 "C 0 :::. g 6 C\I 6 al· in 4 CL 4 >- CL 0 2 [) 2

0 0 Basal Ang II ACTH Forskolin Aldosterone Basal Ang 11 _ACTH Forskolin CortisoV 5 II. MANUSCRIPTS

Invited Review Article

Comparisons of adren·ocortical cell lines as in vitro test systems

Jeniel Parmar and William E. Rainey

Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912

Abbreviated Title: Adrenal glomerulosa targeting

Key words: Cyp11 b2, adrenal cortex, zona glomerulosa, in vivo targeting

Name/Address for Correspondence: Jeniel Parmar Department of Physiology Medical College of Georgia Augusta, Georgia 30912

Telephone: 1-706-721-8363 Fax: 1-706-721-8360 E-mail: [email protected]

76 I. BACKGROUND

The adrenal gland is a compound endocrine organ separated into two distinct developmentally unrelated tissues; an adrenal medulla and an adrenal cortex.

The adrenal medulla is the central core of the adrenal gland; the chromaffin cells of the medulla are the body's main source of the catecholamines. The cells comprising the adrenal cortex are derived from mesoderm of the dorsal coelomic wall and thus have common features to the steroidogenic cells within the gonads.

In 1866, Arnold first described the histology of the adrenal cortex and noted that it was divided into three concentric zones which he named the zona glomerulosa,

43 zona fasciculata, and zona reticularis • While this description was based on the histological organization, it is now accepted that these zones have functionally distinct roles in steroid hormone production. Namely, the glomerulosa synthesizes mineralocorticoids, the fasciculata produces glucocorticoids, and in the human, the zona reticularis produces C19 steroids, including DHEA and

\ DHEA-sulfate. Molecular mechanisms leading to zone specific expression of these steroids are yet to be defined. Each adrenocortical zone synthesizes its steroid products from the common substrate cholesterol. Steroidogenic cholesterol can arise from endogenous cholesterol stores, from serum derived lipoprotein or from de novo synthesis. Within the human adrenal cortex, steroid synthesis involves coordinated actions of five forms of cytochrome P450 and the enzyme 3~-hydroxysteroid dehydrogenase (Fig. 14}; these enzymes are

77 44 distributed between the mitochondria and the endoplasmic reticulum • It is the differential expression of these enzymes within the three adrenocortical zones that allows for the wide array of steroid hormones secreted by this gland.

Although. the enzymes that are differentially expressed between the zones have been identified, the molecular mechanisms causing zone-specific expression patterns are yet to be fully understood. The mechanism regulating production of steroids from each distin_ct zone is quite different and therefore, extremely complex. Angiotensin II (ANG 11) and potassium (K+) are the major regulators of steroidogenesis in the zona glomerulosa while, ACTH is the principal hormone regulating steroidogenesis in the fasciculata and reticularis (Fig. 15). In each case, the rate-limiting step in steroid hormone biosynthesis is the translocation of substrate cholesterol from the outer mitochondrial membrane to cholesterol side­ chain cleavage (CYP11 A), the first enzyme in the steroidogenic pathway, which is located inside the mitochondrion. In addition, regulatory mechanisms control the bioavailability of substrate cholesterol, the synthesis of the enzymes required for steroidogenesis, and the size and structural integrity of the gland.

Investigations into the complexity of the adrenal cortex have been hampered by the unavailability of adrenal tissue samples and cell cultures. This has made the development of viable in vitro cell models an attractive alternative to studies using whole animals. A number of in vitro systems have been investigated,

78 including;· cell suspensions from acutely dispersed tissue, primary cultures from

78 79 normal adrenal glands and adrenal tumors, • and established cell lines from

80 81 85 88 tumors • or from immortalized cells - • Primary cultures of adrenocortical cells have proven useful for examining mechanisms controlling the many aspects of adrenal physiology but the constant requirement for freshly acquired tissue and the difficul,ties associated with the isolation of c~lls has increased the demand for alternatives. Adrenal cell lines, which allow large numbers of functional• cells to be propagated without the need for animal sacrifice or acquisition of human tissue, have yielded considerable progress in defining basic mechanisms involved in adrenal function. Several issues become important for these adrenal model systems, including cell growth, response to agonists and maintenance of steroidogenic capacity. The ability of adrenal primary cultures and cell lines to produce steroids or to respond to ACTH, ANG U and K+ can change over time in culture. Therefore steroid synthesis and cellular responses to agonists must be monitored continuously. The expression of the enzymes involved in steroid hormone biosynthesis can also change under culture conditions. As a result, the steroids produced by cells in culture can be quite different from that seen in vivo. Thus, each of these criteria must also be considered when developing or evaluating cell lines_ to study the adrenal.

79 II. AVAILABLE RODENT ADRENOCORTICAL CELL LINES {see Table IV)

A. The Y1 Adrenal Cell Line

The Y1 adrenal cell_ line was derived from an adrenal tumor that developed in a

LAF1 (C57L x A/HeJ) mouse following exposure -to an atomic blast; a direct relationship of tumor development to the radiation is not clear 143. Gordon Sato and colleagues 144 adapted the transplantable tumor to grow in vitro by

. . alterna~ely propagating dispersed tumor cells as. mono'layer cultures and as tumors in mice. One of the clones developed from the mixed population of tumor cells was named Y1, and it was this clone that was deposited at the American

Type Culture Collection (ATCC CCL-79)80.

Major steroid pathways present in cultured Y1 cells are _shown in Figure 16. In contrast to the normal mouse adrenal gland that produces corticosterone as the major steroid product, Y1 cells in culture produce 20ct-hydroxy-~4-pregnen-3-one

(20a-dihydroxyprogesterone) and 11 (l,20a-dihydroxy-~4 -pregnen-3-one

(11 (l,20a-dihydroxyprogesterone}2°8· 209. This abnormal steroid profile relates to the deficiency in 21-hydroxylase (CYP21 )210 coupled with an increase in 20a­ hydroxysteroid dehydrogenase activity2°8.

The ability of ACTH to regulate Y1 cell steroid synthesis is similar to that seen for normal mouse adrenal cells and other agents that raise intracellular levels of 80 cAMP, which leads _to an induction of a number of genes that are supportive for steroid hormone biosynthesis. Examples include genes encoding the ACTH receptor145• 206, Cyp11 a211 -214, 11 f3-hydroxylase (Cyp11 b1 )214-216·, steroidogenic acute regulatory · protein "(StAR)213· 217· 218, adrenodoxin211 · 212, adrenodoxin reductase212 and· the HDL receptor (SR-Bl} 219. As opposed to the robust stimulation of steroid hormone production by ACTH, Y1 cells do not respond to

ANG II, although transgene expression of the Arig "II receptor in Y1 cells has been described 152.

Yr cells are grown under a humidified atmosphere of 5 % CO2 and 95 % air in a variety of growth media supplemented with serum. The cells approximate doubling time· is 30-40 h220· 221 . Y1 cells, for the most part, grow in culture as flat, adherent cells with polyhedral shapes maintained by a network of stress fibers and focal adhesions near the cell surface. In response to ACTH or to the adenylyl cyclase activator Forskolin, which raises intracellular levels of cAMP, Y1 cells retract their extended plasma membranes, become spherical with short processes and detach easily from the substratum on which they are grown80· 222-

224. The effects of ACTH and cAMP on cell shape are closely linked to the effects of these agents on steroidogenesis and possibly reflect an involvement of the cytoskeleton in the delivery of cholesterol to the mitochondria225-228.

81 The Y1 ceH line, as maintained in continuous culture, started to accumulate in

220 clonal variants with markedly different steroidogenesis • Among the variants,

220 221 229 230 some appeared to be completely resistant to ACTH • • • and some were

231 resistant to ACTH-induced desensitization of adenylyl cyclases • Also, a family

145 232 of ACTH receptor deficient mutants were developed • , and some mutants

233 234 harbored a dominant inhibitory protein kinase A • • These mutants have

proved to be very valuable in assessing the roles of cAMP, protein kinase A,

adenylyl cyclases, ACTH, and ACTH receptor in adrenocortical study.

B. Use of Viral Oncogenes to Develop Rodent Adrena_l Cell Lines

1. Ras transformed rat adrenal cells. Auersperg et al. explored the possibility

of establishing rat adrenal cell lines by infection with Kirsten murine sarcoma

86 92 94 virus • - • Primary cultures of rat adrenal ·cells in early passage were infected with virus and allowed to overgrow the normal cell population. The transformants

metabolized pregnenolone to progesterone and 20a.-dihydroxyprogesterone,

indicating that they retained some steroidogenic potential. However, they did not synthesize 11- or 21-hydroxylated steroids nor was their capacity to form steroids from endogenous ·cholesterol measured. A similar strategy was used to establish

86 an adrenal cell line from rat zona glomerulosa cells • These cells produced basal

levels of steroids and increased steroid production in response to ACTH or

cAMP. However, the stimula~ed levels of steroid production as estimated by

82 radioimmunoassay were very low (8 ng per 105 cells per 24 hours}, and the identity of the steroid product was not rigorously identified.

2. Cell lines from tumors in transgenic animals. Several investigators established immortalized adrenal cell lines from adrenal tumors in mice carrying

87 88 96 98 the Simian Virus 40 T-antigen as a transgene • • - • Using a human CYP11A

87 96 promoter to target expression of T-antigen to the adrenal cortex~ Mellon et a/. • " generated adrenal tumors in female mice and used these tumors to establish cell lines that produced progesterone as the major steroid product. Progesterone synthesis was not responsive to ACTH or ANG II but was stimulated by 8-bromo­ cAMP. Progesterone was synthesiz~d at a rate of 100 ng/µg of DNA per 24 h under stimulated conditions. The cells expressed CYP11A and CYP11 B1 but did not synthesize 11-hydroxylated steroids. Although the cells · produced small amounts of deoxycorticosterone, they did not express measurable amounts of

CYP21, suggesting that another enzyme carried out the steroid 21-hydroxylation

96 reaction. Curiously, these cells also expressed renin-1 mRNA •

The mouse inhibin a-subunit promoter also was used to drive T-antigen expression and to generate adrenal tumors. These tumors were postulated to

97 98 originate from the X zone of the adrenal cortex • • A cell line derived from one of these tumors (clone Ca1} produced progesterone as the major steroid product.

These cells expressed LH receptors, rendering them modestly responsive to

83 stimulation by hCG. Unstimulated Ca.1 cells produced progesterone at a relatively high rate (350 ng/106 cells per hour), and treatment with hCG produced a small but statistically significant (.::; 40%) increase in progesterone production. .

Effects of ACTH on steroid production were not documented. Treatment of Ccx.1 cells with inhibin decreased T-antigen levels and slowed the rate of proliferation.

Effects of inhibin on steroidogenesis were not reported although it would have been interesting to determine if suppression of T-antigen expression affected the rate or profile of steroid production.

Several immortalized adrenocortical ceU lines were generated from mice bearing a transgene for a temperature-sensitive form of T-antigen under control of its own

88 promoter • At permissive temperatures, one group of cells expressed SF-1,

CYP11A, StAR and CYP11 B1, suggesting that they were of fasciculata­ reticularis origin. A second group of' cells expressed SF-1, CYP11A, and StAR but not CYP11 B 1, suggesting that they had lost differentiated function or were derived from an undifferentiated · cell layer situated between the glomerulosa zone and the fasciculata zone. Mukai et al. suggested that these adrenal cell isolates may be useful in exploring the basis for functional zonation of the adrenal cortex. Major steroid products of these cells appeared to be pregnenolone and progesterone. None of the cells were responsive to ACTH or

ANG II and none of the cells expressed aldosterone synthase (CYP11 B2). At permissive temperatures, the phenotypes of the cell lines remained stable for ~

84 200 passages. Shifting cells to non-permissive temperatures or treating cells with dibutyryl cAMP had variable effects on gene expression; either treatment converted the undifferentiated cells to fasciculata-like phenotypes and also induced CYP21 and HSD3B expression.

Perhaps the most successfully established mouse adrenocortical tumor cell lines

95 were recently described characterized by Ragazzon et a/., • Using genetically targeted oncogenesis, the first immortalized cell lines, ATC1 and ATC7-L, were developed. Both ATC1 and ATC7-L have retained a complete ZF phenotype.

This phenotype was manifested at multiple levels: steroidogenic capacity, ACTH responsiveness, and expression profile of specific genes. The two cell lines were established' from adrenal tumors of two transgenic mice harboring the large T­ antigen of SV40 under the control of adrenocortical-specific promoter of Akr1 b7.

Both lines showed a significant stimulation of corticosterone secretion upon treatment with 10-11 M ACTH and then increased in a dose-dependent manner, reaching the highest values at 10-7 M ACTH. Detailed examination of expression of hormonal responsiveness of the mRNAs for the genes involved in the different steps in glucocorticoid synthesis revealed detectable levels of Mc2r (ACTH receptor mRNA) and Cyp21a1 mRNAs but were unresponsive to ACTH in both cell lines. By contrast, mRNA levels for Sr-b1, StAR, Cyp11 a 1, Cyp11 b1 and

Akr1 b7 were strongly induced in a time-dependent manner by ACTH treatment, and these inductions were not abolished by the protein synthesis inhibitor,

85 cycloheximide. Thus, the effect of ACTH on accumulation of these mRNAs is essentially transcriptional. The ATC cell lines have been developed. as novel in

vitro models, maintaining differentiated endocrine functions of ZF adrenocytes.

They have proven very useful in investigating the mechanisms of steroidogenic gene regulation in response to the physiological activator of glucocorticoid synthesis ACTH.

Ill. Bc;>VINE ADRENOCORTICAL CELL LINES (see Table IV)

Primary cultures of bovine adrenal cells have been a favored model to study adrenal cell function. This is likely due to the fact that bovine adrenal glands are readily available at abattoirs. and the large size of the gland allows for isolation of sufficient cells for large experiments. Bovine adrenal cells were the first adrenal model system to be used for the development of cells Unes through viral gene transformation. Hornsby and colleagues used SV40 to create a series of clonal

153 bovine adrenal cell lines • The addition of SV40 to the cells greatly enhanced the cell culture lifespan of the cells. The cells continued to respond to cAMP­

related agonists by induction of 17a-hydroxylase 17, 20-lyase (CYP17) and

CYP11A. However, cellular expression of CYP21 and CYP11 B were found to

154 require specialized growth conditions •

86 More recent studies by Hornsby and colleagues have combined human telomerase reverse transcriptase with SV 40 T antigen- and ras oncogene to

235 immortalize bovine adrenocortical cells • The immortalized bovine adrenal cells have been used in vivo to rescue adrenalectomized SCID (Severe combined immunodeficient) mice from glucocorticoid insufficiency through the production of cortisol. These data suggest that such strategies could be used in the future as a means of adrenocortical replacement.

IV. HUMAN ADRENOCORTICAL CELL UNES (see·Table V}

A. Development of Human Adrenal Cell Lines Using Viral Oncogenes

In a strategy similar to that used for bovine adrenal cells, Hornsby and colleagues used the SV40 T-antigen to isolate clonal populations of proliferating human fetal adrenal cells. These human adrenal clones responded to cAMP by increasing both CYP17 and CYP11A, but levels of CYP21 and CYP1181 were low. Transformed cells could be maintained in culture for 30 to 40 population doublings after isolation, but would then enter "crisis". and stop dividing. The cause of this late passage crisis is the result of telomere shortening that occurs with each population doubling and is normally not corrected by SV40

236 expression •

B. The NCI-H295 Cell Line and Related Strains

87 · 1. Origins and Steroid Synthesis. A 48-year-old African American female from

whom cell line, NCI-H295, was established, had the typical clinical, biochemical,

and pathological profile of adrenocortical carcinoma. She was diagnosed· with

acne, facial hirsutism, edema, diarrhea, weight loss, and a recent cessation of

84 menses • Computer assisted tomography revealed a large locally invasive

malignant tumor, which later metastasized to the lungs and liver, had clinical and.

biochemical evidence of excessive secretion of glucocorticoids and ketosteroids.

The excised tumor was 14- x 13- x 11-cm and was used to establish the original

NCI-H295 cell line. Tumor tissue was finely minced and the resulting suspension

was maintained in various serum-containing and serum-free culture media for a

84 one year period • Gas chromatography / mass spectroscopy (GCMS) and

radioimmunoassay were used to identify the production of steroids by these cells.

Of the 30 steroids detected, approximately 20 were identified. Based on secreted

steroids, these cells appeared to contain all of the adrenocortical enzyme

systems which presumably were present in the original tumor, including CYP11 A,

HSD3B2, CYP11 B1, CYP21, CYP17, CYP11 B2, 3(3-hydroxystJlfotransferase,

and low levels of aromatase (CYP19). Cytogenetically the H295 cell is highly

aneuploid and hypertriploid, with 30% of the cells containing a modal

chromosome number of 62. This cell line is presently available from the

American Type Culture Collection as ATCC CRL-10296 and will grow in

suspension.

88 2. Growth and Morphology. Initial tumor cell growth in conventional serum­ containing medium was unsuccessful due to fibroblast over-growth. The culture was later adapted to grow in such media. Initial or continued growth of NCl~H295 cells did not require fibroblast growth factor, a potent stimulator of normal and cultured adrenocortical cells. Due to continuous growth and development, multiple sub-strains have been adapted from the NCI-H295 cell line using alternative growth conditions to encourage substrate attachment and shorter cell cycle times. The cell culture medium growth supplement, Ultroser G (2% -

BioSepra SA, Villeneuve la Garenne Cedex, France) is a relatively defined bovine derived serum substitute which was used to increase cell growth rate.

This supplement was selected due to previous reports suggesting that it helped

237 238 retain steroidogenic cell function , • NCI-H295 cells grown in Ultroser­ supplemented medium demonstrated increased growth rate, but the cells continued to grow as floating aggregates or loosely attached cells. Over a three- · month period, changing culture medium every three days and discarding unattached cells selected cells that maintained attachment to plastic culture dishes. After characterization, it was subsequently designated as H295R to differentiate this strain from the original cells. In comparison to the parent H295 ceH line, H295R cells grow as an adherent monolayer, and population doubling time was reduced from five to two days. This substrain is propagated in

Dulbecco's modified Eagle's and Ham's F12 medium, 1% ITS plus (Collaborative

89· Biomedical Products, Bedford, MA, USA, 1% penicillin/streptomycin (Gibco},

0.01 % gentarnicin, and 2% Ultroser G (BioSepra SA, Villeneuve la Garenne

Cedex, France). Because of the difficulty in the importation of the Ultroser G serum substitute required for maintaining the NCI-H295R,. a population of these cells was selected to grow in a commercially available serum substitute (Nu­

Serum type I - Collaborative Biomedical Products, Bedford, MA, USA). The strain that can be grown in Nu-Serum is available from the American Type

Culture Collection as ATCC CRL-2128. These cells have decreased responses to ANG It and K+ but continue to respond to agonists of the protein . kinase A signaling pathways. In an attempt to develop a strain of H295 cells that would grow in co.mmerci~I serum and retain ANG II and K+ treatment, a series of sera were tested. Growth of cells in DME/F12 medium with 5' % supplementation with

Cosmic Calf Serum (CCS) (Hyclone, Logan, UT) was found to maintain cell growth and responses to ANG II and K+. This strain of H295R is advantageous because the growth medium is considerably less expensive and no supplementation with ITS-plus is needed. Also, in comparison to the original

H295 cells that grew in suspension, these also grow as a monolayer.

Subsequently, another strain of cells, designated NCI-H295A, have been

81 described which also grow as a monolayer • The method for isolation of the NCI­

H295A strain is similar to that described above, relying on the removal of non­ attached cells with medium changes and, therefore, selecting a population of

90 cells that grow as a monoJayer. It has been recently proposed that NCI-H295A

136 ceHs produce more mineralocorticoids than NCI-H295R cells ._ The same study

also showed a lack of ANG II stimulated steroidogenesis in NCI-H295A and the

angiotensin type 1 receptor was highly expressed in NCI-H295R strain compared

to NCI-H295A. This study did not show any K+ (one of the primary

mineralocorticoid regulators) regulated steroidogenesis nor did it show any

· CYP1182 expression by_ NCI-H295A. The study failed to show any legitimate

conclusive evidence about NCI-H295A strain's steroidogenesis.

3. Hormonal Response and Expression of Hormone Receptors on H295

cells. Peptide hormones, ANG 11 and ACTH are the primary ·physiologic

regulators of steroid hormone production in the human adrenal gland, along with

circulating levels of K+ (Fig. 15). The original description of NCI-H295 cells did

84 not report on the hormonal responsiveness of these cells • Subsequently, the

responses to ANG II, K+, and ACTH treatment, as well as expression of trophic

hormone receptors, were characterized for the NCI-H295R cell strain. In vivo

ANG II acts on the adrenal zona · glomerulosa to increase production of

. aldosterone through type 1 ANG II (AT1) receptors. Studies of [1 25 1] radiolabeled

ANG 11 binding to H295R cells in the presence of antagonists to the AT1 and AT2

receptors established that the H295R cells express AT1 receptors almost

103 104 exclusively • • The AT1 receptor is coupled to phosphoinositidase C and

105 increases the production of inositol phosphates in H295R cells • ANG II,

91 through the AT1 receptor, also increases H295R cell production of aldosterone.

- ANG II-mediated production of aldosterone has been shown to be differentially

regulated by the dopamine receptors D2 and D4. Both receptors are found in

H295 cells with D2 inhibiting and D4 activating aldosterone production via ANG

11239_

Under appropriate culture conditions, as described above, the H295R cell line

has shown sustained regulation of AT1 receptor expression and activity.

Regulation of aldosterone production in H295R cells is mediated via the

transcription factors Sp 1 and Sp3, which are required for AT1 receptor

240 expression • These cells readily exhibit detectable. levels of AT1 receptor

125 103 104 241 rnRNA, as well as [ 1] ANG II binding • • • Expression of transcript and

104 241 binding appears to be affected in parallel • • Thus, the H295R cell line may

be useful to define the mechanisms regulating adrenal cell responsiveness to

ANG II.

Extracellular K+ is the other major physiologic regulator of adrenal aldosterone

production. K+ increases intracellular calcium levels in H295R cells, which

appears to be the mechanism to increase aldosterone biosynthesis. Recent

studies have also shown that K+ stimulation increases the production of both

ANG I and ANG 11, suggesting that these cells may provide a model to study the

242 hypothesis that an intra-adrenal renin/angiotensin system may exist •

92 An alternate pathway for the regulation of adrenal aldosterone production is through the action of parathyroid hormone (PTH) and parathyroid hormone­ related peptides· (PTHRP), which have been shown to regulate aldosterone

243 production in freshly isolated glomerulosa cells • PTH and PTHRP also

244 stimulate H295R cells to increase aldosterone synthesis • PTH and PTHRP activate steroidogenesis in a cAMP-dependent manner in normal cells and the

H295R cell line. Considering these observations, the H295R cell strain appears to be an appropriate model to study several of the major and minor physiologic regulators of aldosterone biosynthesis.

The primary hormonal regulator of adrenal cortisol production is ACTH. The

H295R ceH line is only mildly responsive to ACTH, and most strains are completely resistant. While ACTH treatment did cause an acute increase in

244 aldosterone synthesis, long-term stimulation could not be maintained • The low response to ACTH _may reflect the low level of ACTH receptor expression in

206 the H295R cell • Therefore, most experiments designed to examine the cAMP­ dependent pathway requires the addition of either forskolin (to activate adenylyl

104 105 113 115 127 241 cyclase) or cAMP analogues • • • • • • The low response to ACTH is therefore, a drawback of this cell model. Studies directed toward ACTH action would need to be pursued using either primary cultures of adrenal cells or the Y1

220 mouse adrenal cell line, which retains ACTH responsiveness • An alternate

93 strategy would be to use transgenic technology to reinstate ACTH receptor

expression in the H295 cell line.

The ·responsiveness of the H295R cell strain is highly dependent on growth

conditions. Thus, when adapting the cells to alternate growth conditions, a

characterization of responsiveness may be necessary. Because of the bovine

components of Ultroser G, its importation into the USA from France requires a

specific permit from the U.S. Department of Agriculture. For that reason, the

· selection of a strain of H295R cells that grow in CCS (commercially available

from Hyclone) has made it very easy to maintain the cells.

4. Steroid biosynthesis. As previously noted, adrenocortical steroid hormone

biosynthesis is complex and varies dramatically between the glomerulosa,

fasciculata, and reticularis zones. The clinical features of the patient with the

adrenal tumor that gave rise to the H295 cell line indicated that steroids which

normally arise from each of the adrenal zones were produced by the tumor. The

steroids produced by the original H295 cell line, as well as the H295R strains,

maintain this diversity. The ability of the cells .to produce steroids which originate

from multiple zones of the adrenal suggests that the H295R cell line remains

pluripotent with regard to adrenocortical differentiation. In Gazdar's initial report84

concerning these cells, a broad spectrum of basal steroid hormone synthesis

was noted (30 steroids}. The steroid profile was greatly influenced by serum

94 conditions. Using the H295R cell line we have shown that these cells also

105 127 produce an array of steroids even under basal. conditions • • However, treatment with agonists appears to selectively prom

103 113 245 wm promote the cells to produce aldosterone • - • While aldosterone does not constitute the major product, even under these conditions, the H295R cell can be used to study aldosterone synthesis and factors which preferentially activate its synthesis. This is particularly important when one considers the difficulty in obtainin·g primary cultures of aldosterone-producing cells. Treatment of the H295R cell strain with agonists working through the cAMP pathway produce a pattern of steroids approaching those of the zona fasciculata and

reticularis. Steroids produced during treatment with forskolin include -cortisol,

11 f3-hydroxyandrostenedione, DHEA, DHEA-sulfate, corticosterone, 11-

127 deoxycortisol, and androstenedione • These data support the proposition that the H295R cell line can act as an appropriate model for adrenocortical steroid hormone biosynthesis and may be useful in defining the various mechanisms causing the synthesis of the "zone-specific" steroids.

5. Expression of steroid-metabolizing enzymes in H295 cells. The original

NCI-H295, as well as the H295R and H295A cell strains, have been used as

-genetic models for studying steroidogenic enzyme gene expression. The NCI­

H295 adrenocortical cells express all of the enzymes participating in normal

95 197 human adrenal steroidogenesis • In addition, the genes that encode these enzymes respond to the same second messengers controlling normal human adrenocortical function. Expression of genes encoding CYP11A, CYP17, and

CYP21, together with CYP11 B1 and CYP11 B2, were first studied in the NCI­

197 246 H295 cells • • The transcripts encoding these enzymes accumulated in response to agonists (8-bromo-cAMP, forskolin, cholera toxin, and 3~isobutyl-1- methylxanthine) which activate the protein kinase-A pathway. Consistent _with normal adrenocortical tissue, stimulating the protein kinase-C pathway. using phorbol esters resulted in decreased CYP11A1 and CYP17, but accumulation of

CYP21. In a similar manner, 8-bromo-cAMP increased CYP11 B1, CYP11 B2 and unexpectedly, aromatase (CYP19) mRNA levels. Recent studies have demonstrated that mRNAs encoding the StAR gene as well as the five forms of cytochrome P450 known to be involved in adrenal steroidogenesis (CYP11 A 1,

CYP17, CYP21, CYP11B2 and CYP11B1) are also detectable in the NCI-H295R cell sub-strain 103-1os, 112-116, 121,241, 244_

247 248 Similar to other mammalian adrenal cells • , the levels of mRNA encoding

249 CYP17 and HSD3B2 appear to be differentially regulated in NCI-H295R cells •

ANG II promoted a significant increase in mRNA level for l-:fSD3B2 and CYP11 A 1 but only marginally increased the mRNA levels of CYP17. Potent activators of the protein kinase A pathway (forskolin and dbcAMP), while increasing the message for HSD3B2, have a greater effect on levels of CYP17 mRNA. Thus,

96 activation of the protein kinase C and- calcium signaling- pathways tend to support­ a glomerulosa-like steroidogenic enzyme expression, while -activation of the protein kinase A pathway promotes fasciculata-like enzyme expression.

Another pair of steroidogenic enzymes that exhibit an adrenal zone-specific distribution includes CYP1182 and CYP1181. CYP1182 is expressed only in the zona glomerulosa and is essential for corticosterone conversion to aldosterone

(Fig. 14). On the other hand, CYP1181 is expressed at higher levels in the zona fasciculata/reticularis and is essential for the production of cortisol from deoxycortisol (Fig. 14 ). The transcripts encoding these two enzymes are increased by tre?ttment of H295R cells with activators of the protein kinase-A pathway, although the effect on CYP11 B1 mRNA levels is greater.- In addition, levels of CYP1182 mRNA are increased in H295R cells by treatment with ANG

103 116 250 251 105 116 250 11 , • • or K+ • • • The effects of ANG II on the expression of

CYP1181 were not as pronounced as was· observed for CYP1182, suggesting that the protein kinase-A and calcium pathways differentially regulate the expression of CYP1182 and CYP1181.

The H295R and H295A cell lines have also been shown to be useful model systems to define the mechanisms controlling transcription of the steroid­ metabolizing genes. Fusion genes containing the 5'-flanking DNA from

CYP1181, CYP1182, CYP17 and HSD382 have been studied using these cell

97 81 117 118 lines • - • Further regulati_on of genes in the steroidogenic pathway has been

119 121 demonstrated by a number of transcription factors identified in H295 celfs - •

The H295R cell line is the first steroidogenic cell line to maintain the full complement of adrenal steroid-metabolizing enzymes and therefore may continue to be useful in defining the elements in the 5'-flanking region of these genes that regulate transcription.

C. The SW13 human adrenal carcinoma-derived cell line. These cells were derived from a human adrenal carcinoma 142 and are available from the American

Type Culture Collection (CCL-105). The SW13 is an interesting cell culture model that has a mosaic pattern of vimentin expression and is deficient in the

252 mammalian homologues of Brahma genes, Brm and BRG1 • Although adrenal in origin, SW13 cells produce no steroids and it is not clear that it derived from steroidogenic adrenal cells. Therefore its usefulness as an adrenocortical model system is limited.

D. Human adrenocortical carcinoma (HAC) cell line and related clones. The above mentioned NCI-H295 cell line and its strains have proven useful due to the retention of ANG II dependent aldosterone production; however, they lack ACTH response. In addition, the H295 models are not clonal but represent a mixture of cells isolated from primary cultures of the original tumor. In an attempt to develop a HAC cell line, our group recently isolated clonal populations of cells from an

98 adrenal tumor. Primary adrenocortical carcinoma cells were isolated from a

· tumor removed from a child diagnosed with hypertension, obesity, and hi~sutism.

Cells were maintained in DME/F12 medium supplemented with 1Qc % CCS, ITS+

premix, penicillin, streptomycin, and gentamicin. Parental tumor cells were used

to develop forty seven discrete clones, six of which grew well enough for further

characterization. Preliminary results suggest that ACTH increased cortisol

253 production in two of the clones • The cells also retained responsiveness to Ang

254 II and K+ through the production of aldosterone • Further characterization is

·ongoing, but these cell lines promise to provide an improvement in the available

systems to study human adrenal steroid production.

V. SUMMARY

The mechanisms involving hormonal regulation of adrenocortical cell growth,

morphology, and steroidogenesis remain a topic of research. The murine Y1 and

human H295 adrenocortical cell· lines are the most widely used adrenal cell

culture models. Together, they provide unique systems to study both the

molecular and biochemical characteristics ·of adrenal function. The Y1 cell line

has provided a unique experimental system to dissect the mechanisms of ACTH

action on steroidogenesis, cell proliferation, morphology, and the factors that

govern the expression of genes required for steroidogenesis. The ability of H295

cells to produce mineralocorticoids, glucocorticoids, and C19 steroids based on

99 culture conditions has provided a valuable model to study the processes involved

in adrenocortical differentiation. However, both model systems have limitations.

The Y1, due to the deficiency of 21-hydroxylase, cannot produce corticosteroids.

The H295 models have low or no ACT~ response. The recent development of a corticosterone-producing mouse adrenal cell line and an ACTH responsive

human adrenal cell line should improve our ability to better study adrenal

toxicology and cellular physiology.

100 Table IV: Summary of Information on Rodent and Bovine Adrenal Cell Lines

Cell Line Name ACTH K+ ANG II cAMP Steroids produced Availability Reference Cited:

220 Y1 and its mutant strains + - - + Progesterone metabolites ATCC and Schimmer Lab 255 196,' 256, ' 257

258 86 Rat adrenal cells: 2FASC and + - - + Progesterone metabolites Roskelley and 7GLOM Auersperg ' Auersperg 93 94 Rat adrenocortical line: TRA + + Progesterone metabolites Lab - - . '

95 Mouse adrenal cell-lines: ATC1 + ND - + Reported glucocorticoids Martinez lab and ATC7-L

88 Immortalized mouse adrenal cell - - - + Progesterone metabolites lshimura lines: AcA201, AcE60 and AcA 101 lab Immortalized mouse adrenal cell lines: ST2, ST5-R, and ST5-L - ND ND + Progesterone metabolites Mellon and Weiner Lab 87 r Immortalized mouse adrenal cell 96 line $T5Lc - ND ND + Progesterone metabolites Mellon Lab SBAC and other Bovine Adrenocortical Cell Lines ND None reported ATCC 2~9 260 - + + ' .. (+)Positive;(-) Negative; ND: Not documented

101 Table V: Summary of lnformatio'n on Human Adrenal Cell Lines

Cell Line ACTH K+ ANG II cAMP Steroids produced Availability Reference Name.. Cited: Mineralocorticoids ' NCI-H295 - ND ND ND Glucocorticoids ATCC 84 Adrenal Androgens Mine'ralocorticoids 113 NCI-H295A - ND - + Glucocorticoids Miller Lab Adrenal Androgens 261 262' ' NCI-H295R -/+ + + + Mineralocorticoids ATCC 105 113 245. 114-116' ' Strains Glucocorticoids And ' Adrenal Androgens Rainey Lab Mineralocorticoids · HAC15 + + + + Glucocorticoids Rainey Lab 253,254 Adrenal Androgens (+)Positive;(-) Negative; ND: Not documented

102

-\ Fig. 14: Human adrenal steroid biosynthetic pathways illustrating the three main products of the adrenal: aldosterone, cortisol, and DHEA/S and the enzymes that synthesize these products. Classification for the enzymes in the P450

263 superfamily follow the guidelines previously reported • Nomenclature for the enzymes is as _followed: StAR: Steroidogenic acute regulatory protein; CYP11 A 1: cholesterol side-chain cleavage; CYP17: 17a hydroxylase 17, 20 lyase;

SULT2A 1: DHEA-sulfotransferase; HSD3B2: 3P hydroxysteroid dehydrogenase type 11; CYP21 : 21-hydroxylase; CYP 11 B 1: 11 p hydroxylase; _CYP 11 B2: aldosterone synthase.

Fig. 15: Signaling pathways that regulate steroid production in human adrenal cells. The primary regulators of adrenal steroid hormone biosynthesis are ACTH,

Angiotensin 11 and potassium, which work at the level of the cell membrane. Both the calmodulin-dependent protein kinases (CaM Kinases) and protein kinase A pathways are able to activate acute and chronic steroid hormone production.

Fig. 16:. Rodent adrenal steroid biosynthetic pathways for the production of aldosterone and corticosterone. Also illustrated are the variations seen Y-1 adrenal cell steroid production, which produces primarily 20a­ dihydroprogesterone and 11 J3,20a-dihydroxyprogesterone. Abbreviations are as described in Figure 14.

103 Fig. 14:

Cholesterol StAR CYP11A1 SULT2A1 7aOH DHEA­ sulfate

17aOH Progesterone Progesterone CYP21

Deoxycorticosterone Deoxycortisol CYP11B1 . CYP11B2 Corticosterone - Cortisol (CYP11B2]

Aldosterone

104 Fig. 15:

ACTH ANG II Calcium

ATP cAMP i Protein Kinase A ~ Cam Kinases ! ~ ! Increased StAR ~iat~ :,. , Cholesterol StAR with the ,_ J/,. ~ / surface of ~ mitochondria facilitates cholesterol entry

release of steroids Chronic Effects Acute Effects

105 Fig.. 16:

Cholesterol StAR CYP11A1

HSD382

Progesterone ~-CY-P2-1

Deoxycorticosterone--. CYP.1182

Corticosterone 11 ~ 20a-dihydroxyprogesterone (CYP11B2]

Aldosterone Corticosterone

. 106 II. MANUSCRIPTS

· Book Chapter

Adrenocortical Cell Lines

Jeniel Parmar, Anita Kulh~rya, and William E. Rainey

Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912

Abbreviated Title: Adrenal glomerulosa targeting

Key words: Cyp11 b2, adrenal cortex, zona glomerulosa, in vivo targeting

Name/Address for Correspondence: Jeniel Parmar Department of Physiology Medical College of Georgia Augusta, Georgia 30912

Telephone: 1-706-721-8363 Fax: 1-706-721-8360 E-mail: [email protected] .

107 4.2.1 Introduction

Initially, primary cultures of adrenocortical cells have traditionally been utilized to study the mechanisms controlting adrenocortical steroidogenesis. However, the eventual onset of senescence in culture· creates a recurring need for the costly and difficult isolation of fresh cultures, and subsequently increases the risk of contamination. For these reasons, the use of primary cultures has been increasingly replaced by immortalized cell lines. This chapter describes the major adrenocortical cell lines that are available for the study of physiologic and pathologic adrenocortical functions. Adrenocortical cell lines· that have been derived from adrenal tumors that have developed spontaneously or experimentally from a variety of species will be discussed. These in vitro models have allowed a multitude of studies that have broadened our understanding of normal adrenoco·rtical endocrine function as well as the properties of adrenal tumor cells.

The adrenal gland is an integral part of the endocrine system, as it regulates essential physiological functions in all vertebrates. It is noteworthy, however, tha_t considerable variations exist with regard to the anatomical locations of the adrenal and its significance as an endocrine organ among different animals. In mammals, the gland is subdivided into two different compartments-the outermost cortex and the innermost medulla. This subdivision does not exist in amphibians or fish; in fact, adrenal cells of some fish are found within the

108 kidneys, where they are collectively termed the "intrarenal" gland. During mammalian embryonic adrenocortica-t development, the adrenocortical mesenchymal cells originate from the coelomic cavity near the urogenital ridge.

These cells go on to form first the adrenogonadal primordium, which is the precursor of the adrenal cortex and the somatic cells of the ovaries. Later the· adrenocortical primordium separates to form the fetal adrenal gland, which becomes encapsulated by surrounding mesenchymal cells. With ongoing development, adrenal medullary cells start to migrate into the fetal adrenal gland, eventually forming the adrenal medulla. In a last step the adult adrenal cortex

·arises with its classical zonation and the fetal adrenal cortex regresses.

The histologic zonation of the cortex in three functionally distinct regions (zona glomerulosa, zona fasciculata, and zona reticularis) was first described by Arnold

43 in 1866 • For all adrenocortical steroidogenesis, the rate-limiting step is the translocation of cholesterol from the outer mitochondrial membrane into the mitochondrion for cholesterol side-chain cleavage by the cytochrome P450

44 CYP11 A 1, the first enzyme in the steroidogenic pathway (Fig. 17) •

In the zona glomerulosa cholesterol is used to synthesize mineralocorticoids in response to Angiotensin II (ANG II}. and potassium (K+) stimulation (Fig. 18). In the zona fasciculata cholesterol is used for glucocorticoid synthesis under the regulation of ACTH. In humans, the zona reticularis metabolizes cholesterol to

19-carbon (C 19} steroids, including DHEA and DHEA-sulfate, a process that is

109 r regulated by ACTH and yet-to-be determined factors. There is no physical barrier separating the three different adrenocortical zones. The molecular mechanisms that cause the zone-specific expression pattern of these enzymes are yet to be resolved. However, it has been established that these regulatory mechanisms control the relative expression of the enzymes required for steroidogenesis as well as the size and structural integrity of each of the zones.

There are still numerous molecular and cellular studies needed to better define the mechanisms regulating adrenal cell steroid production, prompting the need for further studies both in vivo and in vitro. Appropriate, well-characterized adrenal model systems are critical for-these studies. Initially, primary cultures of adrenocortical cells proved useful for examining the mechanisms controlling many aspects of adrenal physiology. However, several issues limit the use of the primary adrenal cell culture models, including cell growth in culture, responses to agonists and maintenance of steroidogenic capacity. It is well-documented that the ability of adrenal primary cultures and cell lines to produce steroids and/ or to respond to ACTH, ANG II and K+ can change over time. The expression of the enzymes involved in steroid hormone biosynthesis can also change under in vitro culture conditions. As a result, the steroids produced by cells in culture can be quite different from those produced in vivo. Perhaps the most practical limitation to the use of primary adrenal cell cultures is the constant requirement for freshly acquired tissue and the difficulties associated with the isolation of cortical cells in the numbers needed to accomplish in vitro studies. Taken together the problems

110 and limitations of primary cultures have caused many groups to attempt the use of adrena~ tumors cells as model systems. Moreover, these cell lines can· be used in xenotransplant models and in vitro studies to- research adrenocortical cancer. A number of in vitro tumor cell models have been investigated, including

77 79 cell suspensions from acutely dispersed adrenal tumors, - , established cell

80 84 lines from tumors - and the isolation of immortalized cells from transgenic

85 88 animals - . Herein, we provide a summary of the adrenoco_rtical cell lines and. provide some information as to their development and 'utility.

111 4.2.2 Human Adrenocortical Cell Lines (see Table VI)

4.2.2.1 The NCI-H295, H295R and H295A Adrenocortical Carcinoma Cell

Lines ·

Origin and development

The NCI-H295 cell line was established from a female patient diagnosed with an

84 adrenocortical carcinoma • A large 14- x 13- x 11-cm adrenocortical invasive tumor was detected by CT imaging and was later reported to have metastasized to the lungs and liver. In the initial process of developing the original NCI-H295 cell line, the tumor tissue was finely minced, defragmented and maintained in

84 various serum-containing and serum-free cult_ure media for a one year period •

Monoclonal cell lines were not established and therefore, the NCI-H295 cell line represents a mixed population of tumor cells from the original carcinoma.

Cell line growth and characterization

Since parental NCI-H295 cells sustained a slow rate of growth in the initially described culture media, alternative growth conditions were sought in order to segregate a population of cells with better attachment and growth. Initially, the cell culture medium growth supplement, Ultroser G, a relatively defined bovine­ derived serum substitute, was used to increase cell growth rate. Previous

112 assessments. of this media supplement reported increased adrenal cell growth

237 238 and retention of steroidogenic function • • Despite the increased growth rate

· of NCI-H295 cells grown in Ultroser supplemented medium, the cells continued

to grow as floating aggregates or loosely attached cells, making it difficult to

experimentally treat and manipulate these cells in culture.

In an attempt to establish a superior cell model, the parental NCI-H295 cells were

continuously flushed ·with fresh growth medium for a period of three months to

deliberately remove the floating suspended cells and keep the viable attached

subtype. The newly selected population of cells was subsequently designated as

H295R to differentiate this strain from the original cells. H295R cells grow as an

adherent monolayer (Fig. 18A). The population doubling time of the new H295R

c~lls was approximately 2 days in comparison to the original parental NCI-H295

cell line which grows in suspension and takes approximately 5 days to double in

population. Unfortunately, the success of the H295R was challenged by the

limited availability of the Ultroser-G medium in select countries.

Consequently, a series of sera were tested in an attempt to develop a population

of H295R cells that would grow in a relatively inexpensive commercial serum and

retain ANG II and K+ responsiveness. Growth of cells in DME / F12 medium

supplemented with 5 % Cosmic Calf Serum (CCS) (Hyclone, Logan, UT) was

found to maintain cell growth and responsiveness ·to ANG 11 and K+. Another

strain of H295R cells was also selected to grow in commercially available Nu-

113 Serum type I (10 % - Collaborative Biomedical Products, Bedford, MA). This

strain is available from .the American Type Culture Collection· as ATCC CRL-

. 2128._ These cells however have a decreased response to ANG II and K+,

though the cell doubling time· is relatively similar to those grown in other sera.

Since their first establishment, H295R cells have been cultured with variety of

media in which they have shown a range of steroidogenic responses and

adherent properties. As a result the agonist responses and growth

characteristics of H295H cells vary depending on individual laboratory conditions.

As with H295R, a second population of the parental NCI-H295 cell line was

81 se~ected to grow as a monolayer, yielding another strain of the original cell line ,

designated H295A. The method for isolation of the H295A strain was similar to

that described for H295R, relying on the removal of non-attached cells with

medium changes and, therefore, selecting a population. of cells that grow as a

monolayer.

Expression of hormone receptors and hormonal responsiveness

Although ANG 11, K+ and ACTH are the primary hormonal regulators of steroid

hormone production in the human adrenal cortex (Fig. 17), there is no description

84 of hormonal responsiveness in the original description of the NCI-H295 cells .

Subsequently, the responses to ANG 11, K+, and ACTH treatment, as well as

expression of hormone receptors, were cha~acterized for the H295R cell strain.

114 ANG H is the primary hormonal. regulator within the renin-angiotensin-aldosterone system (RAAS}; upon conversion from its precursor by. angiotensin converting­ enzyme, it acts on the adrenal glomerulosa by binding to type 1 ANG II (AT1) receptors to increase the production of aldosterone. H295R cells have proven to be a useful model to study ANG II regulated aldosterone production. Only AT1 receptor antagonists have a significant inhibitory effect while type 2 ANG 11 (AT2) receptor antagonists have little impact on ANG II stimulation, demonstrating that

103 104 H295R cells respond almost exclusively to the AT1 receptors • •

Subsequent studies also revealed AT1 receptor couple with phosphoinositidase

105 C and increased production of inositol phosphates in H295R cells •

Circulating extracellular K+ is the other major physiologic regulator of adrenal aldosterone production. An increase in intracellular calcium levels in response to elevated K+ mediates an increase in aldosterone biosynthesis. In addition, there

242 is evidence of an intra-adrenal renin / angiotensin system , in which K+ stimulation increases the production of both ANG I and ANG II. The H295R cells are used by many laboratories as a model to study the mechanisms of K+

106 111 regulation of adrenal steroid production - •

ACTH is the primary hormonal regulator of adrenal cortisol production. The

H295H cell line is only mildly responsive to ACTH, while most other

82 adrenocortical cell lines are completely unresponsive • ACTH treatment of

115 H295~ cells does cause an acute increase in aldosterone biosynthesis, though

83 244 the ceH line lacks long term responsiveness • • The low response to-ACTH is therefore, a drawback of this cell- model. Poor ACTH response is in part a reflection of low levels of ACTH receptor expression in the H295R cell, however even after induction of receptor expression the cells still exhibit a poor

83 206 response • • Since ACTH primarily regulates cortisol production through cAMP signaling, the addition of either forskolin (to activate adenylyl cyclase) or

104 105 113 115 127 241 cAMP analogues • • • • • can be used to overcome this dilemma. An alternate strategy could involve the use of transgenic technology to reinstate

ACTH receptor expression in the H295R cell line; however, attempts to do so have increased receptor expression but not response to ligand (unpublished observation). At present, investigations of_ ACTH responsiveness rely heavily on

220 the mouse adrenal cell line Y1, which retains ACTH responsiveness •

Steroidogenesis

Most research focuses. on adrenal production of aldosterone, cortisol and DHEA for which the steroidogenic pathways are well defined (Fig. 19). After isolation and establishment of the parental NCI-H295, the cell line was found to produce an array of steroids, including those characteristic of the adrenal cortex and some unique to tumor cells. Primary assessment of steroidogenic capacity was performed using gas chromatography / mass spectroscopy (GCMS) and

radioimmunoassay. Of a total of 30 different steroids reportedly synthesized and

116 secreted by NCI-H295, approximately 20 were formally identified as known steroid hormones. The production of these steroids suggested the presence of all of the adrenocortical steroidogenic enzymes including CYP11 A 1, HSD382,

CYP1181, CYP21, CYP17, CYP1182, and 3(3-hydroxysulfotransferase. The

H295R strain also maintains this steroidogenic profile; however, it is greatly influenced by serum conditions.

The ability of these cells to produce steroids which originate from multiple zones of the adrenal suggests that the H295R cell line remains pluripot.ent with regard to adrenocortical differentiation. These cells produce an array of steroids even

105 127 under basal conditions • • This is particularly important when one considers the difficulty in obtaining primary cultures of aldosterone-producing cells. It is noteworthy that treatment with agonists appears to selectively promote the

103 113 245 synthesis of certain zone-specific steroid hormone groups in H295R • • •

For instance, production of aldosterone drastically increases following ANG II and K+ stimulation, while treatment with forskolin induces cortisol, 11 (3- hydroxyandrostenedione, DHEA, DHEA-sulfate, corticosterone, 11-deoxycortisol,

127 and androstenedione.production • While H295R cells do not represent a single zone of the adrenal cortex, the cell line provides ample flexibility for studies on adrenocortical steroidogenesis. As stated above, with a wide variety of effective agonists, a mosaic of unique steroid expression profiles are possible in H295R cells, thus making this cell line a potential model for steroid hormone

61 128 135 biosynthesis in all adrenocortical zones • - •

117 · Steroidogenic enzyme expression

-The original NCI-H295 adrenocortical cells express all of the enzymes

99 101 participating in normal human adrenal steroidogenesis • • Along with the

parental NCI-H295, the H295R and H295A cell strains have also been used as

genetic models for studying steroidogenic enzyme gene expression. Transcripts

encoding· StAR, as well as the five forms of cytochrome P450 . known to be

involved in normal adrenal steroidogenesis (CYP11A1, CYP17, CYP21,

103 105 112 116 CYP1182 and CYP-1181), are detectable in the H295R cell strains • • - •

127 241 244 • • • Moreover, as with primary cultures and other mammalian adrenal

247 248 cells • , the levels of mRNA encoding CYP17 and HSD382 appear to be

249 differentially regulated in H295R cells • Furthermore, the transcripts encoding

CYP1181 and CYP1182 enzymes are increased by treatment of H295R cells

with activators of the protein kinase-A pathway, although the effect on CYP1181

mRNA levels is greater (Fig. 17). In addition, ANG II and K+ cause a drastic

103 105 116 250 251 increase in CYP1182 mRNA encoding aldosterone synthase • , • , •

The original NCI-H295 and its strains have proven to be useful models for

defining the mechanisms controlling adren_ocortical steroidogenesis. The basal -

promoter and enhancer elements of the genes encoding CYP1181, CYP1182,

81 117 118 CYP17 and HSD382 . • - together with_the transcription factors mediating the

118 expression of these genes has been studied in great detail using these cell lines11e-121; 111, 122;.12a, 1~. 131-141_

NCI-H295 cell line· as adrenal cancer therapy tool

In vitro cancer therapy screening represents an important mechanisms of drug

testing264. The National Cancer Institute has made wide use of. neoplasms and

-carcinoma cell lines for anticancer drug screening and drug discovery265-268 .

Below is a brief description of the use of the NCI-H295 and its sub-strains as

model systems for the study of cancer therapies.

NCI-H295 has been widely used as a model for screening anti-cancer drugs.

-Suramin, one of the first anti-cancer drugs tested in the NCI-H295 cells reduced

the production of glucocorticoids, mineraloco~icoids and adrenal androgens in

this cell line269. Schteingart and colleagues screened mitotane and mitometh,

two distinct anti-cancer drugs in NCI-H295. Mitotane strongly suppressed cell

growth and mitometh was not effective270. Fallo et al. tested the cytotoxic/ anti­

proliferation drugs, - taxol and paclitaxel, which effectively exhibited dose­

dependent inhibition of cellular growth and reduced steroidogenesis in NCI­

H295271· 272. Adrenostatic compounds aminoglutethimide (AG), metyrapone

(MTP) and etomidate (ETO} were also tested in NCI-H295 for their

antiproliferative property. AG and ETO inhibited cell proliferation and ETO was

much more potent273. Thiazolidinediones (TZDs) are specific peroxisome

119 proliferator-activated receptor (PPAR)-gamma ligands. Examination in NCI-H295 suggest that TZDs might have favorable effects in the treatment of a variety of

274 tumors as differentiation-inducing agents • NCI-H295 cells have also been utilized to assess the role of. certain interferons, chemokines and growth factors

215 2 in adrenocortical ~ancers - n. Preclinical assessment of anti-cancer compounds with potent antiproliferative activity in NCl-295 cells is an essential first step before any clinical evaluation in Phase I, II and Ill programs could be commenced.

Recent studies have utilized NCI-H295 cells in cell culture and as a xenograft model to evaluate the efficacy of a variety of new potential targeted therapies. T­ cell factor / J3-catenin antagonists, PKF115-584278 and type I insulin-like growth

279 factor receptor inhibitors have been recently tested in the NCI-H295 • These studies are aimed at a limited number of -known genetic mutations in adrenocortical carcinoma that are present in the NCI-H295 cells.

4.2.2.2 Human Adrenocortical Carcinoma, Clone 15 (HAC15) Cell Line and

Related Clones

The above mentioned NCI-H295 cell line and its strains have proven useful due to the retention of ANG ll~dependent aldosterone production. However, as previously discussed, they lack significant ACTH responsiveness. In addition, the NCI-H295 models are polyclonal, as they represent a mixture of cells isolated

120 from primary cultures of the original tumor. In an attempt to develop a new human adrenocortical carcinoma (HAC) cell line, the authors recently isolated

82 83 monoclonal populations of cells from an adrenal tumor • • Primary adrenocortical carcinoma cells were isolated and used to develop 47 discrete clones, six of which grew well enough for further characterization. Initially, the ceHs were maintained in DME / F12 based growth medium supplemented with 10

% Cosmic Calf Serum (CCS), penicillin, streptomycin, gentamicin and ITS+

Premix (a solution containing human recombinant insulin, human transferrin, selenious acid, BSA, and linoleic acid). Preliminary results suggested that ACTH . stimulation increased cortisol production in two of the clones. Same clones also retained responsiveness to ANG II and K+ through the production of aldosterone.

Further analyses of these cells will judge, whether they are an improvement over the currently available systems for the study of human adrenocortical steroid

83 production •

HAC15 origin and development

HAC15 was established from an adrenocortical carcinoma removed from an 11 month old female who presented with hypertension, hirsutism, and associated obesity. The carcinoma was initially fragmented and tissue was finely minced into small pieces and incubated in DME / F12 medium containing 0.1 % collagenase. For a final digestion and dispersion, the cells were further incubated with the addition of 0.01 % deoxyribonuclease (DNase I). Once finely

121 dispersed, parental HAC cells were plated at approximately 1000 cells per 100 mm petri-dish. Sterilized cloning rings were placed around each of the clones and trypsin was used to remove the cells (Fig•. 20). After a close assessment of several steroidogenic clones, the most responsive clone to ANG II, K+ and ACTH

83 treatments-HAC15-was further characterized •

HAC15 cell line growth, steroidogenesis and enzyme expression

HAC15 ·cells demonstrate structural features previously observed for the H295R carcinoma cells, including the loss of contact inhibition which leads to cells growing as attached clumps (Fig 18B). As with H295R, HAC15 cells grow as attached cells; however, unlike H295R, they seem to aggregate and grow as clusters of cells in a staggered-stacking pattern.

Upon stimulation with ANG II and K+, a robust increase in aldosterone level is

observed in the HAC15 cell line. These cells respond to ACTH as it causes a

modest yet significant increase in cortisol and DHEA. Other HAC clones have

reportedly responded with increased aldosterone and cortisol production to ANG

11 but have lost ACTH responsiveness. Bypassing the ACTH receptor by treating with forskolin significantly increases both aldosterone and cortisol production in

HAC15. Other clones also respond to forskolin, with a similar increase in both steroids.

122 The HAC15 cell line has proven practical for steroidogenic studies as it expresses all of the genes involved in the adrenocortical pathway including- StAR,

CYP11A1, CYP17, CYP21, HSD3B2, CYP11B1, CYP11B2, and SULT2A1.

Treatment with ANG II, K+, ,ACTH and forskolin causes significant increase in

StAR, CYP11A1, CYP17, CYP21, CYP11 B1, and CYP11 B2 mRNA as compared

83 to untreated cells •

The melanocortin 2 receptor (MC2R) is expressed in HAC15 cells with mRNA levels that are significantly higher than in the H295R cell line. However, the expression of the MC2R and response of the HAC clones is not as robust as that seen in the Y1 adrenal cell line. In summary, the HAC15 cell line exhibits hormonal response, steroidogenesis and expression of steroid-metabolizing enzymes and improved ACTH response compared to the H295R.

4.2.2.3 Cytogenetic Analysis of H295R and HAC15 Cell Lines

The HAC15 and H295R cell lines were karyotyped using standard trypsin

Giemsa banding methods. Briefly, each cell type was exposed to colcemid for 2 h at 37 °C. In HAC15, the chromosome_ number ranged from 68-74 chromosomes with modal chromosome number of 70. It appeared that there were two populations in the HAC15 cells that exhibited different karyotypes. One population exhibited a near pseudotriploid karyotype with a modal chromosome

123 number of 70 (Fig. 21A). This was not a true triploid state because of the loss of certain chromosomes and gain of two copies of other chromosomes. A true triploid has three copies of each chromosome as compared to the two copies of each chromosome seen in a normal diploid female.

The other population of HAC cells exhibited a hyperdiploid karyotype with a modal chromosome number of 52. This HAC15 cell population demonstrated the same abnormalities as seen in the near triploid cell population but had a loss of several of the abnormal and normal chromosomes. No new abnormalities were present in the hyperdiploid clone. It is likely that the hyperdiploid clone evolved from the near triploid cell population.

In the H295R cell strain used in our laboratory, the chromosome number ranged from 52 to 58 with a modal chromosome number of 55 (Fig. 21 B). This · represents a shift from the modal chromosome number of 62 observed in the

84 original NCI-H295 cell population described in 1990 by Gazdar and colleagues •

A composite description of the H295R and HAC15 cells with each structural and numerical chromosome abnormality, as well as chromosome markers have been summarized in Table VII. It is interesting that both the H295R and HAC15 cell lines demonstrated some similar chromosome abnormalities that include abnormal chromosome 3 with interstitial deletion, chromosome 9 with additional material on 9p, deleted 11 p, deleted 12p, chromosome 16 with additional

124 material on the distal end of the long arm, and chromosome 21 with additional material on distal 21 q. Also, there were at least three marker chromosomes in both cell lines that appear to be similar by morphology and banding analysis.

However, it is likely that the karyotype pattern will change as the cells are maintained in culture for extended periods and moved from laboratory to laboratory.

4.2.2.4 Pediatric Adrenocortical Adenoma Derived Cell Line

Origin and development

Primary adrenocortical adenoma cells were isolated from a non-hemorrhagic region of a functional tumor from a 1 year old female that presented with

275 virilization and Cushing syndrome . This pediatric adrenocortical cell line was last reported to have sustained growth· and viability after eight passages.

Sections. of tumor were initially fragmented and subjected to collagenase digestion. Fine tumor fragments were then segregated and plated in two different media; DMEM and Opti-MEM 1 (reduced serum medium of MEM) and supplemented with 1O and 2 % fetal bovine serum (FBS) and 1 % penicillin / streptomycin maintained under standard culture conditions (37 °C in 95 % air-5

%CO2).

125 Growth and steroidogenesis

The cell line iniUally grew at a slow rate with a spindle ~ike morphology in both of the media. As with the HAC15 cell line, a flattened homogeneous morphology, characteristic of a single cell-type origin was noted. Melan-A, a melanocytic differentiation marker, which is expressed in steroid hormone producing adrenal

280 adenomas and carcinomas , was seen in all examined cells. All steroidogenic studies were performed during the ·fifth .passage in medium from 5 day experiments. Biosynthesis of cortisol, aldosterone, androstenedione, and 17-0H progesterone was observed along with the expression of steroidogenic enzymes namely; HSD382, CYP1181 and CYP21 were reported.

Taking into consideration that this newly developed pediatric cell line has last reported to have only reached eight passages, the likelihood of the cell line's viability remains to be established. As these cells are part of a mixed tumor cell population further clonal selection may be necessary.

4.2.2.5 The SW13 Human Adrenal Carcinoma Derived Cell Line

Although adrenal in origin, SW13 cells produce no steroids and it is unclear whether the cell li~e was derived from steroidogenic adrenal cells. These cells were derived from a human adrenal· carcinoma 142 and are available from the

126 American Type Culture Collection (CCL-1_05). ·The SW13 is an interesting cell culture model th~t has a mosaic pattern of vimentin ~xpression and is deficient in the mammalian homologues of Brahma genes, Brm and BRG1 252. Due to the lack of steroidogenesis, its usefulness as an adrenocorticaJ model system is limited.

4.2.2.6 The ACT-1 Human Adrenal Carcinoma Derived Cell Line

Tumor cells were isolated from a 62 year old male who was initially diagnosed with a left adrenocortical carcinoma. After a surgical removal of tumor tissue, aggregated pieces were excised, minced and passed through a cell strainer to dissociate the cells. Cells were supplemented with 10 % FBS, insulin-transferrin­ selenium solution and appropriate antibiotics maintained at 5 % CO2 and 95 % air. In its initial description, ACT-1 cells were only shown to express HSD3B2 enzymes and were devoid of any adrenocortical steroids281 . Hence, ACT-1 has limited adrenocortical function and therefore serves a restricted role for normal adrenal function. However these cells may prove useful to screen adrenocortical carcinoma therapies.

4.2.2.7 The RL-251 Human Adrenal Carcinoma Derived Cell Line An adrenocortical carcinoma was identified by CT scan in a 75 year old male who presented with high blood pressure (219 / 107 mm Hg} and multiple spells of chills, warmth, sweating and flushing. A 5 x 6 x 7 right adrenal mass was removed by laparotomy and placed in cell suspension. Initially, tumor sample was treated with collagenase and plated in fibronectin-coated plastic with RPMI

1640 medium containing 1O % FBS and fibroblast growth factors with a population doubling time of 36 h. As with H295R and HAC15 cell lines, RL-251 also exhibits abnormal karyotypic profile with diploid karyotype consisting of numerous deletions and translocations. Assessments of adrenocortical function showed atypical steroidogenesis and hormonal response with insignificant

277 cortisol production and no ACTH stimulation • To date, there is no description of any other corticosteroid production by the RL-251 cell line. Intriguingly, in its initial report, RL-251 cells expressed ample amounts of interleukin-a, epithelial cell-derived neutrophil-activating peptide 78, growth regulated oncogene-a and growth regulated oncogene-y. These are CXC chemokine family cytokines that have potent angiogenic activity essential for tumor growth. It is speculated that these chemokines may play a role in enhancement of carcinoma growth and proliferation by an autocrine or paracrine manner. A lack of reported steroidogenesis and hormonal responses do not make these cells an appropriate model to study normal adrenocortical function, however, these cells may prove useful in identifying the role of chemokines in adrenocortical carcinomas.

128 4.2.2.8 Human Adrenal Cell Lines from Viral Oncogenes

Simian virus 40 (SV40) T-antigen is a hexamer protein that is an oncogene which

is capable of transforming many cell types. SV40 T-antigen was used by

282 Hornsby and colleagues to transform human fetal adrenal cells • The strategy

yielded human fetal adrenal cell clones that responded to cAMP with an increase

in both CYP 17 and CYP 11 A, but no · change in CYP21 and CYP 11 B 1.

Transformed cells were maintained in culture for 30 to 40 population doublings

after isolation, but then entered ''crisis" and stopped dividing. The cause of this

late passage "senescence" is the result of telomere shortening that occurs with

236 each population doubling and is normally not corrected by SV 40 expression •

4.2.3 Rodent Adrenocortical Cell Lines (see Table VI)

4.2.3.1 The Y1 Adrenal Cell Line

· Presumably following. exposure to radiation during nuclear weapon testing, an

143 adult LAF1 {C57L x A/HeJ) mouse developed an adrenal tumor • Gordon Sato

and colleagues 144 adapted the transplantable tumor to grow in vitro by alternately

propagating dispersed tumor cells as monolayer cultures and as tumors in mice.

The most robust clones developed from the mixed population of tumor cells was

129 named Y1, and ·it· was this clone that was deposited at the American Type

Culture Collection (ATCC CCL-79)8°.

It is noteworthy that the major steroid pathways present in cultured Y1 cells are unlike the normal physiologic mouse adrenocortical steroid pathway. For instance, Y1 cells in culture produces 2Oa-hydroxy-~4-pregnen-3-one (2Oa - dihydroxyprogesterone} and 11 J3,2Oa-dihydroxy-~4-pregnen-3-one (11 J3,2Oa­ dihydroxyprogesterone)208· 209 . (Fig 22). This abnormal steroid profile relates to the deficiency in 21-hydroxylase (Cyp21 )210 coupled with. an increase in 2Oa­ hydroxysteroid dehydrogenase activity208.

Y1 cells could be grown under a humidified atmosphere of 5 % CO2 and 95 % air in a variety of growth media supplemented with serum. Doubling time for this cell line is merely 30-40 h220· 221 . These cells grow in culture as flat, adherent cells with polyhedral shapes maintained by a network of stress fibers and focal adhesions near the cell surface. Upon stimulation with any of the cAMP inducing agonists, Y1 cells retract their extended plasma membranes, become spherical with short processes and detach easily from the .substrate on which they are grown so, 222-224.

The ability of ACTH to stimulate Y1 cell steroidogenic activity and enzymes is similar to that seen in primary mouse adrenal cells and other agents that raise intracellular levels of cAMP. For instance genes encoding the ACTH receptor145·

130 206 211 214 214 216 13 217 218 211 212 , Cyp11 a • , Cyp11 b1 - , star2 • • , adrenodoxin • , adrenodoxin

212 219 reductase and the HDL receptor (SR-BI) • A wealth of information has been gained through studying this cell line and several of its sub-clones which have proved to be invaluable in assessing the roles of cAMP, protein kinase A, adenylyl cyclases, ACTH, and ACTH receptor in adrenocortical cell physiology.

Y1 cells do not respond to ANG II, although transgene expression of the ANG II

152 receptor in Y1 cells has been described .

4.2.3.2 Experimentally Induced Rodent Adrenal Cell Lines .

Kirsten murine sarcoma virus (MSV) transforms cells resulting in reduced requirement for attachment, density and reliability of other cells in terms of growth and development. Auersperg et al. took primary cultures of rat adrenal cells in early passage and infected them with MSV and allowed them to overgrow

86 92 94 the normal cell population • • _ The clones exhibited little steroidogenic potential and only produced progesterone and 20a-dihydroxyprogesterone.

Similarly, an adrenal cell line from rat zona glomerulosa cells86 was created but found to produce negligible steroids (8 ng per 105 cells per 24 h), despite responsiveness to ACTH and cAMP agonists.

An SV40 T-antigen strategy was also attempted in the quest of developing immortal· rodent cell lines. Several investigators established immortalized adrenal cell lines from adrenal tumors in mice carrying the Simian Virus 40 T-

131 87 88 96 98 antigen as a transgene • • - • Mellon et al. used a human CYP11A promoter

87 96 to target expression of T-antigen to- the adrenal cortex • and generated adrenal

tumors in female mice and used these tumors to establish cell lines. These cell

lines were not responsive to ·either ANG II or ACTH and only produced

progesterone following treatment with cAMP analogues. Progesterone was

synthesized at a rate of 100 ng / µg of DNA per 24 h under stimulated conditions.

96 Noteworthy, these cells reportedly produced renin-1 mRNA •

In terms of developing immortalized rodent adrenocortical cell lines, several

others have utilized similar transgenic approaches such as the use ·of mouse

inhibin a-subunit promoter to drive T-antigen expression to generate adrenal

97 98 tumors • . As with others, these cells only produced progesterone.

· Subsequently, another group used a strategy with mice bearing a transgene with

88 a- temperature-sensitive form of T-antigen under control of T-antigen promoter .

At the permissive temperatures, one group of cells expressed Sf 1, Cyp11 a 1, Star

and Cyp11 b1, suggesting that they were of fasciculata-reticularis origin. A

second cell line expressed Sf1, Cyp11 a 1 and Star but not Cyp11 b1, suggesting

that they had lost differentiated function or were derived from an undifferentiated

cell layer situated between the glomerulosa zone and the fasciculata zone. None

of the cells were responsive to ACTH or ANG II and none of the cells expressed

· aldosterone synthase (Cyp11 b2).

132 Perhaps the most successful oncogene targeted mouse adrenocortical tumor cell

95 lines are described and characterized by Ragazzon et a/. • Using genetically targeted oncogenesis, adrenocortical immortalized cell lines, ATC1 and ATC7-L, were developed. AKr1 b7 gene promoter was used to drive the expression of

SV40 T-antigen. The two cell lines were established from adrenal tumors of two transgenic mice harboring the large T-antigen.of SV40. Both ATC1 and ATC7-L have retained a zona fasciculata phenotype evident by their robust steroidogenic capacity, ACTH responsiveness and expression profile of steroid-metabolizing genes._ Both cell lines responded well to ACTH stimulation with a concentration­ dependent increase in corticosterone production. Examination of hormone responsiveness revealed detectable levels of Mc2r and Cyp21 mRNAs but these transcripts were not responsive to ACTH in either cell line. By contrast, mRNA levels for Srb1, Star, Cyp11 a 1, Cyp11 b1 and Akr1 b7 were strongly induced in a time-dependent manner by ACTH treatment, and these inductions were not abolished by the protein synthesis inhibitor, cycloheximide. The ATC cell lines represent novel in vitro models, maintaining differentiated endocrine functions of zona fasciculata. They may prove to be very useful in investigating the mechanisms of steroidogenic gene regulation in response to the physiological activator of glucocorticoid synthesis, ACTH.

133 4.2.4 Bovine Adrenocortical Cell Lines (see Table VI)

In terms of primary cultures, due to the size and availability of bovine adrenals, bovine adrenocortical cells have remained a critical resource for the study of adrenocortical biology. Hornsby and colleagues have used SV40 to create a

153 series of clonal bovine adrenal cell lines • The introduction of SV40 greatly enhanced the lifespan of the cells in culture. A population of cells continued to respond to cAMP-related agonists by induction of CYP17 and CYP11A.

However, cellular expression of CYP21 and CYP11 B was found to require

154 specialized growth conditions • Subsequently, Hornsby et al. have combined expression of human telomerase reverse transcriptase with SV40 T antigen and

235 ras oncogene to immortalize bovine adrenocortical cells • The immortalized bovine adrenal cells have been used in vivo to rescue adrenalectomized SCIO

(Severe combined immunodeficient) mice from glucocorticoid deficiency through the production of cortisol.

4.2.5 Summary

Adrenal cell- lines represent a crucial tool for molecular and cellular studies that cannot practically be done in animal models. The murine Y1 and ·human H295 adrenocortical cell lines are currently the most widely used adrenal cell culture models. The Y1 adrenal cell line has long provided a unique experimental system to dissect the mechanisms of ACTH action on steroidogenesis, cell

134 proliferation, morphology, and the factors that govern the expression of genes

required for steroidogenesis. The additions of the human H295R and HAC15 adrenal carcinoma cells that produce mineralocorticoids, glucocorticoids, and

C19 steroids has provided valuable models to study the processes involved in

adrenocortical differentiation. However, both the human and murine adrenal

model systems have limitations. The Y1 cell, due to the deficiency of 21-

hydroxylase, cannot produce corticosteroids. On the other hand, the H295 cell

· models have low or no ACTH response. The recently described ACTH

responsive murine ATC1 / ATC7-L and human HAC15 cell lines offer new

opportunities to study the regulation of adrenal steroid production. In addition,

the HAC15 offers a new model to test human adrenal carcinoma therapies.

Together, they provide unique model systems to study both the molecular and

biochemical characteristics of adrenal function.

135 Table VI. Summary of Information on Adrenocortical Cell Lines

Ang Cell Line Name Species . II K+ ·ACTH .cAMP Steroids produced Reference Mineralocorticoids NCI-H295 ND ND ND Glucocorticoids 84 Adrenal Androgens Mineralocorticoids NCI-H295A ND +- Glucocorticoids 261 262 Adrenal Androgens

Mineralocorticoids NCI-H295R 105 113 245. -/+ Glucocorticoids 114-116' ' Strains + + + ' Adrenal Androgens

RL-251 ND ND ND None reported 277

ACT-1 ND ND ND ND None reported 281

SW13 ND ND ND ND None reported 142

Pediatric ad renocortical Mineralocorticoids ND ND ND ND 275 adenoma derived Glucocorticoids cell-line Mineralocorticoids HAC15 + + + + Glucocorticoids 82,83 Adrenal Androgens 220 Y1 and its mutant Progesterone 255 196,' 256, ~ + + strains metabolites ' 257

Rat adrenal cells: Progesterone 2FASC and 258 86 + + metabolites 7GLOM Rat Progesterone 93 94 ad renocortical -' . + + metabolites line: TRA Mouse adrenal - 95 cell-lines: ATC1 v•r··--· ND + + Glucocorticoids and ATC7-L

136 - - Immortalized mouse adrenal Progesterone cell lines: ....._ + 88 - - - metabolites AcA201,AcE60 andAcA101 Immortalized mouse adrenal Progesterone cell lines: ST2, ..___ ND ND 87 - + metabolites ST5-R, and ST5- L Immortalized Progesterone mouse adrenal ND ND - metabolites and 96 ceU-line ~ + Glucocorticoids ST5Lc SBAC and other Bovine .. ... + ND + None reported 259 Adrenocortical ' - Cell Lines Abbre v·IatIon. ND, Not determined

137 Table VII. Comparison of cytogenetic abnormalities in HAC15 and-H295R cell lines.

HAC15 H295R

Modal number of chromosomes: 68-74 Modal number of chromosomes: 52-58 -X -X add (2)( q37) -2,del(2}( q35) del(3)(p13p21) del(3}( q27) -4,-4 del(4)(q31.3) +5,+5,del(5(p13)x2 add(5)(p15) +6,add(6}( q27) +6 del(8)(p21) -8,add(8}(q24) +del(9)(p22},add(9)(p2?)x2 +9,+9,del(9)(p13},add(9)(q24} + 10,add(10)(p13} add(10)(p13) add(11)(p13),+add(11)(p15.5},del(11 }(p11.2) + 11,del(11 )(p11.2),add(11 )(p11.2) + 12,del(12)(p11.2)x2 del(12)(p11.2} +13 -13 +15 -14 add(16}(q24}x2 +15 add(17)(q25} + 16,add(16}(q24} +20,+20 -18 add(21 )(q22} -21,add(21 ):(q22) +9~19 mar +8~13 mar

For chromosome analysis, HAC15 and H295R cells were treated with colcemid for 2 h at 37 °C, followed by trypsin/EDTA treatment. Loose cells were subjected to hypotonic conditions for 15 minutes then fixed (3: 1 methanol: glacial acetic acid) and metaphase chromosomes stained with Trypsin/Giemsa stain .. The slides were analyzed under an

Olympus BX61 microscope and the images were captured with an Applied Imaging

Automated Karyotyping System. Abbreviations used in the table include: add (additional material of unknown origin-unidentifiable); del (deletion of part of a chromosome); -

(loss of an entire chromosome); + (gain of an entire chromosome); mar (marker chromosomes of unknown origin-unidentifiable}.

138 Figure Legends:

Fig. 17: Intracellular adrenocortical cell signaffng and mechanisms leading to adrenocortical steroid synthesis upon stimulation with the physiological agonists

Ang II, K+ and ACTH.

Fig. 18: Panel A: Phase contrast photomicrograph of cultured H295R cells, demonstrating celi growth attached patches of· cells. · Panel B: Phase contrast photomicrograph of cultured HAC15 cells, demonstrating cell growth as clumps or clusters of cells.

Fig. 19: Human adrenocortical steroid biosynthetic pathway.

Fig. 20: Development and isolation- of HAC clones with cylindrical cloning rings.

Fig. 21: Cytogenetic profiles depicted through karyotypes. Difference in chromosome size, position of centromeres, in total number of chromosomes and presence of ambiguous marker chromosomes were apparent when comparing the two adrenocortical cell lines. Panel A: Representative HAC15 karyotype and

Panel B: Representative of H295R karytoype.

Fig. 22: Murine adrenocortical steroid biosynthetic pathway including modifications seen in some mouse cell lines.

139 Fig. 17:

140 Fig,. 18:

141 Fig. 19:

Cholesterol

CYP11A1SIAR i HSD382 Pregnenolori Progesteto Demfycortloosteron~eoo~osteron,~ ~ Aldoste,ooo 1

Nomenclature for·the enzymes

DHEA StAR: ster;pldogenlc acute regulatory protein. p

142 Fig-. 20: .

Examine Select and circle viable Remove medium and place clones clones to be isolated rings over desired clones ...... l Harvest each clone in separate Trypsinize cells from within the flask rings •

143 Fig. 21:

A 12 A

144 Fig. 22:

20a- ~--1.. , 20ij,.­ ----i~. 11~20a­ ditiyd(oxypl;Oge.st_ero_lje· dihydroxyprogesterope

145 Ill. DISCUSSION

A. Specific aim I: CYP1182 Gene Regulation

283 The regulation of mineralocorticoid biosynthesis and secretion is complex • In the present investigation we sought to extend our understanding of the molecular mechanisms regulating the production of the principal mineralocorticoid- aldosterone. It is well documented that CYP1182 conducts the late rate-limiting

61 111 284 step in aldosterone biosynthesis • • • Since the CYP11B2 gene in humans and mice is predominantly expressed in the ZG of the adrenal cortex, we proposed to define the regions responsible for controlling in vivo ZG-specific expression. We demonstrated that a 985 bp promoter of the Cyp11 b2 gene is sufficient to target the adrenal ZG with expression of a Lacz reporter in mice. The

985 bp Cyp11 b2 promoter-driven Lacz was detected solely in the ZG of the

_adrenal cortex. Upon qPCR examination of 16 key mouse tissues, no aberrant extra-adrenal Lacz transcript was found. Hence, the major findings of this project are that Cyp11 b2 gene expression is controlled by its 985 bp 5' flanking promoter region and that the expression of the Cyp11 b2 gene is primarily confined to the adrenal cortex under normal physiological conditions in mice.

146 In terms of the history of ZG-specific targeting with the Cyp11 b2 promoter, others have previously utilized a bacterial artificial chromosome (BAC): containing, the entire Cyp11 b2 gene (unpublished data)- or a large segment (>5 kb} of the

176 Cyp11 b2 gene promoter • Morley et al generated 7 independent Tg lines with a construct harboring 5.4 kb of mouse Cyp 11 b2 5' flanking region and the lacZ reporter gene, which resulted in no, detectable lacZ expression. In contrast, we used less than a 1 kb region from the same gene to successfully target the adrenal ZG layer. These results suggest that the additional 4.4 kb region many contains further regulatory element(s) that have silenced or limited lacZ reporter expression in the 5.4 kb Tg mice. The additional 4.4 kb sequence, however, does not show any homology to the corresponding human CYP1182 region and, in fact, most of this sequence is repetitive. Because our NS adrenals show faint X­ gal staining that could go unnoticed without meticulous inspection, it is also possible that the 5.4 kb Tg mice .may have expressed low levels under NS condition which could have been amplified by the administration of LS diet. There is a substantial amount of evidence to support the contention that changes in dietary Na+ and K+ have a direct effect on Cyp11 b2 expression and serum aldosterone levels. Rats given a LS diet or a high K+ diet respond with a profound

285 287 increase in Cyp11 b2 levels - • In-vitro analyses using human· and rodent

57 58 141 288 adrenocortical cell lines have also shown matching results • • • • Our results complement these previous reports with robust increases in endogenous

Cyp11 b2 and Lacz mRNA, as well as serum aldosterone levels noted in the LS groups compared to NS control groups. To some extent, we owe our successful

147 characterization of the Tg mouse to the LS diet and its strong activation of the

RAAS.

It is noteworthy that_part of the success may be attributed to the SIB cassette that

we used as our reporter gene in the development of Tg mice. The SIB cassette

contains a splice donor / splice acceptor (SD/SA) sequence, which allows the

spliceosome to bring together neighboring exons and increase transgene

89 stability2 • The SIB cassette also includes an internal ribosomal entry site (IRES)

sequence, located between _-985 bp Cyp11 b2 promoter and the Lacz reporter

gene, which allows ribosomal subunits to bind our Tg mRNA independently of the

290 291 5'-cap initiation of translation, enhancing the stability of our construct • •

Lastly, the SIB cassette harbors a long poly A tail, which should further stabilize

the transgene in the mouse genome.

Several reports have shown targeting of the adrenal cortex utilizing other

163 172 177 182 adrenocortical-specific gene promoters • - • • Morley et al have also

utilized a 5.0- kb Cyp11 b1 promoter fragment that failed to target the adrenal

177 gland • In another study, however, the same group successfully targeted

adrenal cortex with a 6.4 kb regulatory fragment of the Cyp21 promoter. This

Cyp21 promoter driven Tg construct exhibited scattered-inconsistent targeting

thro~ghout the adrenal cortex and hence ·no further analyses have been

182 conducted using this strategy • Martinez et al. reported adrenal targeting by

17 174 - using regulatory regions from aldo-keto reductase 1B7 (Akr1 b7) ~· • The

148 Akr1 b7 Tg construct contained 5'-flanking and intragenic sequences of the

Akr1 b7 gene which targeted vas deferens and adrenal glands in the reported Tg mice.

Adrenal targeting has allowed examination of genes believed to control adrenal development and function. The transcriptional coactivator beta-catenin, along with SF-1, reportedly regulates Wnt family members, which play a crucial role in the development and maintenance of the adrenal gland. The role of beta-catenin as part of the Wnt signaling pathway has been assessed through the use of Tg mice in terms of both embryonic development of the . adrenal cortex and in

163 172 183 maintenance of the adult organ • • • Hammer et a.I. also utilized SF-1/Cre Tg mice to conditionally inactivate beta-catenin alleles and showed adrenal aplasia in newborn Tg mice. Although adrenocortical-specific, all of the above mentioned

Tg strategies have limited efficacy since they also permit extra-adrenal expression. Our current 985 bp Cyp11 b2 sequence is unique and distinct as it induces transgene expression selectively and exclusively in the adrenal ZG layer.

Non-steroidogenic gene promoters have also been .used to target the adrenal gland. Jorgez et al. utilized 6 kbp of the promoter of mouse transforming growth factor ·~ superfam1ly member inhibin a subunit promoter, which targeted the

184 gonads, pituitary, and the adrenal gland • The 6 kbp a subunit promoter-driven

Cre construct targeted both the adrenal cortex and medulla in three separate Tg lines and only the medulla in two other Tg lines. Hence, these animals may be

149' useful for recombination of flexed alleles, specif~cally in the adrenal medulla. Tg studies have also shown telomerase function in adrenocortical physiology, as

292 telomeres reportedly operate to replenish the cells within the adrenal cortex -

294. While important for their respective analyses and applicable to adrenal cortex, these Tg studies also allowed extra-adrenal expression. Hence, any further genetic manipulation studies utilizing these strategies would affect not only the adrenal cortex, but also all other cell types that are targeted.

Nevertheless, since our transgene solely targeted adrenal ZG cells, it will allow tissue-specific downstream operations such as ZG-specific knock-in and_ knock­ out.

A substantial amount of evidence supports the contention -that low levels of

Cyp11 b2 gene are expressed in extra-adrenal tissues such as the heart, brain,

187 195 295 304 and vasculature - • - • The function of locally produced extra-adrenal aldosterone remains unclear; however, it is well understood that cardiovascular­ and CNS-derived aldosterone does not contribute to the. total pool of circulating aldosterone. Nonetheless, in order to assess the extra-adrenal expression of the endogenous Cyp11 b2 gene, as well as our transgene, we -investigated the ex-pression profile of Cyp11 b2 in 16 key organs in mice. As we anticipated, exceptionally low levels Cyp11 b2 mRNA in most of the extra-adrenal tissues, we therefore, designed primer sets that were highly specific and sensitive to extremely low mRNA lev~ls. We began by designing a DNA template for

Cyp11 b2 that was identical to the target mRNA sequences _to be amplified.

150 Previous qPCR methods measured product relative to the adrenal Cyp11 b2 mRNA levets or relative·to a housekeeping gene, such as 18S ribosomal RNA; however, by using known quantities of the DNA transcripts as standards, we were able to express the number of Cyp11b2 mRNA copies in absolute terms.

Expressing mRNA levels in terms of copy number provides sustained reproducibility and permits quantification of the low levels of Cyp11 b2 mRNA present in extra-adrenal tissues. In order to eliminate concerns regarding cross­ reactivity between the two isozymes, we tested ·our. in-house designed primer and probe sets for Cyp11 b1 and Cyp11 b2 against their respective transcripts and found little or no cross-reactivity.

Under a NS diet, we detected Cyp11 b2 transcript in all organs tested, however, the levels of mRNA in extra-adrenal organs were extremely low compared to the adrenal gland. Intriguingly, a LS diet induced Cyp11 b2 mRNA up-regulation in thoracic aorta, thymus, and kidney, although their fold changes were small compared to the adrenal glands. While this extra-adrenal Cyp11 b2 expression may have little impact on systemic plasma aldosterone levels, it may activate the local aldosterone-mineralocorticoid receptor system, and hence it should not be overlooked. Nonetheless, qPCR analysis of transgene showed that our Tg mice have no detectable reporter gene expression in any of the extra-adrenal tissues tested under NS or LS diet. This phenomenon may be due to the limitations in our qPCR assay sensitivity as the NS adrenals in Tg mice showed minimal transgene expression. However, even under a LS diet none of the extra-adrenal

151 organs exhibited transgene expression. Further analyses under various pathological conditions are necessary to shed more light on this issue.

Subsequently, because transgene expression was solely localized to the adrenal glands, even under a LS diet, we investigated the expression of the 13- galactosidase reporter protein. X-gal staining was performed for Tg and wild-type littermate mice on all extra-adrenal tissues aforementioned, on LS and NS diets.

Besides the apparent (3-galactosidase staining in the Tg adrenals, as compared to the wild-type, we found no specific staining in any other tissue. The uniform 13- galactosidase staining seen in the Tg whole mount adrenals and immunohistochemistry (IHC) slides did not appear to be due to artifacts of endogenous lacZ staining because we eliminated that possibility. As described by Bolon et al., the complexity involved in background (3-galactosidase staining

305 was taken into consideration • X-gal staining time points including 2 h, 4 h, 6 h,

8 h, 10 h, 12 h, 14 h, and overnight were assessed utilizing Tg and wild-type control adrenals on LS and NS diets. Little or no background staining was observed up un~il 8 h of X-gal staining. Hence, for all the mounting and IHC experiments, we assigned 8 h staining time as the standard.

185 186 In agreement with pervious reports • , a LS diet in both wild-type C57BU6 and Tg mice triggered ZG cell proliferation. The ZG layer expanded from 3-4 layers to a 12-15 layered zone. ZG cells under LS conditions began to proliferate and migrate down towards the ZF, which has a distinctively straight-cords-like

152 _ appearance. Co-staining experiments, consisting of X-gal staining, followed by

primary Cyp11 b2 antibody staining of the Tg adrenocortical tissue sections, show·

ZG-specific co-localization of both endogenous Cyp 11 b2 and '3-galactosidase

proteins under NS ~nd LS diets. Although both proteins were co-localized in ZG

layer in LS adrenals, there was a slight difference in their staining patterns.

Within the expanded the ZG layer, the X-gal staining was more prominent on the

outer (capsule) side, whereas the Cyp11 b2 immunostaining was more intense on

the inner (medulla) side· of the ZG. This difference may be explaine.d by the

current hypotheses that adrenocortical stem cells in the capsule or sub-capsular

175 proliferate, differentiate, and move inward to generate adrenal cortical cells •

We believe that upon a LS diet administration, the ZG cells start inward

proliferation and simultaneous activation of Cyp11 b2 promoter. This would lead

to Cyp11 b2 and '3-galactosidase protein production throughout the· ZG layer,

which would result in increased aldosterone production and intense X~gal

staining, respectively. Because Cyp11 b2 enzyme is required for aldosterone

synthesis it would presumably be stablilized in the rapidly dividing ZG cells as

compared to ~-galactosidase protein. Hence we observe the accumulated

Cyp11 b2 enzyme but not the ~-galactosidase protein in the inner-most ZG or

outer ZF cells.

Cumulatively, this suggests that the 985 bp consensus promoter fragment

exclusively targets the ZG and responds to RAAS activation; however, regions

required for extra-adrenal expression of Cyp11 b2 are not present in this promoter

153 fragment. This phenomenon is consistent with the literature, as there are abundant reports that show varied lengt~s of gene promoters capable of driving·

164 165 306 308 tissue and cell-specific expression of transgenes • • - • Alternatively, we must acknowledge that our transgene does not exactly emulate the endogenous

Cyp11 b2 gene, as it is solely expressed in the ZG cells. Perhaps other regulatory regions exist outside of 985 bp promoter region or within the Cyp11 b2 gene that allow its expression in extra-adrenal organs. Indeed, the 985 bp promoter of

Cyp11 b2 has experimental limitations due to the absence of appropriate enhancers that would allow us to study Cyp11 b2 gene expression as it occurs normally.

Conversely however, this unique promoter fragment will allow several possibilities in terms of ZG-specific genetic manipulations. The characterization of this sequence should facilitate further experiments designed to specifically activate or sitence desired genes linked to human adrenocortical diseases, such as hyperaldosteronism. Conditional Cre/loxP system could be utilized to understand adrenocortical zonation . especially during development and throughout adulthood. Nevertheless, this regulatory fragment will be an invaluable tool for the advancement of res.earch associated with adrenal development, physiology, and pathology.·

154 B. Specific aim II: A Paucity of Adrenal Cell Lines

The purpose of this study was to develop a well-characterized human adrenocortical cell line that would be appropriate for many in vitro analyses.

Previously Gazdar et al. demonstrated that an adrenocortical cell line could be established from a heterogeneous population of cells from an adrenocortical carcinoma. Our study confirms-the work of Gazdar and colleagues, as a human adrenocortical carcinoma was utilized in development of the HAC15 cell line. The major findings of this project are that carcinomas provide a viable population of immortalized cells and that these cells can be utilized in the development of cell lines. Data from our current study indicate that a mixed p9pulation of adrenocortical carcinomas could be subjected to a series of cloning experiments in the development of cell lines from a single-cell origin. Moreover, along with

HAC15, several other clones have been isolated from carcinomas. Although the

HAC clones share the same origin, the adrenocortical steroidogenesis and responsiveness to physiological agonists by each clone is quite different.

According to our numerous repeated analyses, the most dominant clone overall, exhibiting the best steroidogenesis and the best responsiveness to physiological and pharmacological agonists proves to be HAC15.

From a clinical standpoint, our collaborative work with St. Jude's Children's

Research Hospital (SJCH} (Zambetti laboratory) has allowed the assessment of chemotherapy agents on HAC15 cells in culture. In addition, at SJCH, HAC15

155 cells have been injected into immunodeficient mice, which led to tumor formation,

allowing exploration .of the efficacy of chemotherapy agents in vivo~ Moreover,

HAC15 gene expression profiling via DNA microarray analysis confirmed high

309 310 expression of marker genes to be expressed in adrenal carcinomas • •

Collectively, results from the HAC15 cell line suggest it to be a good· tool for

studying chemotherapy ·agents both in vitro and in vivo and perhaps changes

· associated with carcinomas.

As aforementioned, HAC15 cells were developed from a naturally occurring

carcinoma, and these cells have been growing steadily in standard culture

conditions for over two years. Tumors, naturally occurring or experimentally

generated, have been utilized to characterize and develop viable adrenal cell

lines. Several cell lines of different origins, cell type- and organ-specific, have

been established that are extremely useful for in vitro analyses. Hela cells were

the first immortalized human cells derived from a cervical carcinoma. Since its

establishment, the Hela cell line has been a great resource for the scientific

community, as it represented the prototype for the establishment of stable

311 immortalized cell lines • Moreover, recent advances in cell line development

have revealed other techniques for the isolation of immortalized cells by ·

utilization of oncogenesis and Tg animals to artificially induce tumors. There have

been a number of rodent adrenal cell lines developed using Tg mice with

82 176 196 281 adrenal-targeted oncogenes • • • • In terms of success rate for the

development of cell lines and their viability as cell culture models, the newer

156 techniques appear to be inferior to naturally occurring tumors. Because cell lines developed from oncogene simulated tumors have a tendency to reach senescence, as is the case with primary cultures. In this regard, our HAC15 cell line is superior . because it has been effectively psssaged for numerous generations and has entered crisis. Taking long-term steroidogenesis and hormonal response into consideration, artificially generated tumor-derived cell lines are also inferior, as they tend to lose responsiveness to agonists and synthesize less steroid hormones with time.

Evidence suggests that rarely occurring tumors may represent the best source for the development of cell lines. The Y1 mouse adrenal cell line originated from a tumor that presumably occurred due to an atomic blast. This cell line has been widely used for studying acute regulation of steroidogenesis, as well as the mechanisms controlling transcription of genes . encoding steroidogenic

210 220 312 313 enzymes • • • • The Y1 cell line has a number of positive attributes, including rapid growth, ACTH response, and easy trasfection with plasmid DNA.

Our HAC15 cells have a much slower doubling time as compared to the Y1 cells.

In terms of ACTH responsiveness, the youngest population of HAC15 cells exhibits an excellent and significant response to ACTH stimulation; however, after extended population doublings, the ACTH response tends to decrease. The lessening trend of ACTH responsiveness seen in HAC15 cells is not uncommon; the human adrenocortical H295R cells have also shown a similar trend.

157 Nevertheless, in our laboratory, we have also seen a time/passage-dependent decline in Y1 cell, responsiveness to ACTH_ stimulation. The reduction in response to ACTH by these cell lines is likely due to the down- regulation of its

83 205 314 receptor, melanocortin type 2 receptor (MC2R) • • • ACTH i$ a unique ligand for its receptor, as it has been shown to maintain the expression of MC2R in vivo

315 316 and in vitro • • Certainly, the lack of ACTH in culture media may somewhat explain the phenomenon of reduction in ACTH responsiveness exhibited by cell lines over time. The greatest drawback of the Y1 cells is their inability to produce

196 mineralocorticoids, glucocorticoids, or adrenal androgens •

Two other mouse adrenocortical cell lines, ATC1 and ATC7-L, have been

95 produced using genetically targeted oncogenesis • Both cell lines represent a ZF phenotype, which is manifested at multiple levels: ·corticosterone secretion,

ACTH responsiveness, and expression profile of specific genes. Corticosterone secretion is increased by ACTH in both cell lines in a dose-dependent manner; consequenUy, in this regard, these cells may prove superior to the Y1 cells. As with the Y1 cells, the ATC cells do not respond to Ang II nor do they secrete significant amounts of aldosterone. Hence, while these cell lines will be useful for certain adrenal research projects, they, like other mouse adrenal cell lines, will not be useful as a human model for most studies because they do not produce aldosterone, cortisol or adrenal androgens, as is seen in the H295R and HAC15 cell lines.

158 In terms of the availability of human adrenocortical cell lines, for several years,

84 13 the NCI-H295 and its substrains were the only option • t _ One substrain;

H295R, is immensely useful due the retention of Ang II- responsiveness. The

H295R cell line has also been ext~nsively utilized as a model to define the

mechanisms controlling transcription of the steroid-metabolizing genes. Promoter

constructs containing the 5'-flanking DNA from CYP11A 1, CYP11 B1, CYP11 B2,

57 81 CYP17, CYP21, and HSD3B2, have been studied ustng this strain • • Although

the H295R cell line express low levels of MC2R, this cell line _has been used by a

number of investigators to study regulation of receptor expression, as well as

206 207 acute actions of ACTH • • Along. with H295R, another substrain, designated

H295A, was developed from the original NCI-H295. This substrain has also

proven superior to the original NCI-H295, as it has been characterized and

81 199 shown to grow as a monolayer • • However, these cells reportedly lack Ang II

responsiveness.

Our study has put forth HAC15 cells as an alternative to the currently available

H295 cell models. As with the H295R cells, our recurrent characterization of

HAC15 cells is promising, as the cell line expresses all of the genes involved in

-the adrenocortical pathway, including StAR, CYP11A1, CYP17, CYP21,

HSD3B2, CYP1181, CYP1182, and SUL T2A 1. Upon stimulation with Ang II and

K+, an increase in aldosterone levels and the expression levels for all of ·the

enzymes involved in its production is observed. ACTH causes a modest, but

significant, increase in aldosterone, cortisol, and DHEA, along with enhanced

159 expression of all of the enzymes in their respective pathways. As with H295R,

HAC15- cells grow as attached cells; however, unlike H295R, they seem to· aggregate and grow as clusters of cells in a staggered-stacking pattern. HAC15 cells demonstrate structural features previously observed for the H295R cells, including the loss of contact inhibition, which leads to cells growing as attached clumps. It is noteworthy that the H295 cell models represent a mixture of cells isolated from primary cultures of the original tumor. On the other hand, HAC15 ceHs possess a single cell origin, as they have been derived from a series of

82 83 clonal experiments • •

A. considerable body of evidence now exists that supports the contention that gene · expression profiling through microarray analysis can assist in tumor

317 322 classification and cancer outcome prediction - • Over the past decade, genome-wide microarray analyses have been actively explored, as adrenocortical carcinomas have increasingly risen to clinical attention. We have assessed the role of the two available adrenocortical carcinoma-derived cell lines, HAC15 and H295R, in DNA microarray analysis. Overexpression in adrenocortical carcinomas of a number of genes that have been designated as biomarkers for malignancy or survival have been concurrently· identified in the

HAC15 and H295H cell lines. Further studies are needed in which objective measurements of genome-wide expression profiles are elucidated so that the associations found in this study, and in the · studies of others, can be substantiated.

160 We demonstrated that the HAC15 cells secrete . mineralocorticoids, glucocorticoids, and adrenal androgens and express all of the enzymes involved in the generation of these hormones. In addition, the levels of these enzymes are shown to increase upon treatment with ACTH, Ang II, and K+. In summary, we show that under conventional culture conditions, HAC15 cells exhibit hormonal responses, steroidogenesis, and expression of steroid-metabolizing enzymes.

This cell line represents only the second human adrenocortical cell model and the first that has retained significant ACTH responsiveness. HAC15 cells have thus proven to be a stable cell model for studies directed towards understanding physiological, as well as pathophysiological, adrenocortical functions.

161 IV. BIBLIOGRAPHY

-1. Colombo L, Dalla Valle L, Fiore C, Armanini D; Belvedere P. Aldosterone and the conquest of land. J Endocrinol Invest. Apr 2006;29(4):373-379. 2. Aburomia R, Khaner 0, Sidow A. Functional evolution in the ancestr~ lineage of vertebrates or when genomic complexity was wagging its morphological tail. J Struct Funct Genomics. 2003;3(1-4):45-52 .. 3. Bury NR, Sturm A. Evolution of the corticosteroid receptor signalling pathway in fish. Gen Comp Endocrinol. Aug-Sep 2007;153(1-3):47-56. 4. Nonaka Y, Tak:emori H,-Halder SK, et al. Frog cytochrome P-450 (11 beta,aldo), a single enzyme involved in the final steps of glucocorticoid and mineralocorticoid biosynthesis. Eur J Biochem. Apr 1 1995;229(1):249-256. 5. Bottner B, Denner K, Bernhardt R. Conferring aldosterone synthesis to human CYPl lB 1 by replacing key amino acid residues with CYPl lB~-specific ones. Eur J Biochem. Mar 15 1998;252(3):458-466. 6. Momet E, Dupont J, Vitek A, White PC. Characterization of two genes encoding human steroid 11 beta-hydroxylase (P-450(11) beta). J Biol Chem. 1989;264(35}:20961-20967. 7. Chua SC, Szabo P, Vitek A, Grzeschik KH, John M, White PC. Cloning of"cDNA encoding steroid 11 beta-hydroxylase (P450cll)-; Proc Natl Acad Sci U SA. 1987;84(20):7193-7197. 8. Kawamoto T, Mitsuuchi Y, Toda K, et al. Role of steroid 11 beta-hydroxylase and steroid 18-hydroxylase · in the biosynthesis of glucocorticoids and mineralocorticoids in humans. Proc Natl Acad Sci U S A. Feb 15 1992;89(4 ): 1458-1462. 9. Heitzmann D, Derand R, Jungbauer S, et al. Invalidation of TASKl potassium channels disrupts adrenal gland zonation and mineralocorticoid homeostasis. Embo J. Jan 9 2008;27(1):179-187. 10. Curnow KM, Tusie-Luna MT, Pascoe L, et al. The product of the CYP11B2 gene is required for aldosterone - biosynthesis in the human adrenal cortex. M ol Endocrinol. 1991;5(10):1513-1522. 11. Rainey WE. Adrenal zonation: clues from 11 beta-hydroxylase and aldosterone synthase. Mol Cell Endocrinol. 1999;151(1-2):151-160. -12. Bassett MH, Zhang Y, White PC, Rainey WE. Regulation of human CYP11B2 and CYPllBl: comparing the role of the common CRE/Adl element. Endocr Res. Nov 2000;26(4):941-951. 13. Kakiki M, Morohashi K, Nomura M, Omura T, Rorie T. Regulation of aldosterone synthase cytochrome P450 (CYP11B2) and 11 beta-hydroxylase cytochrome P450 (CYPl lBl) expression in rat adrenal zona glomerulosa cells by low sodium diet and angiotensin II receptor antagonists. Biol Pharm Bull. Sep 1997 ;20(9):962-968.

16-2 - 14. Rainey WE, White P. Functional adrenal zonation and regulation of aldosterone biosynthesis. Curr opin Endocrinology & Diabetes. 1998;5: 17 5-182. 15. Fallo F, Pezzi V, Barzon L, et al. Quantitative assessment of CYPllBl and CYP11B2 expression in aldosterone-producing adenomas. Eur J Endocrinol. 2002;147(6):795-802. 16. Pascoe L, Curnow KM, Slutsker L, et al. Glucocorticoid-suppressible hyperaldosteronism results from hybrid genes created by unequal crossovers between CYPllBl and CYP11B2. Proc Natl Acad Sci US A. 1992;89(17):8327- 8331. 17. Baker ME. Evolution of adrenal and sex steroid action in vertebrates: a ligand­ based mechanism for complexity. Bioessays. Apr 2003;25(4):396-400. 18. Barnes K, Hardisty MW. Ultrastructural and histochemical studies on the testis of the river lamprey, Lampetra fluviatilis (L.). J Endocrinol. Apr. 1972;53(1):59-69. 19. · Bridgham JT, · Carroll SM, Thornton Nv. Evolution of hormone-receptor complexity by molecular exploitation. Science. Apr 7 2006;312(5770):97-101. 20. Nunez S, Trant JM. Regulation of intei;renal gland steroidogenesis in the Atlantic stingray (Dasyatis sabina). J Exp Zool. Oct 1 1999;284(5):517~525. 21. Baker ME. Evolution of glucocorticoid and mineraloco_rticoid responses: go fish. Endocrinology. Oct 2003; 144(10):4223-4225. 22. Truscott B. Corticosteroids of the coelacanth Latimeria chalumnae Smith: a provisional study on their identity. Gen Comp Endocrinol. Jul 1980;41(3):287- 295. 23. Campbell CM, Postier A, J alabert B, Truscott B. Identification and quantification of steroids in the serum of rainbow trout during spermiation and oocyte maturation. J Endocrinol. Jun 1980;85(3):371-378. 24. Joss JM, King J A, Millar RP. Identification of the molecular forms of and steroid hormone response to gonadotropin-releasing hormone in the Australian lungfish, Neoceratodus forsteri. Gen Comp Endocrinol. Dec 1994;96(3):392-400. 25. J anssens PA, Vinson GP, Jones IC, Mosley W. Amphibian Characteristics of the Adrenal Cortex of the African Lungfish (Protopterus Sp.). J Endocrinol. Jul 1965;32:373-382. . 26. Shrimpton JM, McCormick SD. Regulation of gill cytosolic corticosteroid .receptors in juvenile Atlantic salmon: interaction effects of growth hormone with prolactin and triiodothyronine. Gen Comp Endocrinol. Nov 1998; 112(2):262-274. 27. Joss JM, Itahara Y, Watanabe TX, Nakajima K, Takei Y. Teleost-type angiotensin is present in Australian lungfish, Neoceratodus forsteri. Gen Comp Endocrinol. May 1999;114(2):206-212. 28. Jones IC, Phillips JG, Bellamy D. The adrenal cortex throughout the vertebrates. Br Med Bull. May 1962;18:110-114. 29. Phillips JG, Jones IC, Bellamy D~ Biosynthesis of adrenocortical hormones by adrenal glands of lizards and snakes. J Endocrinol. Oct 1962;25 :233-237. 30. Devuyst 0, Beaujean V, Crabbe J. Aldosterone interaction on sodium transport and chloride permeability: influence of epithelial structure. Biochim Biophys Acta. May 4 1995;1235(2):443-451.

163 31. Gaeggeler HP, Duperrex H, Hautier S, Rossier BC. Corticosterone induces 11 beta-HSD and mineralocorticoid specificity_in an amphibian urinary bladder cell line. Am J Physiol. Feb 1993;264(2 Pt 1):C317-322. 32. Dauphin-Villemant C, Leboulenger F, Xavier F, Vaudry H. Interrenal activity in the female lizard Lacerta vivipara J.: in vitro response to ACTH 1-39 and to [Sarl, Val5] angiotensin II (ANG II). J Steroid Biochem. 1988;30(1-6):457-460. 33. Rosenberg J, Hurwitz S. Concentration of adrenocortical hormones in relation to cation homeostasis in birds. Am J Physiol. Jul 1987;253(1 Pt 2):R20-24. 34. Zhou MY, Gomez-Sanchez EP, Foecking MF, Gomez-Sanchez CE. Cloning and expression of the rat adrenal cytochrome P-450 11B3 (CYP11B3} enzyme cDNA: preferential 18-hydroxylation over 11 beta-hydroxylation of DOC. Mol Cell Endocrinol. Oct 30 1995;114(1-2):137-145. 35. Borroel M, Borroel P, Damasco MC, Lantos CP. The in vitro investigation of four radioactive corticosteroids in the Antarctic seal (leptonychotes weddellii). Gen Comp Endocrinol. Jan 1974;22(1):1-12. 36. Carballeira A, Brown "JW, Fishman LM, Trujillo D, Odell DK. The adrenal gland of stranded whales (Kogia breviceps and Mesoplodon europaeus): morphology, hormonal contents, and biosynthesis of corticosteroids. Gen Comp Endocrinol. Nov 1987;68(2):293-303. · 37. Hardisty MW. Quantitative and experimental studies on the interrenal tissues of the upstream migrant stage of the river lamprey, Lampetra fluviatilis L. Gen Comp Endocrinol. Jun 1972;18(3):501-514. 38. Manire CA, Rasmussen LE, Maruska KP, Tricas TC. Sex, seasonal, and stress­ related variations in elasmobranch corticosterone concentrations. Comp Biochem Physiol A Mol Integr Physiol. Dec 2007;148(4):926-935. 39. Butler DG, Youson JH.· Steroidogenesis in the yellow corpuscles (adrenocortical homolog) in a holostean fish, the bowfin, Amia calva L. Gen Comp EndocrinoL Jul 1986;63(1):45-50. 40. Y ouson JH. Effects of mammalian corticotrophin on the ultrastructure of presumptive interrenal cells in the opisthonephros of the lamprey, Petromyzon marinus L. Am J Anat. Oct 1973;138(2):235-251. 41. al-Mahrouki AA, Youson JH. Immunohistochemical studies of the endocrine cells within the gastro-entero-pancreatic system of Osteoglossomorpha, an ancient teleostean group. Gen Comp Endocrinol. May 1998;110(2):125-139. 42. Nishimura H. Physiological evolution of the renin-angiotensin system. Jpn Heart J. Sep 1978;19(5):806-822. 43. Arnold J. Bin Beitrag zu der feiner Struktur und dem Chemismus der Nebennieren. Virchows Arch. 1866;35:64-107. 44. Simpson ER WM. Regulation of the synthesis of steroidogenic enzymes in adrenal cortical cells by ACTH. Annu Rev Physiol. . 1988;1988;50:427-40. 45. Yabu M, Senda T, Nonaka Y, Matsuka~a N, Okamoto M, Fujita H. Localization of the gene transcripts of 11 beta-hydroxylase and aldosterone synthase in the rat adrenal cortex by in situ hybridization. Histochemistry. 1991;96(5):391-394. 46. McDonald SE, Henderson TA, Gomez-Sanchez CE, Critchley HO, Mason JI. 1 lBeta-hydroxysteroid dehydrogenases in human endometrium. Mol Cell Endocrinol. Mar 27 2006;248(1-2):72-78.

164 47. Domalik LJ, Chaplin DD, Kirkman MS, et al. Different isozymes of mouse 11 beta-hydroxylase produce mineralocorticoids and glucocorticoids. Mol Endocrinol. 1991;5(12):1853-1861. 48. Yanagibashi K, Shackleton CH, Hall PF. Conversion of 11-deoxy.corticosterone and corticosterone to aldosterone. by cytochrome P-450 11 beta-/18-hydroxylase from porcine adrenal. J Steroid Biochem. Jun 1988;29(6):665-675. 49. Hashimoto T, Morohashi K, Omura T. Cloning and characterization of bovine cytochrome P450(1 la) genes. J Biochem. 1989-;105:676-679. 50. Mathew PA, Mason JI, Trant JM, Sanders D, Waterman MR. Amino acid substitutions phe66----leu and ser126----pro abolish cortisol and aldosterone synthesis. by bovine cytochrome p450(1l)beta. J Biological Chem. 1990;265(33):20228-20233. 51. Kirita S, Hashimoto T, Kitajima M, Honda S, Morohashi K, Omura T. Structural analysis of multiple bovine P450(1 la) genes and their promoter activities. J Biol Chem. 1990;108:1030-1041. · 52. Honda S, Morohashi K, Omura T. Novel cAMP regulatory elements in the promoter region of bovine P450(1la) gene. J Bzochem. 1990;108:1042-1049. 53. Oertle M, Muller J. Two types of cytochrome P-450(11 beta) in rat adrenals: separate regulation of gene expression. Mol Cell Endocrinol. 1993;91(1-2):201- 209. 54. Bassett MH, Zhang Y, Clyne C, White PC, Rainey WE. Differential regulation of aldosterone synthase and 11 beta-hydroxylase transcription by steroidogenic factor-1. J Mol Endocrinol. Apr 2002;28(2): 125-135. 55. Bassett MH, Suzuki T, Sasano H, White PC, Rainey WE. The NGFIB family of nuclear receptors regulates adrenal aldosterone production. Mol Endocrinol. 2003. 56. Wang XL, Bassett M, Zhang Y, et al. Transcriptional regulation of human 1 lbeta­ hydroxylase (hCYPl lBl). Endocrinology. 2000;141(10):3587-3594. 57. Clyne CD, Zhang Y, Slutsker L, Mathis JM, White PC, Rainey WE. Angiotensin II and potassium regulate human CYP11B2 transcription through common cis­ elements. Mol Endocrinol. 1997;11(5):638-649. 58. Clyne CD, White PC, Rainey WE. Calcium regulates human CYP11B2 transcription. Endocr Res. 1996;22(4 ):485-492. 59. Coulombe~' Lefebvre A, LeHoux JG. Characterization of the hamster CYP11B2 gene regulatory regions~ Endocr Res. Nov 1996~22(4):653-661. 60. Shibata H, Kobayashi S, Kurihara I, et al. COUP-TF and transcriptional co­ regulators in adrenal steroidogenesis. Endocr Res. Nov 2004;30(4):795-801. 61. Ye P, Nakamura Y, Lalli E, Rainey WE. Differential effects of high and low steroidogenic factor-1 expression on CYP11B2 expression and aldosterone production in adrenocortical cells. Endocrinology. Mar 2009; 150(3): 1303-1309. 62. Bassett MH, Suzuki T, Sasano H, White PC, Rainey WE. The orphan nuclear receptors NURRl and NGFIB regulate adrenal aldosterone production. Mol Endocrinol. Feb 2004;18(2):279-290. 63. Taymans SE, Pack S, Pak E, Torp-y DJ, Zhuang Z, Stratakis CA. Human CYP11B2 (aldosterone synthase) maps to chromosome 8q24.3. J Clin Endocririol Metab~ Mar 1998;83(3):1033-1036.

165 64. White PC. Inherited forms of mineralocorticoid hypertension. Hypertension. Dec ·1996;28( 6):927-936. 65. Carron MC, Palsdottir A, Belt KT, Porter RR. Deletion of complement C4 and steroid 21-hydroxylase genes in the HLA class· Ill region . .Embo J. Oct 1985;4( 10):2547:.. 2552. 66. Davies LA, Hu C, Guagliardn NA, et al. TASK channel d~letion_ in mice causes primary hyperaldosteronism. Proc Natl Acad Sci U S · A. Feb 12 2008; 105(6):2203-2208. 67. Inglis GC, Ingram MC, Holloway CD, et al. Familial pattern of cort;icosteroids and their metabolism in adult human subjects--the Scottish Adult Twin Study. J Clin Endocrinol Metab. Nov 1999;84(11):4132-4137. 68. de Simone G, Tommaselli AP, Rossi R, et al. Partial deficiency of adrenal 11- hydroxylase. A possible cause of primary hypertension. Hypertension. Mar-Apr 1985;7 (2):204-210. 69. Honda M, Nowaczynski W, Guthrie GP, Jr., et al. _Response of several adrenal steroids to ACTH stimulation in essential hypertension. J Clin Endocrinol Metab. Feb 1977;44(2):264-272. 70. Takeda Y, Furukawa K, Inaba S, Miyamori I, Mabuchi H. Genetic analysis of aldosterone synthase in patients with idiopathic hyperaldosteronism. J Clin Endocrinol Metab. May 1999;84(5):1633-1637. 71. Davies E, Holloway CD, Ingram MC, et al. Aldosterone excretion rate and blood pressure in essential hypertension are related to polymorphic differences in the aldosterone synthase gene CYP11B2. Hypertension. 1999;33((2)):703-707. 72. · Paillard F, Chansel D, Brand E, et al. Genotype-phenotype relationships for the renin-angiotensin-aldosterone system in a normal population. Hypertension. Sep l 999;34(3):423-429. 73. Lim PO, Macdonald TM, Holloway C, et al. Variation at the aldosterone synthase (CYP11B2) locus contributes · to hypertension in subjects with a raised aldosterone-to-renin ratio. J Clin Endocrinol M etab. Sep· 2002;87 (9):4398-4402. 74. Nicod J, Bruhin D, Auer L, Vogt B, Frey FJ, Ferrari P. A biallelic gene polymorphism of CYP11B2 predicts increased aldosterone to renin ratio in selected ·hypertensive patients. J Clin Endocrinol Metab~ Jun 2003;88(6):2495- 2500. 75. Morley SD, Viard I, Chung BC, Ikeda Y, Parker KL, Mullins JJ. Variegated expression of a mouse steroid 21-hydroxylase/beta- galactosidase transgene suggests centripetal migration of adrenocortical cells. Mol Endocrinol. 1996; 10(5):585-598. 76. Morley SD, Viard I, Parker KL, Mullins JJ. Adrenocortical-specific transgene expression directed by steroid hydroxylase gene promoters. Endocr Res. 1996;22(4 ): 631-639. 77. Cardoso CC, Bornstein SR, Hornsby PJ. New methods for investigating experimental human adrenal tumorigenesis. Mol Cell Endocrinol. Mar 5 2009;300( 1-2): 17 5-179. 78. O'Hare MJ, Neville AM. Morphological responses to corticotrophin and cyclic AMP by adult rat adrenocortical cells in monolayer culture. Journal of Endocrinology. 1973;56(3):529-536.

166 79. Gospodarowicz D, 11 CR, Hornsby PJ, Gill GN. Control of bovine adrenal cortical cell proliferation by_ fibroblast growth factor. Lack of effect of epidermal growth factor. Endocrinology. 1977;100(4):1080-1089. · 80. Yasumura Y, Buonassisi V, Sato G. Clonal analysis of differentiated function in animal cell cultures. I. Possible correlated maintenance of differentiated function and the diploid karyotype. Cancer Res. 1966;26(3):529-535. 81. Rogriquez H, Hum DW, Staels B, Miller WL. Transcription of the human genes for cytochrome P450scc and P450cl 7 is regulated differently in human adrenal NCI-H295 cells than in mouse . adrenal Yl cells. J Endocrinol Metab. 1997 ;82(2):365-371. 82. Parmar J, Rainey WE. Comparisons of adrenocortical cell lines as in vitro test systems. Adrenal toxicology book.. 2008; 11 :in press. 83. Parmar J, Key · RE, Rainey WE. Development of an adrenocorticotropin­ responsive human adrenocortical carcinoma cell line. J Clin Endocrinol Metab. Nov 2008;93(11):4542-4546. 84c Gazdar AF, Oie HK, Shackleton CH, et al. Establishment and characterization of a human adrenocortical carcinoma cell line that expresses multiple pathways of steroid biosynthesis. Cancer Res. Sep 1 1990;50(17):5488-5496. 85. Auersperg N, MacLaren IA, Bissell MJ. V-K-ras transformation induces reversion to an earlier developmental form in adult rat adrenal cells. Differentiation. 1990;43( 1}:29-36. 86. Pan J, Roskelley CD, Auersperg N. Influence of cell type on the steroidogenic potential and basal cyclic AMP levels of ras-oncogene-transformed rat cells. Differentiation. 1995;58(5):321-328. 87. Mellon SH, Miller WL, Bair SR, Moore CC, Vigne JL, Weiner RI. Steroidogenic adrenocortical cell lines produced by genetically targeted tumorigenesis in transgenic mice. Mol Endocrinol. 1994;8(1):97-108. 88. Mukai K, Nagasawa H, Agake-Suzuki R, et al. Conditionally immortalized adrenocortical cell lines at undifferentiated states exhibit inducible expression of glucocorticoid-synthesizing genes. Eur J Biochem. 2002;269(1 ):69-81. 89. Alberts B, Johnson. A, Lewis J, Raff M, Roberts K, Walter P. Isolating Cells and , Growing Them in Culture. Vol Fifth Edition London: Taylor and francis; 2002. 90. Ham RG. Clonal Growth of Mammalian Cells in a Chemically Defined, Synthetic Medium. Proc Natl Acad Sci USA. Feb 1965;53:288-293. 91. Avery RJ, Norton JD, Jones JS, Burke DC, Morris AG. Interferon inhibits transformation by murine sarcoma viruses before integration of provirus. Nature. Nov 6 1980;288(5786):93-95. 92. Auersperg N, Wan MW, Sanderson RA, Wong KS, Mauldin D. Morphological and functional differentiation of Kirsten murine sarcoma virus;_transformed rat adrenocortical cell lines. Cancer Res. 1981;41 1763-1771. 93. Auersperg N. Effects of culture conditions on the growth and differentiation of transformed rat adrenocortical cells. Cancer Res. 1978;38 1872-1884. 94. Auersperg N, Hudson JB, Goddard EG, Klement V. Transformation of cultured rat adrenocortical cells by Kirsten murine sarcoma virus (Ki-MSV). Int J Cancer. 1977;19:81-89.

167 95. Ragazzon B, Lefrancois-Martinez AM, Val P, et al. Adrenocorticotropin­ dependent changes in SF-l/DAX-1 ratio influence steroidogenic genes expression in a novel model of glucocorticoid-producing adrenocortical- cell lines derived' from targeted tumorigenesis. Endocrinology. Apr 2006;147(4):1805-1818. 96. Compagnone NA, Bair SR, Mellon SH. Characterization of adrenocortical cell lines produced by genetically targeted tumorigenesis in transgenic mice. Steroids. 1997;62:238-243-. 97. Kananen K, Markkula M, Mikola M, Rainio EM, .McNeilly A, Huhtaniemi I. Gonadectomy permits adrenocortical tumorigenesis in mice transgenic for the mouse inhibin alpha-subunit promoter/simian virus 40 T-antigen fusion gene: evidence for negative autoregulation of the inhibirt alpha-subunit gene. Mol Endocrinol. 1996; 10 1667. 98. Rilianawati, Paukku T, Kero J, et al. Direct luteinizing hormone action triggers adrenocortical tumorigenesis in castrated mice transgenic for the murine inhibin alpha-subunit promoter/simian virus 40 T-antigen fusion gene. Mol Endocrinol. 1998; 12(6): 801-809. 99. Villee DB. The development of steroidogenesis. Am J Med.- Nov 1972;53(5):533- 544. 100. Deshpande N, Jensen V, Bulbrook RD, Doouss TW. In vivo steroidogenesis by the_human adrenal gland. Steroids. Apr 1967;9(4):393-404. 101. · Lanman JT, Silverman LM. In vitro steroidogenesis in the human neonatal adrenal gland, including observations on human adult and monkey adrenal glands. Endocrinology. Apr 1957;60(4):433-445. 102. Bird IM, Mathis JM, Mason JI, Rainey WE. Ca(2+)-regulated expression of steroid hydroxylases in H295R human adrenocortical cells. Endocrinology. Dec 1995; 136(12):5677~5684. 103. Bird IM, Mason JI, Oka K, Rainey WE. Angiotensin-II stimulates an increase in cAMP and expression of 17 alpha-hydroxylase cytochrome P450 in fetal bovine adrenocortical cells. Endocrinology. 1993;132(2):932--934. · 104. Bird IM, Mason JI, Rainey WE. Regulation of type 1 angiotensin II receptor messenger ribonucleic acid expression in human adrenocortical carcinoma H295 cells. Endocrinology. 1994;134(6):2468-2474. 105. Bird IM, Hanley_ NA, Word RA, et al. Human -NCI-H295 adrenocortical carcinoma cells: a model for angiotensin-II-responsive aldosterone secretion. Endocrinology. Oct 1993;133(4).: 1555-1561. 106. Romero DG, Plonczynski MW, Gomez-Sanchez EP, Yanes LL, Gomez-Sanchez CE. RGS2 is regulated by angiotensin II and functions as a negative feedback of aldosterone production in H295R human adrenocortical cells. Endocrinology. Aug 2006;147(8):3889-3897. 107. Inagaki K, Otsuka F, Suzuki J, et al. Regulatory expression of bone morphogenetic protein-6 system in aldosterone prodllction by human adrenocortical cells. Regul Pept. Feb 1 2007;138(2-3):133-140. 108. Shah BH, Baukal AJ, Chen HD, Shah AB, Catt KJ. Mechanisms of endothelin-1- induced MAP kinase activation in adrenal glomerulosa cells. J Steroid Biochem · Mol Biol. Dec 2006;102(1-5):79-88.

168 109. Otani H, Otsuka F, Inagaki K, et al. Aldosterone breakthrough caused by chronic blockage of angiotensin II type 1 receptors in human adrenocortical cells: possible involvement of bone morphogenetic protein-6 actions. Endocrinology. Jun 2008; 149(6):2816-2825. 110~ Nogueira EF, Vargas CA, Otis M, Gallo-Payet N, Bollag WB, Rainey WE. Angiotensin-II acute regulation of rapid response genes in human, bovine, and rat adrenocortical cells. J Mol Endocrinol. Dec 2007;39(6):365-374. 111. Nogueira EF, Xing Y, Morris CA, Rainey WE. Role of angiotensin II-induced rapid response genes in the regulation of enzymes needed for aldosterone synthesis. J MolEndocrinol. Apr 2009;42(4}:319~330. 112. Rainey WE, Bird IM, Byrd W, Carr BR. Effect of angiotensin II on human . luteinized granulosa cells. Fertil Steril. 1993;59(1):143-147. 113. Rainey WE, Bird IM, Mason n. The NCI-H295 cell line: a pluripotent model for human adrenocortical studies. Mol Cell Endocrinol. Apr 1994;100(1-2):45-50. 114. Bird IM, Word RA, Clyne C, Mason JI, Rainey WE. Potassium negatively regulates angiotensin II type 1 receptor expression in human adrenocortical H295R cells. Hypertension. 1995;25(6):1129-1134. 115. Bird IM, Pasquarette MM, Rainey WE, Mason JI. Differential control of 17 alpha-hydroxylase and 3 beta-hydroxysteroid dehydrogenase expression in human adrenocortical H295R cells. Journal of Clinical Endocrinology Metabolism. _1996;81(6):2171-2178. 116. Denner K, Rainey WE, Pezzi V, Bird IM, Bernhardt R, Mathis JM. Differential regulation of 11 beta-hydroxylase and aldosterone synthase in human adrenocortical H295R cells. Mol Cell Endocrinol. 1996;121(1):87-91. 117. Clyne CD, Zhang Y, Slutsker L, Mathis JM, White PC, Rainey WE. Angiotensin II and potassium regulate human CYP11B2 transcription through common cis­ elements. Mol Endocrinol. May 1997; 11(5):638-649. 118. Leers-Sucheta S, Morohashi K, Mason JI, Melner MH. Synergistic activation of the human type II 3a-hydroxysteroid dehydrogenase/delta 5 - delta 4 isomerase promoter by the transcription factor steroidogenic factor-1/adrenal 4-binding protein and phorbol ester. J Biol Chem. 1997;272(12):7960-7967. 119. Gizard F, Lavallee B, DeWitte F, Teissier E, Staels B, Hum DW. The transcriptional regulating protein of 132 kDa (TReP-132) enhances P450scc gene transcription through interaction with steroidogenic factor-1 in human adrenal cells. J Biol Chem. Oct 18 2002;277(42):39144-39155. · 120. Vilain E, Guo W, Zhang YH, McCabe ER. DAXl gene expression upregulated by steroidogenic factor 1 in an adrenocortical carcinoma cell line. Biochem Mol Med. Jun 1997;61(1):l-8. 121. Guo W, Burris TP, McCabe ER. Expression of DAX-1, the gene responsible for X-linked adrenal hypoplasia congenita and hypogonadotropic hypogonadism, in the hypothalamic-pituitary-adrenal/gonadal axis. Biochem Mol Med. Oct 1995;56(1):&-13. 122. Sugawara T, Sakuragi N, Minakami H. CREM confers cAMP responsiveness in human steroidogenic acute regulatory protein expression in NCI-H295R cells rather than SF-1/Ad4BP. J Endocrinol. Oct 2006;191(1):327-337.

169 123. Romero DG, Vergara GR, Zhu Z, et al. Interleukin-8 synthesis, regulation, and steroidogenic role in H295R human adrenocortical cells. Endocrinology. Feb 2006; 147(2):891-898. 124. Burton TJ, Cope G, Wang J, et al. Expression of the· epithelial Na(+) channel and other components of an aldosterone response pathway in human adrenocortical cells. Eur J Pharmacol. Apr 13 2009. 125. Isaka T, Ikeda K, Takada Y, Inada Y, Tojo K, Tajima N. Azelnidipine inhibits aldosterone synthesis and secretion in human adrenocortical cell line NCI-H295R. Eur J Pharmacol. Mar 1 2009;605(1-3):49-52. 126. Qin H, Kent P, Isales C, Parker P, Wilson M, Bollag W. The Role of Calcium Influx Pathways in Phospholipase D Activation in Bovine Adrenal Glomerulosa Cells. J Endocrinol. Apr 16 2009. 127. Rainey WE, Bird IM, Sawetawan C, et · al. Regulation of human adrenal carcinoma cell (NCI-H295) production of C19 steroids. J Clin Endocrinol Metab. 1993;77(3):731-737. 128. Romero DG, Welsh BL, Gomez-Sanchez EP, Yanes LL, Rilli S, Gomez-Sanchez CE. Angiotensin II-mediated protein kinase D activation stimulates aldosterone and cortisol secretion in H295R human adrenocortical cells. Endocrinology. Dec 2006;147(12):6046-6055 . . 129. Noda M, Ohno S, Nakajin S. Mono-(2-ethylhexyl) phthalate (MEHP) induces nuclear receptor 4A subfamily in NCI-H295R cells: a possible mechanism of aromatase suppression by MEHP. Mol Cell Endocrinol. Aug 15 2007;274(1-2):8- 18. 130. Stigliano A, Cerquetti L, Borra M, et al. Modulation of proteomic profile in H295R adrenocortical cell line induced by mitotane. Endocr Relat Cancer. Mar 2008; 15( 1): 1-10. 131. Kanczkowski W, Zacharowski K, Wirth MP,·Ehrhart-Bomstein M, Bornstein SR. Differential expression and action of Toll-like receptors in human adrenocortical cells. Mol Cell Endocrinol. Mar 5 2009;300(1-2):57-65. 132. Lucki N, Sewer MB. The cAMP-responsive element binding protein (CREB) regulates the expression of acid ceramidase (ASAHl) in H295R human adrenocortical cells. Biochim Biophys Acta. Mar 16 2009. 133. Song R, Duarte TL, Almeida GM, et al. Cytotoxicity and gene expression profiling of two hydroxylated polybrominated diphenyl ethers in human H295R adrenocortical carcinoma cells. Toxzcol Lett. Feb 25 2009;185(1):23-31. 134. Xing Y, Saner-Amigh K, Nakamura Y, et al. The famesoid X receptor regulates transcription of 3beta-hydroxysteroid dehydrogenase type 2 in human adrenal cells. Mol Cell Endocrinol. Feb 27 2009;299(2):153-162. 135. Ye P, Yamashita T, Pollock DM, Sasano H, Rainey WE. Contrasting effects of eplerenone and spironolactone on adrenal cell steroidogenesis. Horm Metab Res. Jan 2009;41(1):35-39. 136. Fluck CE, Martens JW, Conte FA, Miller WL. Clinical, genetic, and functional characterization of adrenocorticotropin receptor mutations using a novel receptor . assay. Journal of Clinical Endocrinology Metabolism. 2002;87 4318-4323.

170 137. Bollag WB, Kent P, White S, Wilson MV, Isales CM, Calle RA. Phorbol ester increases mitochondrial cholesterol content in NCI H295R cells. Mol Cell Endocrinol. Dec 16 2008;296(1-2):53-57. 138-. Brenner T, O'Shaughnessy KM. Both TASK-3 and TREK-1 two-pore loop- K channels are expressed in H295R cells and_ modulate their membrane potential and aldosterone secretion. Am J · Physiol Endocrinol Metab. Dec 2008;295(6):E1480-1486. 139e Lehoux JG, Lefebvre A. Angiotensin II activates p44/42 MAP kinase partly through PKCepsilon in H295R cells. Mol Cell Endocrinol. Feb 2007;265- 266:121-125. 140. Kempna P, Hofer G, Mullis PE, Fluck CE. Pioglitazone inhibits androgen production in NCI-H295R cells by regulating gene expression of CYP17 and HSD3B2. Mol Pharmacol. Mar 2007;71(3):787-798. · 141. Muller-Vieira U, Angotti M, Hartmann RW. The adrenocortical tumor cell line NCI-H295R as an in vitro screening system for the evaluation of CYP11B2 (aldosterone synthase) and CYPl lBl (steroid-1 lbeta-hydroxylase) inhibitors. J Steroid Biochem Mol Biol. Aug 2005;96(3-4):259-270~ 142. Leibovitz A, Mccombs WM, 3rd, Johnston D, McCoy CE, Stinson JC. New human cancer cell culture lines. I. SW-13, small-cell _carcinoma of the adrenal cortex. J Natl Cancer Inst. Aug 1973;51(2):691-697. 143. Cohen AI, Bloch E, Celozzi E. In vitro response ofexperimental adrenal tumors to corticotropin (ACTH). Proc Soc Exp Biol Med. 1957;95 304-309. 144. Buonassisi V, Sato G, Cohen Al. Hormone-producing cultures of adrenal and pituitary tumor origin. Proc NatlAcad Sci US A. 1962;48 1184-1190. 145. Schirnmer BP, Kwan WK, Tsao J, Qiu R. Adrenocorticotropin-resistant mutants of the Yl adrenal cell line fail to express the adrenocorticotropin receptor. J Cell Physiol. 1995; 163(1): 164-171. 146. Es-souni Routhier M, Boumot P, Ramirez LC. Aldosterone synthase activity in the Y-1 adrenal cell line. J Steroid Biochem Mol Biol. 1995;52(6):581-585. 147. Mitchell J, Northup JK, Schirnmer BP. Defective guanyl nucleotide-binding protein beta gamma subunits in a forskolin-resistant mutant of the Yl adrenocortical cell line. Proc Natl Acad Sci US A. 1992;89(19):8933-8937. 148. Es-souni M, Ramirez LC, Boumot P. 18-hydroxylation in the Y-1 adrenal cell line: response to ACTH and to culture conditions. J Steroid Biochem Mol Biol. Nov 1992;43(6):535-541. 149. Szyf M, Milstone DS, Schirnmer BP, Parker KL, Seidman JG. Cis modification of the steroid 21-hydroxylase gene prevents its expression in the Yl mouse adrenocortical tumor cell line. Mol Endocrinol. 1990;4 1144-1152. 150. Ramirez LC, Es-souni M, Boumot P. 18-Hydroxylase activity in the Yl adrenal cell line. Mol Cell Endocrinol. Sep 1989;66(1):83-91. 151. Schirnmer BP. Adenylate cyclase activity in adrenocorticotropic hormone­ sensitive and mutant adrenocortical tumor cell lines. J Biol Chem. 1972;247(10):3134-3138. 152. Endoh A, Kristiansen SB, Casson PR, Buster JE, Hornsby PJ. The zona reticularis is the site of biosynthesis of dehydroepiandrosterone and dehydroepiandrosterone sulfate in the adult human adrenal cortex resulting from its low expression of 3

171 beta-hydroxysteroid dehydrogenase. Journal of Clinical Endocrinology Metabolism. 1996;81(10):3558-3565. 153. Cheng CY, Ryan RF, Vo TP, Hornsby PJ. Cellular senescence involves stochastic processes causing_ loss of expression of differentiated function genes: transfection with SV40 as a means for dissociating, effects of senescence on growth and on differentiated function gene expression. Exp-Cell Res. Jan 1989;180(1):49-62. 154. Chang CW, Naseeruddin SA, Hornsby PJ. The response of 21-hydroxylase messenger ribonucleic acid levels to -adenosine 3',5'-monophosphate and 12-0- tetradecanoylphorbol-13-acetate in bovine adrenocortical cells is dependent on culture conditions. Endocrinology. 1991;128(1)-:604-610. 155. Connell JM, Davies E. The new biology of aldosterone. J Endocrinol. Jul 2005;186(1): 1-20. 156. Mitchell KD, Navar LG. The renin-angiotensin-aldosterone system in volume control. Baillieres Clin Endocrinol Metab. Aug 1989;3(2):393-430. 157. Imai M, Shimada H, Okada Y, Matsushima-Hibiya Y, Ogishima T, Ishimura Y. Molecular cloning of a cDNA encoding aldosterone synthase cytochrome P-450 in rat adrenal cortex. FEBS Lett. 1990;263(2):299-302. · 158. Nomura M, Morohashi K, Kirita S, et al. Three forms of rat CYPl lB genes: 11 beta-hydroxylase gene, aldosterone synthase gene, and a novel gene. J Biochem. 1993; 113(2): 144-152. 159. Zhang G, Miller WL. The contains only two CYPl lB (P450cl 1) genes. J Clin Endocrinol Metab. Sep 1996;81(9):3254-3256. 160. - Fardella CE, Hum DW, Rodriguez H, et al. Gene conversion in the CYP11B2 gene encoding P450cl 1AS is associated with, but does not cause, the syndrome of corticosterone methyloxidase Il deficiency. J Clin Endocrinol Metab. Jan- 1996;81(1):321-3-26. 161. Li Y, Hough RB, Piatigorsky J. Tissue-specific activity of the blind mole rat and the two nucleotide-mutated mouse alphaB-crystallin promoter in transgenic mice. Proc Natl Acad Sci US A. Feb 20 2007;104(8):2608-2613. 162. Seki T, Yun J, Oh SP. Arterial endothelium-specific activin receptor-like kinase 1 expression suggests its role in arterialization and vascular remodeling. Circ Res. Oct 3 2003;93(7):682-689. 163. Zubair M, Parker KL, Morohashi K. Developmental links between the fetal and adult zones of the adrenal cortex revealed by lineage tracing. Mol Cell Biol. Dec 2008;28(23):7030-7040. 164. Zubair M, Ishihara S, Oka S, Okumura K, Morohashi K. Two-step regulation of . Ad4BP/SF-1 gene transcription during fetal adrenal development: initiation by a Hox-Pbxl-Prep-1 complex and maintenance via autoregulation by Ad4BP/SF-l. Mol Cell Biol. Jun 2006;26(11):4111-4121. 165. Shima Y, Zubair M, Ishihara S, et al. Ventromedial hypothalamic nucleus-specific enhancer of Ad4BP/SF-1 gene. Mol Endocrinol. Nov 2005;19(11):2812-2823. 166. Latinkic BV, Mo FE, Greenspan JA, et al. Promoter function of the angiogenic inducer Cyr61gene in transgenic mice: tissue specificity, inducibility during wound healing, and role of the serum response element. Endocrinology. 2001; 142(6):2549-2557.

172 167. Xiong W, Wang J, Chao L, Chao J. Tissue-specific expression and promoter analyses of the human tissue kallikrein gene in transgenic mice. Biochem J. Jul 1 1997;325 ( Pt 1):111-116. 168. -Paz J, Wade K, Kiyoshima T, et al. Tissue~ and bone cell-specific expression of bone sialoprotein is directed by a 9.0 kb promoter in transgenic mice. Matrix Biol. Aug 2005;24(5):341-352. · 169. Miskimins R, Knapp L, Dewey MJ, Zhang X. C_ell and tissue-specific expression of a heterologous gene under control of the myelin basic protein gene promoter in transgenic mice. Brain Res Dev Brain Res. Feb 21 1992;65(2):217-221. 170. Goldberg H, Helaakoski T, Garrett LA, et al. Tissue-specific expression of the mouse alpha 2(1) collagen promoter. Studies in transgenic mice and in tissue culture cells. J Biol Chem. Sep 25 1992;267(27):19622-19630. 171. Sigmund CD, Jones CA, Fabian JR, Mullins JJ, Gross KW. Tissue and cell specific expression of a renin promoter-reporter gene construct in transgenic mice. Biochem Biophys Res Commun. Jul 16 1990;170(1):344-350. 172. Bingham NC, Verma-Kurvari S, Parada LF, Parker KL. Development of a steroidogenic factor· 11Cre transgenic mouse line. Genesis. Sep 2006;44(9):419- 424. 173. Lambert-Langlais S, Val P, Guyot S, et al. A transgenic mouse line with specific Cre recombinase expression in the adrenal cortex. Mol Cell Endocrinol. Mar 5 2009;300( 1-2): 197-204. 174. Martinez A, Lefrancois-Martinez AM, Manin M, et al. 5'-flanking and intragenic _ sequences confer androgenic and developmental regulation of mouse aldose reductase-like gene in vas deferens and adrenal in transgenic mice. Endocrinology.· Mar 1999; 140(3): 1338.-1348. 175. Kim AC, Barlaskar FM, Heaton JH, et al. In search of adrenocortical stem and progenitor cells. Endocr Rev. May 2009;30(3):241-263. 176. Morley SD, Viard I, Parker KL, Mullins JJ. Adrenal-specific transgene expression and derivation of conditionally immortal rat adrenocortical cell lines. Endocr Res. Feb-May 1995;21(1-2):353-357. 177. Morley SD, Viard I, Parker KL, Mullins JJ. Adrenocortical-specific transgene expression directed by steroid hydroxylase gene promoters. Endocr Res. Nov 1996;22(4 ): 631-639. . 178. Ovcharenko I, Nobrega MA, Loots GG, Stubbs L. ECR Browser: a tool for _ visualizing and accessing data from comparisons of multiple vertebrate genomes. Nucleic_Acids Res. 2004 Jul 2004;32(Web Server issue):W280-286. 179.. Seki T, Hong KH, Yun J, Kim SJ, Oh SP. Isolation of a regulatory region of actiyin receptor-like kinase 1 gene sufficient for arterial endothelium-specific expression. Circ Res. 2004 Apr 2004;94(8):e72-77. 180. Vallon V, Grahammer F, Volkl H, et al. KCNQl-dependent transport in renal and gastrointestinal epithelia. Proc Natl Acad Sci US A. Dec 6 2005; 102(49): 17864- 17869. 181. Shelton JH, Jones AL. The fine structure of the_ mouse adrenal cortex and the ultrastructural changes in the zona glomerulosa with low and high sodium diets. Anat Rec. Jun 1971;170(2):147-181.

173 182. Morley SD, Viard I, Chung BC, Ikeda Y, Parker KL, Mullins JJ. Variegated expression of a mouse steroid 21-hydroxylase/beta- galactosidase transgene suggests centripetal migration of adrenocortical cells. Mol Endocrinol. May 1996; 10(5):585-598. 183. Kim AC, Reuter AL, Zubair M, et al. Targ~ted disruption of beta-"catenin in Sfl­ expressing cells impairs development and maintenance of the adrenal cortex. Development. Aug 2008; 135(15):2593-2602. 184. Jorgez CJ, De Mayo FJ, Matzuk MM. Inhibin alpha-iCre mice: Cre deleter lines for the gonads, pituitary, and adrenal. Genesis. Apr 2006;44(4):183-188. 185. Ogishima T, Suzuki H, Hata J, Mitani F, lshimura Y. Zone-specific expression of aldosterone synthase cytochrome P-450 and cytochrome P-45011 beta in rat adrenal cortex: histochemical basis for the _functional zonation. Endocrinology. 1992;130(5):2971-2977. 186. Tremblay A, Parker KL, LeHoux JG. Dietary potassium supplementation and sodium restriction stirn.ulate aldosterone synthase but not 11 beta-hydroxylase P- 450 messenger· ribonucleic acid accumulation in rat adrenals and require angiotensin II productiori. Endocrinology. 1992; 130(6):3152-3158. 187. Gomez-Sanchez CE, Zhou MY, Cozza EN, Morita H, Foecking MF, Gomez~ Sanchez EP. Aldosterone biosynthesis in the rat brain. Endocrinology. Aug 1997; 138(8}:3369-3373. 188. Hatakeyama H, Miyamori I, Fujita T, Takeda Y, Takeda R, Yamamoto H. Vascular aldosterone. Biosynthesis and a link to angiotensin · ii-induced hypertrophy of vascular smooth muscle cells. J Biological Chem. 1994;269(39):24316-24320. 189. Takeda R, Hatakeyama H, Takeda Y, et al. Aldosterone biosynthesis and action in vascular cells. Steroids. Jan 1995;60(1):120-124. 190. Takeda Y, Miyamori I, Yoneda T, et al. Production of aldosterone in isolated rat blood vessels. Hypertension. Feb 1995;25(2):170-173. 191. Silvestre JS, robert V, Heymes C, et al. Myocardial production of aldosterone and corticosterone in the rat. Physiological regulation. J Biol Chem~ 1998;273:4883- 4891. · 192. Kayes-Wandover KM, White PC. Steroidogenic enzyme gene expression in the human heart. J Clin Endocrinol Metab. Jul 2000;85(7):2519-2525. 193. Young MJ, Clyne CD, Cole TJ, Funder JW. Cardiac steroidogenesis in the normal and failing heart. J Clin Endocrinol Metab. Nov 2001;86(11):5121-5126. 194. Takeda Y. Vascular synthesis of aldosterone: role in hypertension. Mol Cell Endocrinol. Mar 31 2004;217(1-2.):75-79. 195. MacKenzie SM, Clark CJ, Fraser R, Gomez-Sanchez CE, Connell JM, Davies E. Expression of 11 beta-hydroxylase and aldosterone synthase genes in the rat brain. J Mol Endocrinol. Jun 2000;24(3}:321-328. 196. Rainey WE, Saner K, Schimmer BP. Adrenocortical cell lines. Mol Cell Endocrinol. Dec 30 2004;228(1-2):23-38. 197. Staels B, Hum DW, Miller WL. Regulation of steroidogenesis in NCI-H295 cells: a cellular model of the human fetal adrenal. Mol Endocrinol. 1993;7(3):423-433.

174 198. Bird IM, Mason JI, Rainey WE. Protein kinase A, protein kinase C, and Ca(2+)­ regulated expression of 21-hydroxylase cytochrome P450 in H295R human adrenocortical cells. J Clin Endocrinol Metab. May 1998;83(5}:1592-1597. 199. Wijesuriya SD, Zhang G, Dardis A, Miller WL. Transcriptional regulatory elements of the human gene for cytochrome P450c21 (steroid 21-hydroxylase}lie within intron ·35 of the linked C4B gene. J Biol Chem. Dec 31 1999~274( 53):38097-38106. 200. Lin CJ, Martens JW, Miller WL. NF-lC, Spl, and Sp3 are essential for transcription of the human gene for P450cl 7 (steroid l 7alpha-hydroxylase/17,20 lyase) in human adrenal NCI-H295A cells. Mol Endocrinol. Aug 2001; 15(8): 1277-1293. 201. Jimenez P, Saner K, Mayhew B, Rainey WE. GATA-6 is expressed in the human adrenal and regulates transcription of genes required for adrenal androgen biosynthesis. Endocrinology. 2003; 144(10):4285-4288. 202. Fluck CE, Miller · WL. GATA-4 and GATA-6 modulate tissue-specific transcription of the human gene for P450c17 by direct interaction with SPl. Mol Endocrinol. 2004;18 1144-1157. 203. Bassett MH, Suzuki T, Sasano H, et al. The orphan nuclear receptor NGFIB · regulates transcription of 3beta-hydroxysteroid dehydrogenase. implications for the control of adrenal functional zonation. J Biol Chem. Sep 3 2004;279(36):37622-37630. 204. Schimmer BP. Adrenocortical Yl cells. Methods Enzymol. 1979;58:570-574. 205. Penhoat A, Naville D, El Mourabit H, Buronfosse A, Durand P, Begeot M. Functional expression of the human· ACTH receptor gene. Endocr Res. Nov 2000;26(4 ): 549-557. 206. Mountjoy KG, Bird IM, Rainey WE, Cone RD. ACTH induces up-regulation of ACTH receptor mRNA in mouse and human adrenocortical cell lines. Mol Cell Endocrinol. Feb 1994;99(1):R17-20. 207. . Janes ME, Chu KM, Cl~k AJ, King PJ. Mechanisms of Adrenocorticotropin­ Induced Activation of Extracellularly Regulated Kinase 1/2 Mitogen-Activated Protein Kinase in the Human H295R Adrenal Cell Line. Endocrinology. Apr 2008;149(4):1898-1905. 208. Pierson RWJ. Metabolism of steroid hormones in adrenal cortex tumor cultures. Endocrinology. 1967;81 693-707. 209. Kowal J, Fiedler R. Adrenal ·cells in tissue culture. I. Assay of steroid products; steroidogenic responses to peptide hormones. Arch Biochem Biophys. 1968;128:406-421. 210. Parker KL, Chaplin DD, Wong M, Seidman JG, Smith JA, Schimmer BP. Expression of murine 21-hydroxylase in mouse adrenal glands and in transfected Yl adrenocortical tumor cells. Proc Natl Acad Sci US A. 1985;82 7860-7864. 211. Guo IC, Huang C, Chung BC. Differential regulation of the CYPl lAl (P450scc) and ferredoxin genes in adrenal and placental cells. DNA Cell Biol. 1993;12 849- 860. 212. Black SM, Szklarz GD, Harikrishna JA, Lin D, Wolf CR, Miller WL. Regulation of proteins in teh cholesterol side-chain cleavage system in JEG-3 and Y-1 cells. Endocrinology. 1993; 132 539-545.

175 213. Lin X, Takemori H, Katoh .Y, et al. Salt-inducible kinase is involved in the ACTH/cAMP-dependent protein kinase signaling in Yl mouse adrenocortical tumor cells. Mol Endocrinol. 2001;15(8):1264-1276. 214. Wong M, Rice DA, Parker KL, Schimmer BP. The roles of cAMP and cAMP­ dependent protein kinase in the expression of cholesterol side chain cleavage and steroid 11 beta-hydroxylase genes in mouse adrenocortical tumor cells. J Biol Chem.· 1989;264(22): 12867-12871. 215. Mitani F, Mukai K, Miyamoto H, Ishimura Y. Localization of replicating cells in rat adrenal cortex during the late gestational and early postnatal stages. Endocr -Res. 1998;24(3-4):983-986. 216. Rice DA, Aitken LD, Vandenbark GR, et al. A cAMP-responsive element regulates expression of the mouse steroid 11 beta-hydroxylase gene. J Biol Chem. 1989;264(24): 14011-14015. 217. Lin D, Sugawara T, Strauss JF, Ill, et al. Role of steroidogenic acute regulatory protein in adrenal and gonadal steroidogenesis. Science. 1995;267(5205):1828- 1831. 218. Lopez D, Nackley AC, Shea-Eaton W, Xue J, Schimmer BP, McLean MP. Effects of mutating different steroidogenic factor-1 protein regions on gene regulation. Endocrine. 2001;14(3):353-362. 219. Temel RE, Trigatti B, DeMattos RB, Azhar S, Krieger M, Williams DL. Scavenger receptor class B, type I (SR-Bl). is the major route for the delivery of high density lipoprotein ·cholesterol to the steroidogenic pathway in cultured mouse adrenocortical cells. Proc Natl Acad Sci US A. 1997;94(25):13600-13605. 220. Schimmer BP. Adrenocortical Yl cells. Methods Enzymol. 1979;58(570-574). 221. Schimmer BP. Isolation of ACTH-resistant Yl adrenal tumor cells. Methods Enzymol. 1985; 109:350-356. 222. Cuprak LJ, Lammi CJ, Bayer RC Scanning electron microscopy of induced cell rounding of mouse adrenal cortex tumor cells in culture. Tissue Cell. 1977 ;9 667- 680. 223. Voorhees H, Aschenbrenner J, Carnes J, Mrotek J. Round_ing and steroidogenesis of enzyme- and ACTH-treated Y-1 mouse adrenal tumor cells. · Cell Biol Intl. 1984;8:483-497. 224. Mattson P, Kowal J. The ultrastructure of functional mouse adrenal cortical tumor cells in vitro. Differentiation. 1978;11 75-88. 225. Osawa S, Betz G, Hall PF. Role of actin in the responses of adrenal cells to ACTH and cyclic AMP: inhibition by DNase I. J Cell Biol. 1984;99 1335-1342. 226. Shiver TM, Sackett DL, Knipling L, Wolff J. Intermediate filaments and steroidogenesis in adrenal Y-1 cells: acrylamide stimulation of steroid production. Endocrinology. 1992;131 201-207. 227. Whitehouse BJ,- Gyles SL, Squires PE, et al. Interdependence of steroidogenesis and shape changes in Yl adrenocortical cells: studies with inhibitors of phosphoprotein phosphatases. Journal of Endocrinology. 2002; 172 583-593. 228. Cortese F, Wolff J. Cytochalasin-stimulated steroidogenesis from high density lipoproteins. J Cell Biol. 1978;77 507-516. 229. Y asumura Y. Retention of differentiated function in clonal animal cell lines, particularly hormone-secreting cultures. Am Zoologist. 1968(8):285-305. 230. Schimmer BP. Phenotypically variant adrenal tumor cell cultures with biochemical lesions in the ACTH-stimulated steroidogenic pathway. J Cell Physiol. 1969;74(2):115-122. 231. Colantonio CM, Kwan WK, Czerwinski W, Mitchell J, Schimmer BP. Altered G protein activity in a desensitization-resistant mutant of the Yl adrenocortical tumor cell line. Endocrinology. 1998;139(2):626-633. 232. Qiu R, Tsao J, Kwan WK, Schimmer BP. Mutations to forskolin resistance result in loss of adrenocorticotropin receptors and consequent reductions in levels of G protein alpha-subunits. Mol Endocrinol. 1996;10(12)-:1708-1718. 233. Rae PA, Gutmann NS, ·Tsao J, Schimmer BP. Mutations in cyclic AMP­ dependent protein kinase and ·corticotropin (ACTH)-sensitive adenylate cyclase affect adrenal steroidogenesis. Proc NatlAcad Sci US A. 1979;76(4):1896-1900. 234. Olson MF, Krolczyk AJ, Gorman KB, Steinberg RA, Schimmer BP. Molecular basis for the 3',5'-cyclic adenosine monophosphate resistance of Kin mutant Yl adrenocortical tumor cells. Mol Endocrinol. 1993;7(4),:477-487. 235. Thomas M, Suwa T, Yang L, Zhao L, Hawks CL, Hornsby PJ. Cooperation of hTERT, SV40 T antigen and oncogenic Ras in tumorigenesis: a cell transplantation model using bovine adrenocortical cells. Neoplasia. 2002;4(6):493-500~ 236. Cong YS, Wright WE, Shay JW. Human telomerase and its regulation. Microbiol Mol Biol Rev. 2002;66(3}:407-425. 237. Hornsby PJ, McAllister JM. Culturing steroidogenic cells. Methods Enzymol. 1991 ;206:371-380. 238. McAllister JM, Byrd W, Simpson E. The effects of growth factors and phorbol esters on steroid biosynthesis in isolated human theca interna and granulosa-lutein cells in long term culture. Journal of Clinical Endocrinology Metabolism. 1994;79(1):106-112. 239. Wu KD, Chen YM, Chu TS, Chueh SC, Wu MH, Bor-Shen H. Expression and localization of human dopamine D2 and D4 receptor mRNA in the adrenal gland, aldosterone-producing adenoma, and pheochromocytoma. J Clin Endocrinol Metab. Sep 2001;86(9):4460-4467. 240. Zhao X, Martin MM, Elton TS. The transcription factors Spl and Sp3 are required for human angiotensin TI type 1 receptor gene expression in H295-R cells. Biochim Biophys Acta. 2001;1522 (3):195-206. 241. Bird IM, Mason JI, Rainey WE. Hormonal regulation of angiotensin TI type 1 receptor expression and ATl-R mRNA levels in human adrenocortical cells. Endocr Res. 1995;21(1-2):169-182. 242. Hilbers U, Peters J, Bornstein SR,· et al. Local renin-angiotensin system is involved in K+-induced aldosterone secretion from human adrenocortical NCI­ H295 cells. Hypertension. Apr 1999;33(4 ): 1025-1030. · 243. Isales CM, Barr.ett PQ, Brines M, BoUag W, Rasmussen H. Parathyroid hormone modulates angiotensin II-induced aldosterone secretion from .the adrenal glomerulosa cell. Endocrinology. 1991;129(1):489-495. 244. Hanley NA, Wester RM, Carr BR, Rainey WE. Parathyroid hormone and parathyroid hormone-related peptide stimulate aldosterone production in the human adrenocortical cell line, NCI-H295. Endocr J. 1993;1:447-450:

177 245. Clark BJ, Pezzi V, Stocco DM, Rainey WE. The steroidogenic acute regulatory protein is induced by angiotensin Il and K+ in H295R adrenocortical cells. Mol Cell Endocrinol. 1995; 115(2):215-219. 246. Winqvist 0, Karlsson FA, Kampe 0. 21-hydroxylase, a major autoantigen in idiopathic addison's disease. Lancet. 1992;339(8809):1559-1562. 247. McAllister JM, Hornsby PJ. Dual regulation of 3 beta-hydroxysteroid dehydrogenase, 17 alpha- hydroxylase, and dehydroepiandrosterone sulfotransferase by adenosine 3',5'-monophosphate and activators of protein kinase C in cultured human adrenocortical cells. Endocrinology. 1988;122(5):2012-2018. 248. Rainey WE, Naville D, Mason JI. Regulation of 3 beta-hydroxysteroid dehydrogenase in adrenocortical cells: effects of angiote~sin-II and transforming growth factor beta. Endocr Res. 1991;17(1-2):281-296. 249. Bird IM, Imaishi K, Pasquarette MM, Rainey WE, Mason JI. Regulation of 3 beta-hydroxysteroid dehydrogenase expression in human adrenocortical H295R cells. Journal of Endocrinology. 1996;150 Suppl:S165-Sl73. 250. Pezzi V, Clyne CD, Ando S, Mathis JM, Rainey WE. Ca(2+}-regulated expression of aldosterone synthase is mediated by calmodulin and calmodulin­ dependent protein kinases. Endocrinology. Feb 1997;138(2):835-838. 251. Holland OB, Mathis JM, Bird IM, Rainey WE. Angiotensin increases aldosterone synthase mRNA levels in human NCI-H295 cells. Mol Cell Endocrinol. 1993;94(2):R9-13. 252. Mizutani T, Ito T, Nishina M, Yamamichi N, Watanabe A, Iba H. Maintenance of integrated proviral gene expression requires Brm, a catalytic subunit of SWI/SNF complex. J Biol Chem. May 3 2002;277(18):15859-15864. 253. Parmar J, Rainey WE, Key RE, Kulharya AS. Development of an ACTH Responsive Human Adrenocortical Cell Line (HAC). 2006. 254. Parmar J, Rainey WE, Key RE. Angiotensin II and Potassium Regulate Aldosterone Production: the Expression of Steroidogenic Enzymes and Type 1 Angiotensin Il Receptors in a Newly Developed Adrenocortical Cell Line. 2007. 255. Schimmer BP, Zimmerman AE. Steroidogenesis and extracellular cAMP accumulation in adrenal tumor cell cultures. Mol Cell Endocrinol. 1976;4(4):263- 270. 256. Havelock JC, Rainey WE, Carr BR. Ovarian granulosa cell lines. Mol Cell Endocrinol. Dec 30 2004;228(1-2):67-78. 257. Rainey MD, Zachos G, Gillespie DA. Analysing the DNA damage and replication checkpoints in DT40cells. Subcell Biochem. 2006;40:107-117. 258. Roskelly CD, Auersperg N. Rapid ras-oncogene-mediated transformation maintains steroidogenic differentiation in adrenocortical parenchymal cells. Differentiation. 1995 ;59 103-111. 259. Cheng CY, Hornsby PJ. Expression of 11 beta-hydroxylase and 21-hydroxylase in long-term cultures of bovine adrenocortical cells requires extracellular matrix factors. Endocrinology. 1992;130(5):2883-2889. 260. Chang SF, Chung BC. Difference in transcriptional activity of two homologous CYP21A genes. Mol Endocrinol. 1995;9(10):1330-1336.

178 261. Huang N, Dardis A, Miller WL. Regulation of cytochrome b5 gene transcription by Sp3-, GATA-6, and steroidogenic factor 1 in human adrenal NCI-H295A cells. Mol Endocrinol. Aug2005;19(8):2020-2034. 262. Samandari E, Kempna P, Nuoffer JM, Hofer G, Mullis PE, Fluck CE. Human adrenal corticocarcinoma NCI-H295R cells produce more androg~ns than NCI­ H295A cells and differ in 3beta-hydroxysteroid dehydrogenase type 2 and 17 ,20 lyase activities. J Endocrinol. Dec 2007;195(3):459-472. 263. Nelson DR, Koymans L, Kamataki T, et al. P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics. 1996;6(1): 1-42. 264. Shoemaker RH. The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer. Oct 2006;6(10):813-823. 265. Weiss RB, Charles LM, Jr., Macdonald JS. m-AMSA: an exciting new drug in the National Cancer Institute Drug Development Program. Cancer Clin Trials. Fall 1980;3(3):203-209. . 266. Fisher RI, Terry WD, Hodes RJ, et al. Adjuvant immunotherapy or chemotherapy for malignant melanoma. Preliminary report of the National Cancer Institute randomized clinical trial. Surg Clin North Am. Dec 1981;61(6):1267-1277. 267. Bodrogi I. Third-line chemotherapy of resistant advanced testicular cancer. Prog Clin Biol Res. 1989;303:749-758. 268. Lopez Garcia N. [Contribution to the study of adenocarcinoma of the endometrium. Part I: Introduction. Part II: Material and methods]. Rev Esp Oncol. 1980;27(3):443-521 contd. 269. . La· Rocca RV, Stein .CA, Danesi R, Jamis-Dow CA, Weiss OH, Myers CE. Suramin in adrenal cancer: modulation of steroid hormone production, cytotoxicity in vitro, and clinical antitumor effect. J Clin Endocrinol Metab. Aug 1990;71 (2):497-504. 270. Schteingart DE, Sinsheimer JE, Counsell RE, et al. Comparison of the adrenalytic activity of mitotane and a methylated homolog on normal adrenal cortex and adrenal cortical carcinoma. Cancer Chemother Pharmacol. 1993;31(6):459-466. 271. Fallo F, Pilon C, Barzon L, et al. Paclitaxel is an effective antiproliferative agent on the human NCI-H295 adrenocortical carcinoma cell line. Chemotherapy. Mar­ Apr 1998;44(2): 129-134. 272. Fallo F, Pilon C, Barzon L, et al. Effects of taxol on the human NCI-H295 · adrenocortical carcinoma cell line. Endocr Res. Nov 1996;22(4):709-715. 273. Fassnacht M, Hahner S, Beuschlein F, Klink A, Reineke M, Allolio B. New · mechanisms of adrenostatic compounds in a human adrenocortical cancer cell line. Eur J Clin Invest. Dec 2000;30 Suppl 3:76-82. 274. Betz MJ, Shapiro I, Fassnacht M, Hahner S, · Reineke M, Beuschlein F. Peroxisome proliferator-activated receptor-gamma agonists suppress adrenocortical tumor cell proliferation and induce differentiation. J Clin Endocrinol Metab. Jul 2005;90(7):3886-3896. 275. Almeida MQ, Fragoso MC, Lotfi CF, et al. Expression of insulin-like growth factor-II and its receptor in pediatric and adult adrenocortical tumors. J Clin Endocrinol Metab. Sep 2008;93(9):3524-3531.

179 276. van Koetsveld. PM, Vitale G, de Herder WW, et al. Potent inhibitory effects of type I interferons on human adrenocortical carcinoma cell growth. J Clin Endocrinol Metab. Nov 2006;91(11):4537-4543. 277. Schteingart DE, Giordano TJ, Benitez RS, _et al. Overexpression of CXC chemokines by an adrenocortical carcinoma: a novel clinical syndrome. J Clin Endocrinol Metab. Aug 2001 ;86(8):3968-3974. 278. Doghman M, Cazareth J, Lalli E. The T cell factor/beta-catenin antagonist PKF115-584 inhibits proliferation of adrenocortical carcinoma cells. J Clin Endocrinol Metab. Aug 2008;93(8):3222-3225. 279. Barlaskar FM, Spalding AC, Heaton JH, et al. Preclinical targeting of the type I insulin-like growth factor receptor in adrenocortical carcinoma. J Clin Endocrinol Metab. Jan 2009;94(1):204-212. 280. Ghorab Z, Jorda M, Ganjei P, Nadji M. Melan A (A103) is expressed in adrenocortical neoplasms but not in renal cell and hepatocellular carcinomas. Appl Immunohistochem Mol Morphol. Dec 2003;11(4):330.. 333. 281. Ueno M, Nakashima J, Akita M, et al. Characterization of a newly established cell line derived from human adrenocortical carcinoma. Int J Urol. Jan 2001;8(1):17- 22. 282. Hornsby PJ, Ryan RF, Cheng CY. Replicative senescence and differentiated gene expression in cultured adrenocortical cells. Exp Gerontol. 1989;24(5-6):539-558. 283. Quinn SJ, Williams GH,, James VHT. Regulation of aldosterone secretion. The Adrenal Gland. New York: Raven Press; 1992:159-189. 284. Lenzini L, Seccia TM, Aldighieri E, et al. Heterogeneity of aldosterone-producing adenomas revealed by · a whole transcriptome analysis. Hypertension. Dec 2007 ;50( 6): 1106-1113. 285. Muller J, Meuli C, Schmid C, Lauber M. Adaptation of aldosterone biosynthesis to sodium and potassium intake in the rat. J Steroid Biochem. 1989;34(1-6):271- 277. 286. Shibata H, Ogishima T, Mitani F, et al. Regulation of aldosterone synthase cytochrome P-450 in rat adrenals by angiotensin II and potassium. Endocrinology. May 1991; 128(5):2534-2539. 287. Ohnishi T, Wada A, Lauber M, Y amano T, Okamoto M. Aldosterone biosynthesis in mitochondria of i~olated zones of adrenal cortex. J Steroid Biochem. Jul 1988;31(1):73-81. 288. Bassett MH, White PC, Rainey WE. The regulation of aldosterone synthase · expression. Mol Cell Endocrinol. 2004;217(1-2):67-74. 289. Choi T, Huang M, Gorman C, Jaenisch R. A generic intron increases gene expression in transgenic mice. Mol Cell Biol. Jun 1991;11(6):3070-3074. 290. Creancier L, Morello D, Mercier P, Prats AC. Fibroblast growth factor 2 internal ribosome entry site (IRES) activity ex vivo and in transgenic mice reveals a stringent tissue-specific regulation. J Cell Biol. Jul 10 2000;150(1):275-281. 291. Gallardo HF, Tan C, Sadelain M. The internal ribosomal entry si_te of the encephalomyocarditis virus enables reliable coexpression of two transgenes in human primary T lymphocytes. Gene Ther. Oct 1997;4( 10): 1115-1119.

180 292. Else T, Trovato A, Kim AC, et al. Genetic p53 deficiency partially rescues the _-adrenocortical dysplasia phenotype at the expense of increased tumorigenesis. Cancer Cell. Jun 2 2009; 15(6):465-476. 293. Keegan CE, Rutz JE, Else T, et al. Urogenital and caudal dysg~nesis in adrenocortical dysplasia (acd) mice is caused by a splicing mutation in a novel telomeric regulator. Hum Mol Genet. Jan 1 2005; 14(1): 113-123. 294. Else T, Theisen BK,_Wu Y, et al. Tppl/Acd maintains genomic stability through a complex role in telomere protection. Chromosome Res. 2007;15(8):1001-1013. 295. Gomez-Sanchez EP, Gomez-Sanchez CM, Plonczynski MW, Gomez-Sanchez CE. Aldosterone Synthesis in the Brain Contributes to Dahl Salt Sensitive Rat Hypertension. Exp Physiol. Oct 16 2009. 296. Ahmad N, Romero DG, Gomez-Sanchez EP, Gomez-Sanchez CE. Do human vascular endothelial cells produce aldosterone? Endocrinology. Aug 2004; 145(8):3626-3629. 297. Zhou MY, Gomez-Sanchez EP, Foecking MF, Gomez-Sanchez CE. Cloning and expression of the rat adrenal cytochrome p-450 11 b3 (cyp 11 b3) enzyme cdna: preferential 18-hydroxylation over 11 beta-hydroxylation of doc. Mol Cell­ Endocrinol. 1995; 114(1-2): 137-145. 298. Marwood JF, Ilett KF, Lockett MF. Effects · of adrenalectomy and of hypophy.sectomy on the development of sound-withdrawal hypertension. J Pharm Pharmacol. Feb 1973;25(2):96-100. 299. Ilett KF, Lockett MF. A renally active substance from heart muscle and from blood. J Physiol. May 1968;196(1):101-109. 300. Ilett KF, Lockett MF. Effect of age on the secretion of hydrocortisone and corticosterone into the adrenal venous blood of cats. J Endocrinol. Feb 1969;43(2):313-314. 301. Ilett KF, Lockett MF. Adaptation to the natriuretic actions of hydrochlorothiazide, frusemide and amiloride, by rats. J Pharm Pharmacol. Apr 1972;24(4):302-312. 302. Gomez-Sanchez EP, Ahmad N, Romero DG, Gomez-Sanchez CE. Origin of aldosterone in the rat heart. Endocrinology. Nov 2004;145(11):4796-4802. 303. MacKenzie SM, Lai M, Clark CJ; et al. llbeta-hydroxylase and aldosterone synthase expression in fetal rat hippocampal neurons. J Mol Endocrinol. Dec 2002;29(3):319-325. 304. Yu L, Romero DG, Gomez-Sanchez CE, Gomez-Sanchez EP. Steroidogenic enzyme gene expression in the human brain. Mol Cell Endocrinol. Apr 25 2002;190(1-2):9-17. 305. Bolon B. Whole mount enzyme histochemistry as a rapid screen at necropsy for expression of beta-galactosidase (LacZ)-bearing transgenes: considerations for separating specific Lacz activity from nonspecific (endogenous) galactosidase activity. Toxicol Pathol. 2008;36(2):265-276. 306. Widen SG, Papaconstantinou J. Liver-specific expression of the mouse alpha­ fetoprotein gene is mediated by cis-acting DNA elements. Proc Natl Acad Sci US A. Nov 1986;83(21):8196-8200. 307. Feuerman MH, Godbout R, Ingram RS, Tilghman SM. Tissue-specific transcription of the mouse alpha-fetoprotein gene promoter is dependent on HNF- 1. Mol Cell Biol. Oct 1989;9(10}:4204-4212.

181 308. Keplinger BL, Guo X, Quine J, Feng Y, Cavener DR. Complex organization of promoter and enhancer elements regulate the tissue- and developmental stage­ specific .expression of the Drosophila melanogaster Gld- gene. Genetics. ·Feb 2001 ;157(2):699~ 716. 309. Giordano TJ, Kuick R, Else T, et al. Molecular classific.ation and prognostication of adrenocortical tumors· by transcriptome profiling. Clin Cancer· Res. Jan 15 2009; 15(2):668~676. 310. de Reynies A, Assie G, Rickman DS, et al. Gene expression profiling reveals a new classification of adrenocortical tumors and identifies molecular predictors of malignancy and survival. J Clin Oncol. Mar 1 2009;27 (7): 1108-1115. 311. Continuously Cultured Tissue Cells and Viral Vaccines: Potential advantages may be realized and potential hazards obviated by careful planning and monitoring. Science. Jan 4 1963;139(3549):15-20. 312. Schiebinger RJ, Albertson :SD, Cassorla FG, et al. The developmental changes in plasma adrenal androgens during infancy and adrenarche are associated with changing activities of adrenal microsomal 17-hydroxylase and 17 ,20-desmoslase. J Clin Invest. 1981;67:1177-1182. 313. Hum DW, Miller WL. Transcriptional regulation of human genes for steroi_dogenic enzymes. Clin Chem. 1993;39(2):333-340. 314. Schioth HB, Chhajlani V, Muceniece R, Klusa V, Wikberg JE. Major pharmacological distinction of the ACTH receptor from other · melanocortin receptors. Life Sci. 1996;59(10):797-801. . 315. Cooray SN, Chan L,· Metherell L, Storr H, Clark AJ. Adrenocorticotropin resistance syndromes. Endocr Dev. 2008;13:99-116. 316. Clark AJ, Baig AH, Noon L, Swords FM, Hunyady L, King PJ. Expression, desensitization, and internalization of the ACTH receptor (MC2R). Ann N Y Acad Sci. Jun 2003 ;994: 111-117. . 317. Hayes DN, Monti S, Parmigiani G, et al. Gene expression profiling reveals reproducible human lung adenocarcinoma subtypes in multiple independent patient cohorts. J Clin Oncol. Nov 1 2006;24(3 l ):5079-5090. 318. Marchionni L, Wilson RF, Wolff AC, et al. Systematic review: gene expression profiling assays in early-stage breast cancer. Ann Intern Med. Mar 4 2008; 148(5):358-369. 319. Golub TR, Slonim DK, Tamayo P, et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science. Oct 15 1999;286(5439):531-537. 320. Schaner ME, Ross DT, Ciaravino G, et al. Gene expression patterns in ovarian carcinomas. Mol Biol Cell. Nov 2003;14(11):4376-4386. 321. Takahashi M, Yang XJ, Sugimura J, et al. Molecular subclassification of kidney tumors and the discovery of new diagnostic markers. Oncogene. Oct 2 2003;22(43):6810-6818. 322. Giordano TJ, Kuick R, Thomas DG, et al. Molecular classification of papillary thyroid carcinoma: distinct BRAF, RAS, and RET/PTC mutation-specific gene expression profiles discovered by DNA microarray analysis. Oncogene. Oct 6 2005;24( 44): 6646-6656.

182