Transcript

1

Bile Acid Chemistry, Biology, and Therapeutics

During the Last 80 Years: Historical Aspects

Alan F. Hofmann and Lee R. Hagey

Running footline: History of bile acid research

Department of Medicine, University of California, San Diego 92093-063

Correspondence to Alan F. Hofmann ([email protected]) or Lee R. Hagey ([email protected])

Alan F. Hofmann, M.D., Telephone and fax: (858) 459-1754

Lee R. Hagey, Ph.D. Telephone (619) 543-2281

Abbreviations: ASBT, apical sodium co-dependent bile acid transporter; AUC, area under the curve;

BSEP, bile salt export pump; CDCA, chenodeoxycholic acid; CMC, critical micellization concentration;

CMT, critical micellization temperature; CMpH, critical micellization pH; DCA, deoxycholic acid; FATP,

fatty acid transport protein; FGF, fibroblast growth factor; FGFR4, fibroblast growth factor receptor 4;

FRET, Forster resonance energy transfer; FXR, farnesoid X-Receptor; GLP-1, glucagon-like peptide 1;

MDR, multidrug resistance (protein); MRP4, multidrug resistance protein 4; MTBE, methyl tertbutyl

ether; NTCP, Na+ taurocholate co-transporting polypeptide; OATP, organic anion transporting

polypeptide; OST, organic solute transporter; PC, phosphatidylcholine; PFIC2, progressive familial

intrahepatic cholestasis, type 2; SLCO, solute carrier organic anion; TGR5, transmembrane G protein-

coupled receptor 4; UDCA, ursodeoxycholic acid.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

2

Abstract

During the last 80 years there have been extraordinary advances in our knowledge of the chemistry and

biology of bile acids. We present here a brief history of the major achievements as we perceive them.

Bernal, a physicist, determined the x-ray structure of cholesterol crystals, and his data together with the

vast chemical studies of Wieland and Windaus enabled the correct structure of the steroid nucleus to be

deduced. Today, C24 and C27 bile acids together with C27 bile alcohols constitute most of the bile acid

“family”. Patterns of bile acid hydroxylation and conjugation are summarized. Bile acid measurement

encompasses the techniques of gas chromatography, HPLC, mass spectrometry, as well as enzymatic,

bioluminescent, and competitive binding methods. The enterohepatic circulation of bile acids results from

vectorial transport of bile acids by the ileal enterocyte and hepatocyte; the key transporters have been

cloned. Bile acids are amphipathic, self-associate in solution and form mixed micelles with polar lipids --

phosphatidylcholine in bile, and fatty acids in intestinal content during triglyceride digestion. The rise and

decline of dissolution of cholesterol gallstones by the ingestion of 3,7 dihydroxy bile acids is chronicled.

Scientists from throughout the world have contributed to these achievements.

Key words: Bile acid transport, enterohepatic circulation, bile acid physical chemistry; bile acid analysis;

bile acid metabolism

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

3

Table of contents

I. The ebb and flow of medical interest in bile acids 5

II. Bile acid structure: history and current concepts

A. The carbon skeleton 13

B. the A/B ring junction 16

C. Hydroxylation sites 18

D. Conjugation 25

E. Structure-function relationships 28

III. Measurement

A. Isolation and chromatographic separation 30

B. Mass spectrometry 33

C. Enzymatic and competitive binding techniques 34

D. Diagnostic utility of plasma bile acids 35

E. Determinants of bile acid profiles in body fluids 36

IV. Bile acid metabolism: transport and the enterohepatic circulation

A. Bile acid transport 37

B. Enterohepatic circulation in health 48

C. Perturbations of the enterohepatic circulation 57

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

4

V. Physicochemical aspects of bile acids

A. Overview 58

B. Bile acids in aqueous solution 59

C. Solubilization of polar lipids by bile acid micelles 66

D. Physical chemistry of bile and cholesterol gallstone disease 70

E. Physical chemistry of fat digestion 73

VI. Dissolution of cholesterol gallstones

A. Gallstone dissolution by oral bile acids 75

B. Acceleration of dissolution by shock wave lithotripsy 79

C. Topical dissolution with organic solvents 80

D. Concurrent advances in gallstone management 81

VII. Epilogue: omissions and apologies 82

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

5

This article contains aspects of the history of bile acid research during the last eight decades. The

authors, now in the twilight of their careers, are not trained historians, and there will be omissions,

misconceptions, and provocative judgments. Let us hope the omissions are minor, misconceptions are

rare, and provocative judgments stand the test of time. Let us also hope that this effort will be both useful

and entertaining. Each of the topics discussed merits at least a full length article, so there will be of

necessity in many instances consideration of only what we perceive to be the highlights.

I. The Ebb and Flow of Medical Interest in Bile Acids

After the elucidation of the true chemical structure of bile acids in 1932 (see below), there was little

interest in bile acids in the Western world. One exception to this statement was the laboratory of

Siegfried Thannhauser, who wrote the first textbook of metabolic biochemistry in Germany. He studied

cholesterol and bile acid balance in the biliary fistula dog (1). During this time, bile acids were sold as

liver tonics and laxatives, but there were no placebo-controlled studies showing efficacy. Indeed, bile

acids were considered by the medical profession to have no useful therapeutic properties. The tri-oxo

derivative of cholic acid (called “dehydrocholic acid”) was known to induce bile flow in animals (2), and

was occasionally used to stimulate bile flow in patients; but again there were no controlled studies

showing efficacy in hepatobiliary disease.

Albert Boehringer founded a pharmaceutical company in Ingelheim, Germany. His wife was a cousin

of Heinrich Wieland, then Professor in Munich. Wieland was successful in persuading the company to

begin the isolation and marketing of cholic acid from ox bile in 1917. Thus, cholic acid of high purity

soon became available to the scientific community. During this time, Japanese students came to

laboratories such as that of Wieland to pursue their doctoral studies. When they returned to Japan, they

continued their studies on bile acid and bile alcohol structure. Their work, as well as that occurring in

Europe is summarized in the book “Ueber die Chemie and Physiologie der Gallensäuren” authored by

Shimizu in 1935 (3).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

6

During the 1930’s and 1940’s, an enormous international effort went into defining the structure of

the major hormonal steroids. When it was recognized that cortisone had a C-11 oxygen atom, it was

logical to use deoxycholic acid (possessing a C-12 hydroxyl group) as a chemical precursor for the

synthesis of corticosteroids. Deoxycholic acid (DCA) was easily isolated from bovine bile or synthesized

from cholic acid, and it was not too difficult for a talented chemist to move the oxygen from C-12 to C-

11. In 1946, H. Sarett (at the Merck Company) reported a complex synthesis (37 steps!) of cortisone

from DCA (4), and this led to its commercial production on a small scale. Seven years later, Hench, a

rheumatologist at the Mayo Clinic who worked closely with his colleague Kendall, an able steroid

chemist, obtained a small supply of cortisone supplied by Merck (5). Hench showed that this compound

caused a striking symptomatic improvement in patients with rheumatoid arthritis. The translational

research of Hench and Kendall resulted in their being awarded the Nobel Prize in 1950 (6).

As a result of the truly exciting advent of cortisone in the treatment of rheumatoid arthritis, there was

an enormous effort to develop a simple, efficient synthesis of this hormone from DCA. Soon, there was

concern that the world’s supply of DCA (derived from cow and sheep bile) would be insufficient to meet

the medical demand. Then, workers at Upjohn discovered that hydroxylation at C-11 could be achieved

using a fungus. Other workers found a plant saponin that could be used as a substrate for the fungal

hydroxylation at C-11, after which the side chain was easily altered to that of cortisone. (For details, see

Fieser and Fieser) (7). DCA was no longer needed, and chemical interest in bile acids collapsed.

After the Second World War, a few laboratories pursued the search for new bile acids, as well as

defining the metabolism of bile acids in mammals. The availability of 14C and 3H which could be

incorporated into the bile acid molecule, together with the development of automatic liquid scintillation

counters and chromatography enabled biotransformations to be identified and measured. Sune

Bergström, then working in Lund, Sweden recruited a highly talented group of doctoral students -- Jan

Sjövall, Henry Danielsson, Arne Norman, Sven Lindstedt, Bengt Samuelsson, Sven Eriksson, Bengt

Borgström, among others -- who carried out fundamental studies of bile acid metabolism in a variety of

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

7

species including man (8). (It is said that Bergström chose his graduate students on the golf course. It is

also said that the lights never went off at night in the Bergström laboratory).

Sjövall developed gas chromatography and then later, after he had moved to the Karolinska Institute in

Stockholm, developed mass spectrometry for the measurement of bile acids (9). He used these techniques

and others to define important aspects of bile acid metabolism over the next four decades (10). Norman

developed a simple synthesis of conjugated bile acids, prepared the conjugates of all the major bile acids

known at that time (11), and with Sjövall performed metabolic studies in animals which distinguished

primary bile acids (made in the liver from cholesterol) from secondary bile acids (made from primary bile

acids by intestinal bacteria). Their work proved that DCA was a secondary bile acid (12, 13). Lindstedt

measured bile acid kinetics (pool size, turnover rate, and synthesis) in man (14). Danielsson identified

key intermediates in bile acid biosynthesis (15). Samuelsson synthesized ursodeoxycholic acid (UDCA)

and defined its metabolism (16). Eriksson showed that bile acid synthesis in the rat was under negative

feedback control (17). Borgström with his colleagues Sjövall, Lundh, and Dahlquist, intubated healthy

volunteers and defined the site at which individual nutrients are absorbed (18). This study proposed that

the bile acid pool circulates about twice per meal. Borgström became director of the Department of

Physiological Chemistry in Lund in 1958, replacing Bergström who had moved earlier that year to the

Karolinska Institute in Stockholm with the bile acid group. Bergström and his pupil Samuelsson then left

the bile acid field and turned their attention to prostaglandins. Their outstanding work in this field led to

their receiving the Nobel Prize in 1982. However, at the Karolinska Hospital, Swedish scientists

continued to work on bile acids, clinical and biochemical studies being conducted by Kurt Einarsson, Bo

Angelin, Ingemar Björkhem, and Kjell Wikvall, among others.

In 1961, news of the work of Lack and Weiner (19) which showed unequivocally that the ileum was

the site of conjugated bile acid absorption reached Sweden. Borgström directed the senior author, to

perform studies showing that in man, conjugated bile acids are absorbed mainly in the ileum (20).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

8

In London, Geoffrey Haslewood pursued the structure of bile acids in different species, based on his

belief that bile acid composition was a powerful phenotype that would aid in the identification of

evolutionary relationships (21). Edward Doisy, Chairman of the Department of Biochemistry at St. Louis

University, having already won the Nobel prize for his work on vitamin K, pursued the identity of the

3,6,7-trihydroxy bile acids in rats and identified the α- β- and ω-muricholic acids (22) His student,

William Elliott synthesized numerous bile acids and described their chemical and chromatographic

properties (23).

In Japan, a school of bile acid studies was established by Tayei Shimizu (24), who returned to his

native country after having spent three years in the laboratory of Heinrich Wieland. Japanese

investigators pursued the structure of natural bile acids, the work continuing with Taro Kazuno and his

pupil Takahiko Hoshita (25) and, in turn, with Hoshita’s student Mizuho Une (25, 26). Japanese workers

also attempted to define the early steps in bile acid biosynthesis from cholesterol (27).

In the United States, James Carey, working at the University of Minnesota, was directed by his mentor,

Cecil Watson, to pursue bile acid metabolism in man. He identified chenodeoxycholic acid (CDCA) as a

major biliary bile acid (28) and proposed that lithocholic acid, its bacterial metabolite, caused liver injury

in man (29). Edward (“Pete”) Ahrens, working with Lyman Craig at the Rockefeller University, showed

that countercurrent distribution could be used to separate conjugated and unconjugated bile acids (30).

Later, two of his postdoctoral fellows, Scott Grundy and Tatu Miettinen, developed a gas

chromatographic method for measurement of fecal bile acids, whose output, in the steady state, is

equivalent to bile acid synthesis (31). Grundy went on to a distinguished career in human nutrition (32).

Miettinen continued to do sterol balance studies for much of his very productive career (33).

The first symposium devoted solely to bile acids was organized by Leon Schiff, a clinical hepatologist,

who was one of the founders of the American Association for the Study of Liver Diseases. This

symposium held in 1967 was quite exciting for its participants who are shown in Figure 1.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

9

However, it is safe to say that the study of bile acids was pursued by only a small number of

laboratories, some in Departments of Biochemistry and some in Departments of Medicine. Erwin

Mosbach, one of the early workers in bile acid metabolism, once stated to his wife, “Whenever I go to the

podium to give a paper on bile acids, everyone leaves the room”.

In 1965, the senior author, working in the laboratory of E.H. Ahrens, began feeding studies with cholic

acid in a patient with severe hypercholesterolemia, and showed that cholic acid feeding was a potent

suppressor of bile acid and cholesterol biosynthesis (34), based on measurement of fecal bile acids and

sterols, using the newly developed gas chromatographic method for fecal bile acids that had been

developed in this laboratory (32). It was logical to test CDCA, the other primary bile acid, but at that

time, the world’s supply of pure CDCA was thought to be less than 10 grams, and the synthesis from

cholic acid was difficult. However, in the 1960’s, a small English pharmaceutical company (Weddell

Pharmaceuticals) began the manufacture of CDCA for unknown reasons. A kilogram was purchased for

the senior author by the Mayo Clinic in 1967. Leslie Schoenfield returned to the Mayo Clinic in 1966

after having spent a year in the laboratory of Sjövall, and initiated a clinical trial with his fellow Johnson

Thistle to test whether oral cholic acid or hyodeoxycholic acid would lower cholesterol in bile and

ultimately induce cholesterol gallstone dissolution. The senior author persuaded Schoenfield to add

CDCA to his protocol, and this study of Thistle and Schoenfield showed that CDCA feeding decreased

biliary cholesterol saturation, whereas neither cholic acid nor hyodeoxycholic acid had any effect (35). In

1972, the first gallstone dissolution induced by the ingestion of CDCA was observed, initially at the Mayo

Clinic (36), and later in London by a group led by Hermon Dowling (37). (This was not the first time that

the efficacy of oral bile acids had been tested at the Mayo Clinic. In 1938, Philip Hench had fed a

mixture of conjugated bile salts in an unsuccessful attempt to treat rheumatoid arthritis) (38).

The discovery that CDCA would induce gradual dissolution of cholesterol gallstones led to the next

resurgence of interest in bile acids. For the very first time, CDCA was made in kilogram quantities by

several manufacturers, and became the third bile acid available as a fine chemical.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

10

In Japan, ursodeoxycholic acid (UDCA) had been brought to market in the 1950’s by Tokyo Tanabe as

a liver tonic, based on the legendary therapeutic effects of bear bile. It was packaged as 10 mg tablets,

and the recommended dose is unlikely to have had any pharmacodynamic effects. Two decades later in

Japan, UDCA, in much larger doses, was observed to induce cholesterol gallstone dissolution (39, 40).

UDCA soon replaced CDCA for gallstone dissolution throughout the world because UDCA showed an

efficacy similar to that of CDCA, but in contrast, had virtually no hepatotoxicity. To provide UDCA for

therapeutic trials performed around the world, UDCA was synthesized in kilogram quantities and became

the fourth bile acid to become available as a fine chemical. A Japanese perspective on the discovery of

UDCA as a therapeutic agent is available (41).

As discussed later, dissolution of cholesterol gallstones by ingestion of UDCA began to decline in the

1990’s because of the development of laparoscopic cholecystectomy which promised a rapid cure for the

problems of gallstone disease, irrespective of gallstone type. Once again, bile acids seemed to have little

medical value, and interest in their biology and chemistry declined.

Nonetheless, the ready availability of UDCA meant that it continued to be used for gallstone patients

who were poor operative candidates. Ulrich Leuschner, a hepatologist based in Frankfurt, Germany, noted

that UDCA ingestion improved biochemical parameters in patients with primary biliary cirrhosis (42).

UDCA was subsequently shown to slow disease progression in primary biliary cirrhosis, initially by

Raoul Poupon in Paris (43), and later by Keith Lindor and his colleagues at the Mayo Clinic (44). UDCA

treatment of primary biliary cirrhosis is now standard of care. UDCA was also shown to be effective in

cholestasis of pregnancy (45). UDCA has been widely used for other cholestatic diseases and to restore

bile flow following liver transplantation, but proof of efficacy in these conditions based on placebo-

controlled trials has been uncommon.

At present, it is estimated that about 1000 metric tons or about 1 million kilograms -- of cholic acid are

produced globally from bovine bile, the majority of which is used for the production of UDCA (S. Yorke,

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

11

New Zealand Pharmaceuticals, Ltd.; personal communication). Bile acids are also used as precursors for

the synthesis of nuclear receptor agonists such as obeticholic acid (the 6α-ethyl derivative of CDCA), as

building blocks for other molecules (46), and as key constituents of some bacteria media. Bile acids have

even used in the electronic industry as precursors of photoresist molecules in integrated circuits (47).

(This unlikely technology transfer from a biological laboratory to the electronic world resulted from one

of the early chemists in our laboratory, Rick DiPietro, taking a position in the electronics industry and

envisioning a truly novel use for bile acids).

In 1984, the results of the Coronary Primary Prevention Trial showed that the ingestion of

cholestyramine, the first bile acid sequestrant to be marketed, lowered plasma cholesterol levels and

reduced cardiovascular events. This was of considerable interest to the cardiovascular community, as the

study provided the first convincing evidence for the validity of the cholesterol hypothesis (48).

Nonetheless, the demonstrated efficacy of treating hypercholesterolemia with a bile acid sequestrant had a

rather modest effect on clinical practice because of the poor tolerability of cholestyramine and the

emergence of the statins which were still more potent and usually well tolerated.

The current rebirth of interest in bile acids arises from the astonishing discovery of their signaling

properties. Bile acids were found to be the ligand for the orphan nuclear receptor FXR (farnesoid X-

receptor) (49, 50, 51), meaning that bile acids regulate the activity of many genes (52, 53, 54). Bile acid

derivatives that are potent FXR agonists have been shown to induce the synthesis and basolateral release

of the protein fibroblast growth factor 15/19 (FGF19) from the ileal enterocyte (55). FGF19 travels in

portal venous blood to the liver and down-regulates bile acid biosynthesis (55, 56). FGF19 promotes

hepatocyte growth (57), and FXR agonism also has antidiabetic effects mediated by multiple mechanisms

(58), as well as anti-inflammatory activities (59). Several potent FXR agonists have been synthesized (53,

60) and one of these, obeticholic acid (61), is now being tested for its efficacy in primary biliary cirrhosis

and non-alcoholic steatohepatitis (62).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

12

Two Japanese groups discovered a transmembrane G-coupled protein receptor (TGR5) activated by

bile acids (63, 64). TGR5 has a wide tissue distribution, being most heavily expressed in the enteric

nervous system (65); it is also present in spinal cord neurons (66) and the primary cilia of cholangiocytes

(67). The consequences of this activation include release of the hormone glucagon-like peptide 1 (GLP-

1), which has appetite-suppressing and antidiabetic effects (68). TGR5 also has effects on hepatic blood

flow (69), distal intestinal motility (70), and in addition, has anti-inflammatory effects (71) that are under

active investigation.

Discovery of the signaling properties of bile acids is certainly a new paradigm in the bile acid field.

Bile acids have become hormones. Now highly talented molecular biologists in both academe and the

biotechnology industry are pursuing bile acid biology. Bile acid signaling has been discussed only

briefly in this review, as its aim is to focus on events in the past.

As bile acids have been the subject of scientific study for more than 150 years, the bile acid

literature is vast. The early history of bile acid discovery is available in the two monographs by Harry

Sobotka (72, 73), as well as the masterly text on steroids by the Fiesers published in 1955 (7). A small

pharmaceutical company, Dr. Falk Pharma, based in Freiburg, Germany was the first European company

to bring CDCA to market for gallstone dissolution. Since 1972, the Falk Foundation, funded by Dr. Falk

Pharma, has hosted biennial meetings on bile acid biology, chemistry, and therapeutics. The proceedings

of these meetings, usually published in book form, serve to document the advances that have occurred in

the last four decades. G.A.D. Haslewood’s slim monograph summarizes current knowledge in 1966 (74)

and Haslewood later authored a monograph on the biological importance of bile salts (75). The Elsevier

Encyclopedia of Organic Chemistry (76) contains detailed information on the chemistry of bile acids and

their derivatives. A four volume series “The Bile Acids” edited by Nair and Kritchevsky (77) treats many

of the topics discussed in this article in much greater detail. A book entitled “Sterols and Bile Acids” is

Volume 12 of the series New Comprehensive Biochemistry. This volume, published in 1985, has 6

chapters dealing with varying aspects of bile acids and bile alcohols (78). A multiauthor book describing

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

13

many aspects of bile acid research with particular emphasis on Italian contributions was published in

2000 (79). The senior author (80) and Hermon Dowling (81) have both written overviews of the field

from a medical standpoint in 1985. The Journal of Lipid Research published a number of excellent

reviews on the biology and chemistry of bile acids a few years ago.

The remainder of this historical review will discuss selected topics in more detail with an emphasis on

structure-activity relationships. The review will not summarize our extensive work on bile acids in

different vertebrates, as our findings have been published elsewhere (82). Other topics that have been

omitted are discussed in the epilogue.

II. Bile acid structure: History and current concepts

A. The carbon skeleton

1. The steroid nucleus

In December, 1929, a unique event occurred in Stockholm. Two Nobel prizes in Chemistry were

awarded. In 1927, no prize in Chemistry had been awarded because the Nobel Prize Selection Committee

could not agree on a suitable candidate. During the following year, the committee decided to award the

1927 prize to Heinrich Wieland and the 1928 prize to Adolf Windaus. In his Nobel lecture, Wieland

reviewed his laborious attempts to define the structure of bile acids and presented his current vision of the

structure of cholic acid (83). Windaus presented his proposed structure of cholesterol as well as his

brilliant studies on the formation of vitamin D2 from cholesterol by radiation (84). These two

extraordinarily talented chemists, using the most primitive of chemical techniques, had slowly developed

the idea that both cholesterol and bile acids share a four ring structure with a side chain of five carbon

atoms for bile acids and eight carbons for cholesterol. However, the structures of the steroid nucleus

shown in their Nobel lectures were not correct, and each of these giants described their structures as

“probable”.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

14

The path to the correct structure came from an unlikely origin. The polymath Desmond Bernal,

working in the Mineralogical Museum in Cambridge, England decided together with his colleague

William Astbury to undertake X-ray diffraction studies of organic molecules. Bernal, nominally a

physicist, began his studies in 1931. He studied ergosterol, cholesterol, and vitamin D and for all three of

these compounds obtained a probable structure of a flat molecule. He published his findings as a note in

Nature in 1932 (85). This report caught the attention of Rosenheim and King in London, and Wieland

and Dane in Munich. Later, in that same year, Wieland and Dane followed by Rosenheim and King

published the application of Bernal’s pioneering studies to the extensive literature on bile acid structure

(86). Each group proposed the cyclopentanoperhydrophenanthrene structure for the steroid nucleus.

Their final structure has stood the test of time with confirmation by both X-ray diffraction (87) and a total

synthesis of CDCA (88). Figure 2 shows the structure of CDCA and the numbering system for the steroid

nucleus and side chain that was proposed in the 1930’s and has been used ever since.

Bernal’s X-ray diffraction studies were the first biological application of this revolutionary technique.

Bernal later used X-ray diffraction to deduce the structure of other organic molecules, and his pupils such

as Dorothy Crowfoot Hodgkin extended the work, its ultimate triumph being the helical structure of DNA

and RNA by Watson and Crick. Bernal’s left wing political views and his Bohemian life style probably

kept him from being awarded a Nobel Prize (89,90).

2. The side chain

The structure of the C5 side chain of the common natural C24 bile acids – isopentanoic acid or more

specifically γ-methylbutanoic acid – was elucidated by Wieland, and like the structure of the steroid

nucleus, has also been confirmed by X-ray diffraction (87) and total synthesis (88).

The structure of the C8 side chain in C27 bile alcohols and bile acids was more problematic. It was

not clear initially that these molecules possessed the side chain of cholesterol because it was quite

uncertain whether these compounds and even the common natural C24 bile acids were derived from

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

15

cholesterol. The uncertainty arose from the failure of a dietary cholesterol load to increase bile acid

output in the biliary fistula dog. George Whipple, a leading physiologist, wrote in his 1922 review on the

constituents of bile (91). “Cholesterol....has often been suspected as the precursor of cholic acid, ... but

careful experiments negative [sic] this interesting suggestion”. Nonetheless, Shimizu in his book “Ueber

die Chemie und Physiologie der Gallensäuren”, published in 1935 (3) noted that, “Interesting

intermediates in the biological catabolism of cholesterol to bile acids can be found in scymnol and

tetraoxybufostan [both C27 bile alcohols]”. Even earlier, in 1923, Ruzicka had proposed that bile acids are

formed from cholesterol (92).

Proof that cholesterol was converted to bile acids had to await the introduction of isotopes into

biochemistry by Rudolph Schoenheimer (93). Bloch, Berg, and Rittenberg prepared cholesterol labeled

with deuterium, gave it to a dog with a surgical connection between the biliary tract and the renal pelvis;

in this chronic preparation bile is excreted in the urine. Cholic acid was isolated from urine and shown to

have incorporated deuterium into its structure, thus establishing the conversion of cholesterol to C24 bile

acids (94). Figure 3 shows the structure of cholesterol and the changes that occur as it is converted to

CDCA. (Although Konrad Bloch received the Nobel Prize in 1964 for his extraordinary work on fatty

acid and sterol synthesis, his Nobel lecture does not even mention this key experiment).

The first bile acid with a longer side chain than the common natural C24 bile acids was isolated

from the bile of the toad Bufo vulgaris formosus by Shimizu and Oda in 1934 (95). The structure of this

bile acid was established by Takahiko Hoshita et al as a trihydroxy bile acid with a double bond at C-22

and a carboxyl group at C-24 (25). Thus this bile acid is a C28 bile acid having one more carbon than its

precursor cholesterol. Other C28 bile acids with a methyl group on the side chain have been reported (26),

but are quite uncommon in nature.

C27 bile acids are much more common (82). They were isolated from the bile of a number of

early evolving vertebrates. In particular, bile from the alligator, which was readily available in large

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

16

amounts (studies at that time began with up to 5 liters of bile), became the object of intense study as it

contains a great variety of C27 bile acids (96, 97).

The carbon skeleton of the side-chain of C27 bile acids was assumed to have remained unchanged

from that of the parent compound, cholesterol, but the location of the carboxyl group was unclear. In

1952, Bridgwater and Haslewood synthesized a defined side-chain starting from the steroid nucleus.

They then showed that their product was identical to 3α,7α,12α-trihydroxy-5β-cholestan-27-oic acid, the

major bile acid isolated from the bile of crocodiles and alligators, and they confirmed its structure by X-

ray diffraction (98). Subsequently, the laboratory of Takashi Iida reported the synthesis of the 25R and

25S stereoisomers of this compound (99).

The first C27 bile alcohol to be isolated was scymnol, obtained from the bile of the Greenland

shark (Scymnus borealis) by Olof Hammarsten, working in Sweden in 1898 (100). The finding that the

molecule had 27 carbon atoms is a tribute to the precision and accuracy of the combustion techniques that

were used at that time to determine elemental analysis. A half century later, another C27 bile alcohol was

isolated from the bile of the American bullfrog (Rana catesbeiana) by Kazuno et al (101). Subsequently,

Haslewood (102) found the same bile alcohol in the European common frog (Rana temporia), and gave it

the common name “ranol”. Our extensive survey of vertebrate bile acids (82) has established that most

bile alcohols have an iso-octanoic acid (α,ε-dimethyl-hexanoic acid) side chain. The exceptions identified

to date are C26 and C25 alcohols that are present in the bile of some frog species (25, 26), as well as a C24

bile alcohol, petromyzonol that occurs in the lamprey as a disulfate (103,104). Trace proportions of C24

bile alcohols with the nuclear structure of cholic acid and CDCA are found in the bile of many species

(105).

B. The A/B ring junction

In most natural bile acids whether C24 or C27, the junction of the A and B rings is cis. This steric

conformation is indicated by depicting the bond connecting the C-5 hydrogen atom to the A/B ring

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

17

juncture as a solid line or wedge. The much rarer “allo” bile acids (A/B ring junction is trans) are usually

denoted by a dashed line coming from the ring junction to the hydrogen atom. For the non-steroid

chemist, it is not easy to grasp that the orientation of the C-5 hydrogen atom indicates the type of A/B ring

junction, and thus determines the shape of the two markedly different bile acid epimers. The structures of

cis and trans decalin, model compounds for the A and B rings of bile acids are shown in Figure 4. Every

bile acid and bile alcohol has, in principle, a 5α and a 5β epimer.

Both Wieland and Windaus were fully aware of the problem of bile acid geometric isomerism at the

time of their Nobel Prize lectures. A detailed review of some of the experimental work elucidating the

nature of the A/B ring junction in various bile acids is available in Fieser and Fieser’s comprehensive text

on steroids (7). Probably, it was Windaus who introduced the prefix allo to denote 5α (A/B trans) bile

acids.

Elliott has summarized the chemical and biological properties of allo bile acids in his chapter in The

Bile Acids, volume I (106). One way to distinguish 5α bile acids from 5β bile acids is to prepare their

per-oxo derivatives, which are readily distinguished using optical rotatory dispersion (107).

C24 allo bile acids are the dominant primary bile acids only in lizards, although they are present in trace

proportions in the bile of many species (82). If cholestanol (the saturated 5α-derivative of cholesterol) is

administered to rabbits, it is converted to 5α- bile acids (107). Allocholic acid, the biological precursor of

allodeoxycholic acid, has been prepared synthetically and some its biological properties explored (108).

Allo bile acids have also been identified in fecal bile acids (109), presumably generated by bacterial

enzymes acting on the 3-oxo-Δ4,6 intermediate that is formed during the process of C-7 dehydroxylation

(110). Enantiomeric bile acids, in which the steric configuration of all ring junctions is reversed, have

recently been synthesized (111).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

18

Bile alcohols (C27) with a 5α configuration are not uncommon in nature. The simplest

biotransformation of cholesterol (Δ5) into a bile alcohol results in a 5α nucleus, and many early evolving

vertebrates form A/B trans bile alcohols (25, 26, 82).

The preceding discussion thus indicates that there are only four carbon skeletons in most natural bile

acids and alcohols – the C27 cholestane skeleton (with either A/B cis or A/B trans structure) and the C24

cholane skeleton (with either A/B cis or A/B trans A/B structure). The side chain of each of the four

skeletons may end in either a primary alcohol group or a carboxyl group. As noted above, C24 bile

alcohols are very rare in nature. Thus, to simplify bile acid and alcohol nomenclature, it is useful to divide

all “cholanoids” and “cholestanoids” into only three great classes – C27 bile alcohols, C27 bile acids and

C24 bile acids. In each of these classes, there are 5α and 5β epimers. Within these three great classes, and

for a given A/B ring junction, the pattern of hydroxylation – on the steroid nucleus and side chain or both

-- defines each individual bile alcohol or bile acid.

C. Hydroxylation sites

1. The steroid nucleus

a. C24 primary bile acids

Bile acid nomenclature, i.e. the trivial names of bile acids, is awkward because it is based on cholic

acid. Ox bile from the slaughter house was readily available to chemists, and Strecker is given credit for

isolating cholic acid and obtaining the correct empirical formula of C24H40O5 by combustion analysis in

1848 (112). Mylius isolated DCA in 1886 and the term “deoxy” was attached as a prefix because DCA

had one less oxygen atom by combustion analysis (113). When CDCA was isolated from goose bile, it

was named Chenosäure (cheno acid) for its animal source of origin (cheno denotes goose in Greek)

(114), and some years later the adjective “deoxy” was added, based on its elemental composition, giving

rise to the polysyllabic name of chenodeoxycholic (acid). Other trihydroxy bile acids were named for

their animal source: muricholic acid (from rodents), and hyocholic acid (from pigs).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

19

We have proposed (82) that the root C24 bile acid should be CDCA, as every bile acid must have a

hydroxyl group at C-3 (from the C-3 hydroxyl group of cholesterol) and a hydroxyl group at C-7, as 7α-

hydroxylation of cholesterol is the rate limiting step in the major pathway of bile acid biosynthesis. Thus,

all natural primary C24 trihydroxy bile acids can be considered as derivatives of CDCA. The only

exception to this useful convention occurs in those few mammals (nutria, beavers, bears, and some

cavimorphs) where UDCA, the 7β-hydroxy epimer of CDCA, is a primary bile acid. The route of

formation of this 7β-hydroxy epimer of CDCA is unknown. One can speculate on the existence of a

cholesterol 7β-hydroxylase or a pathway that results in the epimerization of the C-7 hydroxy group.

The most frequent site (in terms of the number of vertebrate species) of additional hydroxylation of

CDCA is at C-12 in cholic acid. The second most common site of hydroxylation is probably at C-16,

because so many avian species have the 3α,7α,16α-trihydroxy bile acid as a major biliary bile acid (82).

Another common site of hydroxylation is at C-6, occurring in hyocholic acid (pigs) and the three

muricholic acids (rats and mice). Other “third site” (nuclear) hydroxylations occurring in biliary bile

acids of the healthy animal are at C-1 (α or β), C-2 (β), C-4(�), C-5(β), C-9(β), C-11(α), C-15(α), and C-

19 (β). Table 1 summarizes the species in which these rare biliary bile acids occur, and the appropriate

citation.

C24 bile acids with four hydroxyl groups on the steroid nucleus occur rarely. Some snakes have

3α,7α,12α,16α-tetrahydroxy bile acids (82). Tetrahydroxy bile acids of unknown structure are found in

trace proportions in the bile of many vertebrate species. After experimental ligation of the common bile

duct, tetrahydroxy- and even penta-hydroxy C24 bile acids occur in the plasma and urine of mice (115).

Such bile acids also occur in mice when the canalicular bile salt export pump has been deleted genetically

(116).

An ad hoc committee of bile acid workers met in Freiburg in 1980 under the auspices of the Falk

Foundation. The group proposed working rules for bile acid nomenclature and bile acid abbreviations

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

20

that were published and noted by the IUPAC nomenclature committee. At the suggestion of Gerhart Kurz

of the University of Freiburg, it was agreed that there would be no more trivial names, except for

conversational purposes (117).

b. C24 secondary bile acids

As noted before, bile acids are modified by bacterial enzymes in the distal intestine. Modification

of the hydroxyl groups forms new bile acids that are termed “secondary”. This distinction has limitations,

because bile acids such as UDCA or certain of the hydroxy-oxo bile acids can be both primary and

secondary. A consideration of secondary bile acids is important because they can be major biliary bile

acids. After their formation in the distal intestine, they are absorbed in part. They return to the liver in

portal venous blood, and traverse the hepatocyte where they may or may not undergo further metabolism

before joining the pool of primary bile acids (formed de novo from cholesterol) in the enterohepatic

circulation.

The major bacterial biotransformations of conjugated bile acids are deconjugation, 7-

dehydroxylation, oxido-reduction, epimerization, and side chain desaturation (detailed in 110).

Deconjugation, i.e. hydrolysis of the amide bond between the terminal carboxylic acid group of the bile

acid and the amino group of taurine or glycine, is mediated by many bacterial species and does not

require anaerobic conditions. Unconjugated bile acids formed by bacterial deconjugation are still

considered primary bile acids.

In the large intestine, anaerobic modifications of the hydroxyl groups occur. The most striking change is

7-dehydroxylation that occurs via a 3-oxo-Δ4,6 nucleotide intermediate (110). The 7-dehydroxylation

product of cholic acid is DCA; that of CDCA or UDCA is lithocholic acid. The 7-deoxy derivative of

hyocholic acid and ω-muricholic acid is termed hyodeoxycholic acid and that of α- and β-muricholic acid

is termed murideoxycholic acid. To date, the C-7 deoxy derivative of “avicholic” (3α,7α,16α-trihydroxy)

has not been identified, but bacterial 16-dehydroxylation has been reported for some steroids (118), and is

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

21

likely to occur in some birds. Such 16α-dehydroxylation would result in the formation of CDCA, that in

turn would likely undergo 7α-dehydroxylation to form lithocholic acid.

During transit through the large intestine, any hydroxyl group can be oxidized and then reduced. If the

reduction is to the other possible epimer, a new epimer will be formed. 3β-Hydroxy bile acids (“iso” bile

acids) are major fecal bile acids in man (119). 7β-Hydroxy bile acids and even 12β-hydroxy bile acids

are also formed, but to a far lesser extent. Elimination of the 3-hydroxy group with formation of a Δ2 or

Δ3 bile acid may also occur (120).

Secondary bile acids may undergo additional hydroxylation during hepatocyte transport. Such

rehydroxylation at C-7 generates the original primary bile acid that had undergone bacterial 7-

dehydroxylation in the intestine. This rehydroxylation does not occur in man and the cow, occurs to a

limited extent in the rat and hamster, and is complete in the prairie dog (121). Alternatively, secondary

bile acids may undergo hydroxylation at a new site, generating a novel “tertiary” bile acid. One example

is 16α-hydroxylation of DCA to form “pythocholic” acid (3α,12α,16α- trihydroxy) that occurs in the

python (75). In the wombat, lithocholic acid undergoes hydroxylation at C-15 (122). “Iso” (3β-hydroxy)

bile acids are epimerized to 3α-hydroxy bile acids during transport through the hepatocyte (123,124).

Biotransformation of the 3β-epimers of the muricholic and hyocholic acid families has not been reported,

but is likely to occur. In man, the 7β-hydroxy group of UDCA does not undergo epimerization during

transport through the hepatocyte, and the small proportion of UDCA in the biliary bile acids is likely to be

formed from CDCA in the distal intestine by bacteria. Bacterial modification of bile acids and subsequent

hepatic biotransformation has been termed by us, “damage and repair”.

The percent of secondary bile acids in biliary bile acids is usually less than 50%, but there are

exceptions. The most remarkable example is in the rabbit, in which DCA constitutes 80% of the

circulating bile acids (82). In some snakes, the 3α,12α,16α-trihdroxy bile acid, which can be formed by

16α-hydroxylation of DCA, exceeds 50% (82).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

22

c. C27 bile acids

Nuclear hydroxylation in C27 bile acids occurs at C-3 and C-7 (the default structure), and also at C-12 in

most species. In the Andean condor, hydroxylation at C-16 occurs (82). We have identified a 1β,3α,7α-

trihydroxy C27 bile acid in the bile of the tinamou, an ancient flightless bird (125). A tetrahydroxy bile

acid with hydroxyl groups at C-3, C-7, and C-12 plus a 2β-hydroxy group has been isolated from the bile

of the large freshwater fish Arapaima gigas (126). It seems likely that there are additional sites of nuclear

hydroxylation in C27 bile acids yet to be discovered.

C27 bile acids undergo 7-dehydroxylation in the alligator (97), but the extent of 7-dehydroxylation of

C27 bile acids in other species has received little attention.

d. C27 bile alcohols

In C27 bile alcohols, default nuclear hydroxylation occurs at C-3 and C-7; additional hydroxylation

occurs at C-12, and rarely at C-6 or C-16 (25,26, 127). The C-3 hydroxyl group has a β-configuration in

latimerol and myxinol, meaning that it remains unchanged from that of cholesterol. Hydroxylation at C-

16 occurs in myxinol. The bile of Arapaima gigas contains a bile alcohol with a β-hydroxy group at C-2

in addition to hydroxyl groups at C-3, C-7, and C-12, so that C27 bile alcohols with four hydroxyl groups

on the nucleus do occur (126).

No C27 bile alcohols are recognized as “secondary”, i.e. with hydroxyl groups modified by bacterial

enzymes. However, the presence of 7-deoxyscymnol in the Megamouth shark, as reported by Ishida et al

(128), suggests that bacterial dehydroxylation at C-7 may also occur for C27 bile alcohols.

. 2. The side chain

a. C24 bile acids

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

23

With only four available carbons in the side chain, C24 bile acids are likely to undergo side chain

hydroxylation only at C-23 or C-22, each of which has potential R and S epimers. The most common C-

23 hydroxy bile acid is termed “phocaecholic acid” and has the nucleus of CDCA. (The name “phoca” is

given to the marine mammal family from which Hammarsten (129) isolated the bile acid). The structure

of phocaecholic acid was later established as the 23-hydroxy derivative of CDCA by Windaus and van

Schoor (130). The R and S epimers have been synthesized (131,132) and the R form identified as the

naturally occurring epimer (131). In addition to marine mammals, phocaecholic acid also occurs in

wading birds and some song birds (82). The 12-hydroxy derivative of phocaecholic acid also occurs in

marine mammals and is therefore a natural tetrahydroxy- bile acid.

A 3α,7α-dihydroxy bile acid with a hydroxy group at C-22 was shown to occur in the fish

Parapristipoma trilineatum by the Hoshita laboratory (133) and given the awkward name of

haemulcholic acid. The compound was synthesized by both the Hoshita laboratory (134) and the

Pellicciari laboratory (135), and the S epimer was shown to occur in nature. Some of its physicochemical

and biological properties were characterized by Roda et al (135). Haemulcholic acid was also identified

in freshwater fish by Haslewood during his Zaire River expedition (75).

Side chain desaturation forming Δ22 bile acids is a major pathway in rats (136,137) and cavimorphs

(82). Such bile acids could arise from further β-oxidation of the side chain in the liver and/or by bacterial

action in the distal intestine.

b. C27 bile acids

Elucidation of the structure of the C27 bile acids with hydroxyl group substituents on the C8 side chain

was not simple, as side products such as lactones may be generated during alkaline hydrolysis.

Unfortunately, an enzyme preparation that hydrolyzes the amide bond of C27 taurine conjugated bile acids

is not available at present (In contrast, cholylglycyl hydrolase, which rapidly cleaves the amide bond in

C24 conjugated bile acids, is available commercially).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

24

So far, all reported C27 bile acids have only a single hydroxyl group in the side chain. It occurs at C-22

in turtles, at C-24 in lizards, and at C-26 in bullfrogs (25, 26); each of these bile acids has four potential

epimers. The four cholestanoic acid epimers with a hydroxyl group at C-24 have each been synthesized

by Une et al. (138). The most common diastereoismer is varanic acid, which has the 24R, 25R

configuration. In collaboration with the Iida and Une groups, we have found that the 24R,25S epimer is

present in the bile of several varanids (139).

c. C27 bile alcohols

As noted earlier, Hammarsten determined that he had isolated a C27 bile alcohol from the bile of the

Greenland shark (100). The challenge of determining the structure of this compound would occupy

chemists for many decades. Windaus et al. (140) initially took up the problem of determining the

structure of Hammarsten’s compound (named scymnol) and proposed that it contained one primary and

three secondary hydroxyl groups, with an epoxy ring. The three secondary hydroxyl groups were found

to be located in the sterol nucleus (at positions 3α, 7α, and 12α) as scymnol could be converted into cholic

acid by partial oxidation of the side-chain (141,142). Since the side-chain of cholesterol has a C-27-

methyl group, different paired combinations of carbons 23, 24, and 25 were proposed to contain the

epoxy group, with the primary alcohol group being on C-27. Bridgwater and Haslewood showed (using

Hammarsten’s original sample) that the epoxy group was an artifact of alkaline hydrolysis, and that the

side-chain of scymnol actually contained two secondary hydroxyl groups and a primary hydroxyl group

bonded to sulfate (143,144). In 1961, Cross used 1H-NMR to elucidate the structure of both scymnol and

anhydroscymnol, the epoxide formed during alkaline hydrolysis (145). He found that the side chain

hydroxyl group of scymnol was unexpectedly on C-26 (the only carbon with a single proton), consistent

with a side-chain structure having hydroxyl groups at C-24 and C-26, and a primary hydroxyl group at C-

27. The structure of scymnol was ultimately established as 3α,7α,12α,24,26,27-hexahydroxy-5β-

cholestane by comparison with a synthetic standard (143). Carbon-24 is chiral, and the configuration of

the 24-hydroxyl group was determined to be 24R, in the crystallographic study of Ishida et.al. (146). This

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

25

group has also found that whereas most shark species have the (24R,25R)-epimer of scymnol, a few

species also form the (24R,25S)-epimer (147). Other named C27 bile alcohols are tabulated in our

previous review (82). Additional information is available in the cogent review of Une and Hoshita (26).

Bile alcohols, just as bile acids, have been named for the species in which they occur, and the name

gives no clue whatsoever to the structure of the bile alcohol. It is convenient to think of the trihydroxy

bile alcohol with α-hydroxyl groups at C-3, C-7, and a primary alcohol function at C-27 as the “root” C27

bile alcohol, and to look on other bile alcohols as derivatives of that simplest bile alcohol. At present, this

“root” bile alcohol has no trivial name, but has been synthesized by the Mosbach group (148).

D. Conjugation

1. N-acyl amidation with taurine or glycine.

In virtually all species, bile acids, whether C24 or C27, are found in bile in N-acyl amidated form, i.e.

conjugated in amide linkage with taurine (or a taurine derivative) or far less commonly with glycine.

Taurine conjugation is the rule in fish, amphibians, reptiles, and most avian species. It also occurs to a

variable extent in mammals (82). In some angelfish species, N-methyltaurine conjugates are present

(149), the N-methyltaurine thought to be of dietary origin. Similarly, in some marine fish, bile acids are

conjugated with cysteinolic acid, a hydroxy-ethyl derivative of taurine that is of dietary origin (150).

Glycine conjugation occurs in a few avian species, some bovids, cavimorphs, and primates, including

humans (82). Sarcosine (N-methylglycine) conjugates have been synthesized (151,152,153) and shown to

be more resistant to bacterial deconjugation during enterohepatic cycling (154). Some workers have

considered bile acid amidation to be the final step in bile acid biosynthesis (155). It seems more logical to

us to define the bile acid biosynthesis pathway as ending with a mature bile acid or bile alcohol before it

undergoes a phase II biotransformation step such as N-acylamidation, sulfation, or glucuronidation.

The process of conjugation occurs both in hepatocytes and in cholangiocytes (156). However,

conjugation in the hepatocyte greatly exceeds that occurring in cholangiocytes. The conjugation process

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

26

begins with the formation of a bile acid adenylate (157) followed by formation of the coenzyme A thio-

ester. A bile acid aminotransferase then transfers the CoA bile acid to the amino group of glycine or

taurine; the human enzyme has been cloned and shown to mediate conjugation with both glycine and

taurine (158). Conjugation of newly synthesized bile acids occurs in peroxisomes (159), and recent work

suggests that “reconjugation" of unconjugated bile acids returning from the intestine also occurs in these

organelles (160). Other than their pKa value, which in turn is responsible for the pH-solubility profile (see

later), there is little difference in the biological properties of glycine and taurine conjugated bile acids,

except that taurine conjugates are deconjugated by bacteria more slowly than glycine conjugates

(161,162).

That bile acids are present in bile in “conjugated” form was recognized in the 19th century. At that

time, neither the structure of the bile acid nor the site of linkage was known. This resulted in bile acid

conjugates having a misleading name, in that “taurocholate” implies that the cholic acid moiety is capable

of ionization. We and others have suggested that the name “cholyltaurine” (analogous to glycylglycine)

would be more appropriate (117), but few investigators have followed this proposal. The extremely low

proportion of unconjugated bile acids in biliary bile acids has led some investigators to conclude that the

conjugation process is highly efficient. However, unconjugated bile acids, especially dihydroxy bile

acids, if secreted into bile are rapidly absorbed by the biliary ductules (163,164), resulting in their

removal from ductal bile. Thus, the state of conjugation in canalicular bile can be considerably less than

that of ductal bile.

2. Other types of bile acid conjugation

When the term “conjugated” was applied to bile acids, it denoted N-acyl amidation with taurine or

glycine. When bile acid sulfation and glucuronidation were discovered to be important pathways under

some circumstances, it was necessary to distinguish different modes of “conjugation” for bile acids. In

principle, there are six modes of bile acid conjugation: N-acyl amidation (using glycine, taurine, or

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

27

possibly other amino acids), sulfation, glucuronidation, conjugation with glutathione, and conjugation

with N-acetylglucosamine or glucose, and bile acids may have more than one mode of conjugation. In

vertebrates, the only bile acid sulfates that occur in appreciable proportions in biliary bile acids are the 3-

sulfate of lithocholylglycine and lithocholyltaurine amidates that occur in man (165) and probably great

apes (166). In patients with severe cholestasis, bile acid sulfation (mostly at C-3) followed by urinary

excretion becomes a major pathway (167,168). In the bile duct ligated hamster, esterification with sulfate

occurs at C-7 (169).

In the Mayo Clinic studies of Thistle and Schoenfield, in which bile acids were administered to

gallstone patients (35), one group received hyodeoxycholic acid (3α,6α-dihydroxy-. Since

hyodeoxycholic acid should be absorbed efficiently from the small intestine by passive mechanisms, its

absence in biliary bile acids was completely mystifying. In 1978, B. Almè and J. Sjövall reported a

patient with malabsorption who excreted in urine 6-hydroxy bile acids that were present as O-linked

(ethereal) glucuronides. (170). Five years later, Sacquet et al, working in the laboratory of Reca Infante in

Paris, reported that when hyodeoxycholic acid was administered to human subjects, it was largely

excreted in urine as its glucuronide (171), explaining its absence in biliary bile acids. This group did not

know the site of glucuronidation but proposed that it was at C-3.

Four years later, Anna Radominska-Pyrek and colleagues showed that human microsomes rapidly

glucuronidate hyodeoxycholic acid at C-6 (172). Thus the curious fate of hyodeoxycholic acid in humans

was finally settled. It presumably results from defective N-acylamidation, and hyodeoxycholic acid is

consequently metabolized in the same manner as a C24–nor (C23) dihydroxy bile acid (see below), i.e. by

glucuronidation in the endoplasmic reticulum. To date, this pathway has not been identified in any other

animal. When hyodeoxycholic acid is given to rats, it is probably conjugated with taurine and not

glucuronidated (173).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

28

When C23 dihydroxy-bile acids, are administered to animals including man, little amidation occurs, as

both the CoA ligase and the glycine/taurine amino acid transferase act to only a very limited extent on bile

acids with a shortened (C4) side-chain (174). Such bile acids undergo glucuronidation both at C-3 and C-

23 (163,164). In man, when norUDCA is given, it forms the C-23 ester glucuronide which is excreted in

both bile and urine (175). In rats, N-acyl amidation is also much less efficient for α-hydroxy bile acids

(131,132) and hydrophilic epimers of cholic acid, for example 3α,7β,12β-trihydroxy-cholanoate (176).

In patients ingesting UDCA, UDCA also undergoes conjugation with N-acetylglucosamine at C-7, in

addition to amidation with glycine or taurine at C-24 (177). In Niemann-Pick disease type C, a novel Δ5

3β,7β-dihydroxy cholestanoic acid is formed (178); this compound may be considered an acidic 7β-

hydroxy derivative of cholesterol. This compound undergoes sulfation at C-3, conjugation with N-

acetylglucosamine at C-7 (178), and amidation with glycine or taurine at C-24. In both circumstances,

the multiply-conjugated metabolites are eliminated in urine rather than bile, probably because they are not

substrates for the canalicular transporters. For UDCA, conjugation with N-acetylglucosamine is a very

minor pathway—less than 0.3% of the oral dose.

Glutathione conjugates of bile acids (presumably as thio-esters) have been identified in rat and human

infant bile by the group of Shigeo Ikegawa, but their concentration is several orders of magnitude less

than that of the major biliary bile acids (179).

To date all C27 bile acids have been found to be conjugated exclusively with taurine. Bile alcohols are

esterified with sulfate, and thus have a pKa similar to that of taurine conjugates.

E. Structure-function relationships.

In the steady state adult animal, cholesterol input – from de novo synthesis and dietary cholesterol –

must be balanced by excretion – cholesterol as such and bile acids. There is thus a mole fraction of total

cholesterol excretion composed of bile acids, fecal acidic steroids, divided by total (neutral plus acidic)

steroids. In man, based on balance studies, this figure is 0.46; in the mouse it is 0.45, whereas in the

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

29

hamster it is 0.57, according to John Dietschy and Steve Turley (180). The very low cholesterol

proportions in the biliary lipids of many vertebrate species (181), suggests that in most vertebrates this

number should be well above 0.5. The route of bile acid excretion is solely via bile, as the amount of bile

acids in urine is negligible. In patients with complete biliary obstruction, fecal bile acid output is

extremely low, indicating that possible bile acid extruders in the small intestine epithelium are not

upregulated (182). In patients with biliary obstruction, bile acid elimination is by urinary excretion, albeit

with greatly increased levels in plasma and liver. In addition to cholesterol excretion in bile (as such and

after conversion to bile acids) there is a small flux of cholesterol directly from the small intestine (183).

For a bile acid to serve as the chemical form of cholesterol elimination there are only a few molecular

requirements. The molecule must be too polar to be absorbed passively and too large to pass through the

paracellular junctions between enterocytes. It also must be resistant to pancreatic enzymes.

The bile acid molecule is too large to pass via the tight junctions of the intestinal epithelium.

Amidation with glycine or taurine converts unconjugated bile acids from weak acids (pKa of 5) to a pKa of

4 for glycine amidates and a pKa of less than 2 for taurine conjugates (184). Thus, bile acids are present in

bile and intestinal content as organic anions and therefore will not cross membranes unless a transporter is

present. Bile acid glycine and taurine amidates are resistant to pancreatic carboxypeptidases, whereas bile

acids amidated with amino acids other than glycine or taurine (with the exception of aspartic acid and

cysteic acid) , are hydrolyzed rapidly by pancreatic carboxypeptidases (185).

In vertebrates with a cecum, deconjugation and dehydroxylation convert bile acids to membrane

permeable molecules and they are in part absorbed, as signaled by the presence of unconjugated bile acids

in the systemic circulation. Isao Makino, who had worked in the Sjövall laboratory in Stockholm and

returned to Japan, was among the first to report the presence in healthy individuals of unconjugated bile

acids in plasma using gas chromatography (186). Some decades later, this early finding was confirmed

by Kenneth Setchell et al, using gas chromatography-mass spectrometry (187). Nonetheless, absorption

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

30

from the cecum is not complete, as unconjugated bile acids have poor solubility at the acidic pH (pH 5.5)

of the cecum and are bound to bacteria and unabsorbed dietary components.

Unconjugated (and conjugated) bile acids that are not absorbed are finally eliminated from the body by

the process of defecation. As there is negligible excretion of bile acids in urine, breath, and skin, and as

the bile acid nucleus is considered to be indestructible by bacterial enzymes, fecal excretion of bile acids

is equal to their synthesis from cholesterol. However, it must be stressed that there is little information on

the fecal bile acid composition across the vertebrate spectrum (188). There are some species that by our

analyses have very low fecal bile acids, suggesting that the bile acid molecule can in fact be degraded by

bacteria. The degradation of bile acids by individual bacterial strains with bile acids as the major carbon

source has been characterized in great detail by S. Hayakawa (189). The biological relevance of these

studies is unclear. To date, there are no studies in which bile acids tagged with 14C in the steroid nucleus

are administered and 14CO2 in breath is collected.

There is very little information on the fate of bile alcohols in the distal intestine. In the Asiatic carp we

found unchanged 5α-cyprinol sulfate in the distal intestine (190). Even if there were hydrolysis of the

ester sulfate, the liberated bile alcohol is likely to have negligible passive membrane permeability because

of the number of hydroxyl groups.

For solubilizing lipids in the form of mixed micelles, bile acids must be amphipathic, i.e. they must

have a polar side and a non-polar side. So far as is known, all major natural biliary bile acids and bile

alcohols are amphipathic, although the hydrophobic side (beta face) is somewhat reduced in UDCA.

Figure 5 shows a space-filling model of a bile acid molecule, emphasizing its amphipathic nature. To the

best of our knowledge, no one has synthetized a molecule unrelated to the bile acid molecule which has

the same physicochemical properties of bile acids as regards solubilizing membrane lipids.

III. Measurement

A. Isolation and chromatographic separation

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

31

Elucidation of the metabolism of bile acids in health and disease requires accurate, reliable, and

sensitive methods for measurement, as well as techniques for extracting bile acids (or alcohols) from

complex biological matrices. The complexity of bile acid structure and the enormous range in polarity of

bile acids and their conjugates have always proved a challenge to the analytical chemist. A cogent review

by W. Griffiths and J. Sjövall on current analytical approaches was published in 2010 (9). These two

chemists have contributed greatly to the advances in bile acid analysis.

The modern era of bile acid analysis began with the development of improved chromatographic

methods for separation. Paper chromatography based on liquid-liquid partition was developed in the

1940’s by the great English chemists A.J. P. Martin and R.L. Synge who shared the Nobel Prize in 1952.

In the 1950’s it was first applied to bile acids by Jan Sjövall to separate individual unconjugated bile acids

(191). Sjövall and Haslewood then collaborated to apply the method to Haslewood’s vast collection of

bile samples from different animals (192).

Gas-liquid (partition) chromatography was developed some 10 years later by A.J.P Martin working

with A.T. James. The earliest application to bile acids came in the 1960’s (193,194), and Eneroth and

Sjövall tabulated retention times for different stationery phases in 1971 (195). A decade later capillary

gas chromatography was described, greatly increasing resolving power. Gas chromatography using flame

ionization detectors affords both separation and quantification. However, only when a peak is further

analyzed by mass spectrometry can one be one certain that the peak is truly a bile acid.

Thin-layer adsorption chromatography (TLC) became widely used when a commercial device for

coating the adsorbent (usually silicic acid or alumina) on glass plates became available and excellent

adsorbents were developed (196). TLC offered useful separations of bile acids by class (taurine

conjugates, glycine conjugates, unconjugates) but could not separate individual taurine dihydroxy-

conjugates from each other. For unconjugated bile acids, molecules separate according to the number and

orientation of hydroxyl groups, but a clear separation of DCA from CDCA with the commonly employed

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

32

solvent systems does not occur. Moreover, quantification of individual spots was difficult. Nonetheless,

TLC is a highly useful quasi-quantitative method for assessing purity of bile acids and monitoring

chemical reactions.

The senior author was privileged to have access to the first TLC apparatus to enter Sweden,

purchased in 1960 by Borgström for the separation of lipids. Borgström had been impressed with the

remarkable separations of neutral lipids that were obtained using this technique by Malins and Mangold

(197), then working at the Hormel Institute in Minnesota. With TLC using silicic acid as the stationery

phase and phosphomolybdic acid (in ethanol) as the detection reagent most bile acids (as well as

unsaturated lipids) appear as deep blue spots on a brilliant yellow background (the colors of the Swedish

flag). A technique was described for coating silicic acid on microscope slides (198), and this in turn

permitted screening of multiple solvent systems for optimal bile acid separation. Systems were developed

(199) that our laboratory still uses today.

Reversed phase TLC has also been used for bile acids (200,201), but has not achieved popularity

because of the much lower capacity of the method, as well as difficulties in detecting bile acids after

separation. Eneroth and Sjövall tabulated RF values using TLC for a large spectrum of bile acids and a

number of solvent systems in their review in 1971 (195). Some of their values have been shown as actual

simulated chromatograms in a chapter by the senior author (202).

In our laboratory, we have found TLC to be of great value for quantifying hepatic biotransformation of

radioactive bile acids. Small (1 mm) bands of adsorbent can be directed into scintillation counter vials,

using the automatic device described by Snyder and Kimble (203), or manually, by transferring the

adsorbent to a vial using a brush.

HPLC was developed in the 1970’s and soon applied to bile acids. Today, the technique when used for

bile acid analysis is usually executed in the reversed phase mode, i.e. with C18 or C8 bonded silicic acid as

the lipophilic solid phase. Numerous laboratories have reported the power of this analytical method for

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

33

separating and measuring conjugated bile acids in bile and intestinal content (9). Conjugated, i.e. N-acyl

amidated bile acids have an amide bond that has absorbance at 205 nm, permitting their measurement in

the effluent. Unconjugated bile acids, non-amidated bile acid sulfates, and bile alcohol sulfates are not

detected at 205 nm. HPLC with UV detection is too insensitive for analysis of serum bile acids unless

large volumes of serum are used (204). One solution to the non-detection of unconjugated bile acids and

bile alcohol sulfates is to use a light scattering detector that responds approximately equally to all bile

acids (205). A second approach, useful for unconjugated bile acids, is to couple the bile acid to a

chromophore such as the phenylacyl group (206).

In our laboratory, we developed an HPLC method for separating the 12 conjugated bile acids

predominant in human bile (165), and we have found this method to be essential in our analysis of

vertebrate bile samples (82). Nonetheless, when used alone for bile analysis, there are many unknown

peaks that may or may not be bile acids. Therefore peak identification requires confirmation by mass

spectrometry

B. Mass spectrometry

Mass spectrometry had been developed in the 1930’s and was used to analyze mixtures of

small molecules. Mass spectrometry of large organic molecules emerging from the gas chromatograph

was developed by R. Ryhage in Sweden, and the first commercial instrument for this purpose was

marketed by LKB in 1965. William Elliott wrote in 1988, “Two years later [1965], I returned to the

Karolinska to devote the summer to learning about the GC/MS which Ragner Ryhage had built, and was

then produced by LKB. Our LKB 9000 is still operating, beginning its 23rd year.” Since then, there have

been continuous improvements in techniques for sample volatilization and instrument sensitivity,

culminating in the MALDI analysis of proteins. In our opinion, anyone who wishes to do a complete bile

acid analysis must have HPLC/MS/MS available. It is also desirable to have GC/MS availability when

needed, as capillary gas chromatography has greater resolving power than HPLC.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

34

The wedding of HPLC and MS/MS is thus an essential step in the analysis of unconjugated and

conjugated bile acids. Currently, internal standards containing three or more deuterium atoms are not

available for many bile acids and are sorely needed. For a total analysis, class separation by ion exchange

chromatography developed by Setchell and Sjövall should be performed before HPLC/MS/MS (9). For

state of the art analysis, either capillary GC or capillary HPLC will be required to separate complex

mixtures of bile acids. For unknown bile acids, structure assignment requires NMR for elucidation of

structure, and ultimately synthesis for confirmation of assigned structure.

C. Enzymatic and competitive binding assays

Development of an enzymatic technique for determining total 3α-hydroxy bile acids based on

oxidation of the 3α-hydroxy group by a 3α-hydroxy steroid dehydrogenase was a major advance in bile

acid analysis (207), especially for workers in the field of bile acid physiology. This technique was based

on the pioneering work of Talalay (208), who developed enzymatic analysis of 3α- hydroxy hormonal

steroids using a steroid dehydrogenase isolated from Pseudomonas testosteroni. Details of the method

were clarified by Turley and Dietschy (209). The sensitivity of the method was increased by measuring

the reduced NAD by changes in fluorescence or by coupling the oxidation to reduction of a tetrazolium

dye (210). The enzyme technique using 3α-hydroxysteroid dehydrogenase does not measure 3β-hydroxy

bile acids. Thus, when used for determination of fecal bile acids, it will underestimate total bile acids

(119). An enzyme recycling system has recently been marketed by Diazyme™ and is quite sensitive.

In our laboratory, based on collaboration with Marlene DeLuca. Aldo Roda developed an enzymatic

bioluminescence assay for primary bile acids. The method was based on co-immobilizing on beads a 7α-

hydroxysteroid dehydrogenase, an oxido-reductase, and luciferase (211). The method was validated by

showing excellent agreement with GC/MS measurements (212). However, this method, despite its

simplicity and sensitivity, has not been widely adopted. Limitations in the enzymatic method are

summarized in the review by Griffiths and Sjövall (9).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

35

All of the chromatographic techniques discussed above are labor-intensive and time-consuming. For

measuring diurnal variations in plasma levels of total bile acids, two approaches have been taken to

develop a rapid, simple procedure for measuring total bile acids. The first was the enzyme method that

measures total 3α-hydroxy bile acids, as just discussed. The second approach was the development of

competitive binding techniques, such as radioimmunoassay. What is measured depends on the affinity of

the antibody, and thus one determines “immunoreactive” bile acids. At the Mayo Clinic, Wilfred

Simmonds et al developed a radioimmunoassay specific for cholyl conjugates (213) which was then used

by LaRusso et al to define the time course of the postprandial elevation of such cholyl conjugates in

health and in patients with bile acid malabsorption (214). Later, Schalm et al (215) developed a

radioimmunoassay for chenodeoxycholyl conjugates, and used it to show that the postprandial peak of

CDC conjugates occurs earlier than that of cholyl conjugates (216), as shown in Figure 6. Other

radioimmunoassays have been developed, as summarized in the review by Griffiths and Sjövall (9).

All of the above methods for measuring bile acids isolate the bile acids from the body tissue or fluid,

and then quantify the bile acids. We have long needed a bile acid-sensitive molecule whose signals would

provide information on the concentration of bile acids within the living cell. This need has now been

supplied by the development of an indicator molecule consisting of a protein possessing a bile acid

binding site (based on the binding site of FXR) and two reporter molecules whose fluorescence is

determined by the extent to which bile acids occupy the binding site -- the greater the binding, the less the

fluorescence. The technique, as described by van der Velden et al (217), uses the well-known Forster

Resonance Energy Transfer (FRET) principle, and has already been used to characterize bile acid

transport into isolated hepatocytes.

D. Diagnostic utility of plasma bile acid levels

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

36

Despite the simplicity of competitive binding techniques for measuring conjugated bile acids, they

have not been widely adapted by the clinical chemists. The measurement of plasma bile acids is not part

of the usual panel of liver tests (aminotransferases, alkaline phosphatase, total bilirubin). The major

reason is that bile acid levels are probably inferior in sensitivity and specificity in detecting liver injury

when compared to the standard liver panel. This conclusion is based on clinical studies (218,219). In

addition, a recent animal study (220) in which liver injury was experimentally induced in rats by feeding a

variety of toxic molecules found that serum bile acid levels were not more sensitive than aminotransferase

levels. Thus, because of the lack of greater sensitivity of serum bile acid measurements, there is the belief

that the current liver tests are adequate. Moreover, the instruments that automate determination of a

serum “chemical panel” do not have a bile acid “channel”. For purely cholestatic diseases such as

primary cholestasis of pregnancy, the level of serum bile acids has value for predicting prognosis (221).

E. Determinants of bile acid profiles in body fluids

In assessing the significance of bile acid concentrations that in turn define the bile acid profile, it is

important to realize that plasma, hepatic, biliary, urinary and fecal bile acids all have a different

composition in health. Plasma bile acids, in health, represent the balance between instantaneous intestinal

input and net hepatic uptake, with first pass clearance ranging from 40 to 90%, depending on the bile

acid. Hepatic bile acids (as distinguished from hepatocyte bile acids) represent the balance between input

of unconjugated and conjugated bile acids, subsequent biotransformation of these molecules, as well as

bile acids that have been secreted by transporters into the canaliculus. Urinary bile acids represent

filtered bile acids plus any bile acids secreted by the tubules minus those absorbed by the tubules. Ductal

bile which is usually analyzed differs from canalicular bile by that fraction of conjugated and

unconjugated bile acids that are absorbed by the biliary ductules. Gallbladder bile is ductal bile minus bile

acids absorbed from the gallbladder. Fecal bile acids represent bile acids entering the colon with or

without modification by bacterial enzymes minus those that were absorbed.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

37

The power of current analytical techniques may result in an enormous variety of bile acids being

detected. When such results are tabulated, we suggest organizing bile acids by primary bile acids, and

then listing secondary bile acids derived from the primary bile acid underneath their precursor. Bile acids

that are present in trace proportions should be lumped together to simplify the presentation. The choice of

the analytical method should be determined by the biological question that is being asked. For

nomenclature in reporting bile acid analyses, the recommendations of an ad hoc nomenclature committee

(117) should be followed.

IV. Bile Acid Metabolism: transport and the enterohepatic circulation

A. Bile acid transport

1. Overview

Glycine and taurine conjugated bile acids are fully ionized at the pH conditions prevailing in plasma,

bile, and small intestinal content. Therefore, these molecules will not cross a cell membrane and enter a

cell unless a transporter is present. What is responsible for the enterohepatic circulation is the vectorial

transport of conjugated bile acids through the ileal enterocyte and the hepatocyte. The enterohepatic

circulation of bile acids results from the action of two chemical pumps and three fluid flows. The

chemical pumps are the vectorial transport systems of the ileal enterocyte and the hepatocyte – each of

which has basolateral and apical transporters. The fluid flows are the aboral movement of intestinal

content due to intestinal propulsive activity, portal venous blood flow that transports bile acids from the

intestine to the liver, and biliary flow that returns the bile acids to the intestine. The molecules that

constitute the circulating pool of bile acids must be substrates for the vectorial transport systems of both

the ileal enterocyte and the hepatocyte in order to circulate enterohepatically.

The initial input into the hepatic limb of the enterohepatic circulation consists of primary bile acids

formed and conjugated in the pericentral hepatocytes. The newly synthesized conjugated bile acids are

pumped across the canalicular membrane and flow in canalicular bile from the pericentral region to the

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

38

periportal region, their direction of flow being opposite from that of sinusoidal blood. In the periportal

region, newly synthesized bile acids are joined by bile acids returning from the intestine and pumped into

bile.

The return pathway of the enterohepatic circulation features three inputs into the portal venous blood

from bile acids that have been absorbed by enterocytes. The dominant input into portal venous blood

consists of “undamaged” conjugated bile acids that have been absorbed by the ileal transport system. A

second input is that of unconjugated primary bile acids formed by bacterial deconjugation in the distal

small intestine and large intestine. A third input is that of “secondary (damaged) bile acids that are

formed by bacterial modification of the hydroxyl groups of primary bile acids. Only a fraction of the

secondary bile acids that are formed are absorbed, and those that are not absorbed are eventually

eliminated by defecation.

As discussed previously, those secondary bile acids that are absorbed from the distal intestine may

undergo uptake by the hepatocyte or in some cases, urinary excretion (or both). After hepatocyte uptake,

“damaged” bile acids undergo “repair” in the hepatocyte by “reconjugation” as well as re-epimerization

of hydroxy groups (from β to α) and reduction of oxo groups to hydroxy groups. They are then secreted

into bile to join the recycling primary bile acids. In man, the flux of bile acids returning from the intestine

is at least twentyfold greater than the input of newly synthesized bile acids. More details of bacterial

damage and hepatocyte repair are given in the introduction.

In man, the major secondary bile acids in colonic content are lithocholic acid, formed by 7-

dehydroxylation of CDCA, and DCA, formed similarly from cholic acid. In man, lithocholic acid is not

only N-acylamidated, but also sulfated in part. Such sulfo-lithocholyl amidates are secreted into bile, but

are poorly absorbed from the small intestine (222), so they do not have an enterohepatic circulation.

Those unsulfated amidates secreted in bile return to the liver, undergo sulfation in part, are re-secreted

into bile, and eliminated via the fecal route. The end result is that lithocholic acid has a very rapid

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

39

turnover in man, and sulfated and unsulfated lithocholyl amidates constitute less than 5% of biliary bile

acids (165). In rodents, lithocholic acid is not sulfated but undergoes hydroxylation at C-6 or C-7 (15).

As noted previously, DCA may pass through the hepatocyte without further metabolism, or may be

rehydroxylated to varying degrees in a species dependent manner. The “repaired” and reconjugated bile

acids are secreted from the hepatocyte and join the cycling conjugated bile acids.

Reabsorption of bile acids from the small intestine gives a cycling “pool” for each bile acid. The size

of the bile acid pool and its turnover rate differ for each bile acid, although the turnover rate of CDCA and

DCA are about the same. Most of each individual bile acid pool is stored in the gallbladder during

overnight fasting, and biliary bile acid composition is determined by the relative proportions of each type

of bile acids, this in turn being determined by the sizes of the individual bile acid pools. .

2. Transport by the ileal enterocyte: the apical transporter

The first person to observe preferential absorption of conjugated bile acids by the ileum was

Hermann Tappeiner, who presented his findings to the Vienna Academy of Sciences in 1887 (223).

Tappeiner studied the absorption of taurocholate from different regions of the small intestine using

anesthetized dogs. He found that taurocholate was absorbed preferentially in the ileum. Rather than

concluding that the ileum had special properties, Tappeiner was puzzled by the failure of the proximal

intestine to absorb taurocholate. Verzar, working in Hungary, authored one of the first monographs on

intestinal absorption (224). He was skeptical of the results of Tappeiner and directed his colleague

Fröhlicher to repeat Tappeiner’s experiments. Fröhlicher obtained identical results (225), again showing

that ileal absorption of conjugated bile acids was greater than that of jejunal absorption. However, in

Verzar’s book on intestinal absorption the concept of specific transporters with differing substrate

affinities is not considered. At that time, the mechanism of absorption of small molecules was considered

to be passive, analogous to diffusion.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

40

Baker and Searle, working at the University of Iowa, rediscovered the work of Tappeiner and

Fröhlicher, and confirmed the selective ileal absorption of taurocholate in the rat ileum in 1960 (226).

A decade before, T.H. Wilson, an American working in the laboratory of G. Wiseman at the University

of Sheffield, developed the everted gut sack technique and used this method to show that glucose was

actively transported (227). (It is rumored that this technique was the result of a drunken party in Paris

some years before, where physiologists were bemoaning the difficulty of studying intestinal absorption in

the intact animal. Someone, never identified, said the only solution was to turn the animal inside out.

And someone else is supposed to have said, “Maybe that’s a good idea!” In 1960, Robert Crane, who had

studied glucose absorption since his postdoctoral studies with the Cori’s in St. Louis, published his idea

that glucose absorption was sodium dependent, and that the downhill transport of sodium served to

energize the uphill transport of glucose (228). (The late Robert Crane was a tall, handsome charmer who

always had an eye for attractive women. At a party, he tried to persuade a lovely young scientist to go

home with him; when she refused, he started attacking her data).

In 1961, Leon Lack and Ike Weiner, young faculty members at Johns Hopkins, were discussing how to

teach medical students about the enterohepatic circulation. They realized that nothing was known about

the mechanism of bile acid absorption. They used the everted sac technique to show that the last fourth of

the small intestine of rats and guinea pigs actively transport conjugated bile acids against a concentration

gradient (229). Their data are illustrated in Figure 7. As noted earlier, the senior author, working with

his mentor, Bengt Borgström and Göran Lundh (a surgeon and brother-in-law of Borgström) perfused the

small intestine of healthy volunteers and obtained strong evidence for ileal absorption of conjugated bile

acids in man (20). Peter Holt, working in the laboratory of Kurt Isselbacher at Harvard, showed that

conjugated bile acid absorption in the everted sac preparation was sodium-dependent (230). Thus, it

seemed that a Na+ dependent transporter for conjugated bile acids was present in ileal enterocytes. (The

term “enterocyte” was coined by Christopher Booth, a prominent English gastroenterologist and

bibliophile, who discovered that vitamin B12, like conjugated bile acids, was also absorbed

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

41

predominantly in the ileum). In 1978, H. Lϋcke, G. Stange, R. Kinne and H. Murer, working at the Max-

Planck Institute in Frankfurt, prepared vesicles from ileal enterocyte brush borders, and used this

preparation to show that taurocholate uptake was carrier mediated and Na+ dependent (231). Attempts

were made to isolate the apical bile conjugated bile acid transporter by G. Kurz, working in Freiburg,

using the photoaffinity technique with 3H-tagged azido- bile acids (232). These studies were continued

by his student, Werner Kramer at the Hoechst Company in Frankfurt (233), but they failed to clearly

identify the ileal apical bile acid transporter.

During this time, ideas of cell polarity were developing (234). Thus, it seemed that at a minimum,

four transporters would be required for the enterohepatic circulation of bile acids. These would be an

apical and basolateral transporter in the ileal enterocyte, and an apical (canalicular) and basolateral

(sinusoidal) transporter in the hepatocyte.

The great breakthrough, in our judgment, came in 1987 with the use of expression cloning by Matthias

Hediger working in the laboratory of Ernie Wright at UCLA. Using the technique of injecting mRNA

into frog eggs (obtained from Xenopus laevis), they were able to report the cloning of the sodium coupled

transporter for glucose (235). Seven years later, Paul Dawson decided to attempt to clone the ileal bile

acid transporter. Dawson was well educated for the task, having worked with Joseph Goldstein, Michael

Brown, and David Russell at UT Southwestern in Dallas. Dawson, now relocated to Wake Forest

University, reported the successful cloning of the ileal apical sodium-dependent bile acid transporter

(protein, ASBT; gene, SLC10A2), using a somewhat different expression technique (236). This

achievement, more than a century after Tappeiner’s original study, permitted the testing of individuals for

defects in the gene encoding ASBT, as well as providing a molecular target for pharmaceutical companies

who might wish to develop an inhibitor.

In Dawson’s original paper, he found that ASBT was also present in the kidney, in agreement with

earlier studies by F. Wilson et al (237). Later, the laboratories of G. Alpini in Texas (238) and N. LaRusso

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

42

(239) at the Mayo Clinic showed that ASBT was also present in cholangiocytes. ASBT is highly specific

for bile acids, although the integrity of the 7α-hydroxyl group seems more important than that of the 3α-

hydroxyl group, as some substituents can be added to the 3α- position without impairing transport (240).

As yet, the transporter has not been crystallized, although the structure of a bacterial homologue has

recently been published (241). That paper proposes that two sodium ions act allosterically to open a

binding pocket. An overview of the SLC10 transporter family is available (242).

3. Transport by the hepatocyte: bile acid uptake

Bile acid uptake by the liver was known to be highly efficient. For example, in an isolated perfused

dog preparation studied at Mayo Clinic by N. Hoffman et al, uptake of taurocholate was 100% and that of

the taurine conjugate of CDCA was 70% (243). In a study published that same year from the Erlinger

laboratory in Paris, first pass extraction of taurocholate in the dog was found to be 40-80% and saturable,

consistent with uptake being carrier mediated (244). Conjugated bile acid uptake was saturable, and

using either hepatocytes (245) or the isolated perfused rat liver (245) was sodium-dependent (246). Thus

it seemed highly likely that one or more sodium-coupled transporters was present in the sinusoidal

membrane and mediated the efficient uptake of bile acids.

A sodium-dependent basolateral hepatocyte protein, named NTCP (sodium taurocholate porter) was

cloned in 1991, again using expression cloning, by Bruno Hagenbuch, Bruno Stieger and colleagues

working in the laboratory of Peter Meier in Zurich (247). When the Na+ coupled ileal transporter ASBT

was cloned four years later, it was possible to compare the sequences of NTCP and ASBT and identify

some regions of homology (248). Thus, the ileal uptake protein and the hepatocyte removal protein were

related, and both used the sodium gradient to power the uptake. However, NTCP has a much broader

specificity than ASBT (249,250). NTCP can be considered an anion transporter that transports not only

bile acids but also other organic anions into the hepatocyte from plasma, whereas ASBT selectively

conserves solely conjugated bile acids, thereby generating their enterohepatic circulation.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

43

NTCP is not the only bile acid transporter in the basolateral (sinusoidal) membranes of the hepatocyte.

A family of Na+ independent transporters, organic anion transport polypeptide (OATP) was identified, and

several of these multi-specific transporters have the ability to transport bile acids, as tabulated in cogent

reviews by Hagenbuch and Stieger (249) and Klaassen and Alaskans (250). In man, these include

OATP1A2, OATP1B1, OATP1B3, and OATP1C1. The genes encoding OATP transporters are denoted by

the term solute carrier organic anion (SLCO).

To what extent OATP transporters contribute to the highly efficient uptake of conjugated bile acids

from sinusoidal plasma is not known, but it is believed that the majority of bile acid uptake is mediated by

hepatocyte NTCP. The efficient uptake of dihydroxy conjugated bile acids is truly remarkable, as these

are 99% bound to albumin (251), and presumably uptake is from the extremely low concentration (nM) of

unbound species. In our view, it is reasonable to think that more than one transporter may be involved in

the sinusoidal uptake of conjugated bile acids. Presumably the transporters involved in bile acid uptake

have a zonal distribution and are enriched in periportal hepatocytes.

During enterohepatic cycling, there is continuous bacterial deconjugation in the small intestine

followed by hepatic reconjugation, presumably occurring in the periportal hepatocytes. This was first

recognized in rat studies by Norman, working in Lund, Sweden in 1955 (252) and much later confirmed

for man in our own studies at the Mayo (253). The rate of “reconjugation” in the periportal cells is

several fold greater than the rate of conjugation of newly synthesized bile acids in the pericentral cells.

The uptake of unconjugated bile acids is by both passive and carrier-mediated mechanisms, and

depends on the number and orientation of hydroxyl substituents. Lithocholic acid, CDCA, DCA, UDCA,

and presumably, 3,6-dihydroxy bile acids are highly membrane permeable (221,245) and in our judgment

do not require a transporter for rapid uptake into the hepatocyte. Cholic acid, in contrast, passes across

cell membranes slowly and requires a transporter for rapid cellular entry. A systematic study of the

passive membrane permeability of the three muricholic acid epimers is lacking.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

44

At least two transporters appear to be involved in the uptake of unconjugated trihydroxy- bile acids.

The laboratory of Curt Klaassen in Kansas City showed that OATP1B2 (a sodium-independent

transporter), appears to be involved in the uptake of unconjugated bile acids that return from the intestine

(254). Knockout of the transporter in mice caused a marked elevation in the plasma level of unconjugated

bile acids. Hubbard et al (255) showed that fatty acid transport protein 5 (FATP5) knockout mice have a

major fraction of unconjugated bile acids in bile, and such unconjugated bile acids were enriched in

secondary bile acids. They suggested that FATP5 was involved in both the uptake and CoA formation of

unconjugated bile acids returning from the intestine. It is attractive to postulate that this transporter

mediates not only adenylate formation but also subsequent CoA thioester formation of the unconjugated

bile acids that enter the hepatocyte. Steinberg et al (256) characterized a homolog of a peroxisomal very

long chain CoA synthetase. This homolog was present in the smooth endoplasmic reticulum. They found

that it had CoA synthetase activity for cholic acid, and postulated a role for this enzyme in cholic acid

reconjugation.

In the isolated perfused liver, rapid uptake of DCA occurs, presumably followed immediately by CoA

formation. Formation of the adenylate and subsequent CoA thioester serves to prevent back diffusion

across the sinusoidal membrane and thereby traps the bile acid CoA in the hepatocyte. In contrast, 24-

norDCA, which does not form a CoA thioester, regurgitates back from the hepatocyte into the perfusate

(257).

The basolateral transporter multidrug resistant protein 4 (MRP4) has been shown to mediate the co-

efflux of conjugated bile acids and reduced glutathione, in work from the laboratory of Dieter Keppler

(258). For this work, we provided tritium-labeled conjugated bile acids of high specific activity (259).

Keppler has suggested that conjugated bile acids percolate in and out of the sinusoidal membrane, and

that such efflux from the hepatocyte into the space of Disse may serve to prevent toxic concentrations of

bile acids in the hepatocyte (260).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

45

4. Canalicular secretion of bile acids

The next challenge was to identify the canalicular transporter or transporters. This had to be a

powerful transporter, because the intracellular concentration of bile acids was thought to be in the low µM

range, whereas the concentration of monomeric bile acids in ductal bile, based on the dialysis studies of

Duane (261) was about 1000 µM. There was thus an uphill transport of three orders of magnitude.

To study this transport system, one needed vesicles consisting predominantly of canalicular

membranes. The first preparation was achieved by Inoue and Kinne, then working in the laboratory of

Irwin Arias in New York (262). The canalicular membranes were isolated by nitrogen cavitation and

calcium precipitation. At about the same time, Peter Meier working in the laboratory of James Boyer at

Yale developed a method for preparing canalicular vesicles using zonal flotation and high speed

centrifugation through discontinuous sucrose gradients (263). These vesicles were used by Meier and his

colleagues to show that taurocholate uptake by canalicular vesicles was carrier mediated and probably

electrogenic (264). A similar study was published by Inoue, Kinne, and the Arias group (265).

In 1986, Eamon O’Maille, who had studied bile acid secretion in the intact dog, was working in our

laboratory as a guest investigator. He carefully measured the Tmax of taurocholate transport in the rat and

concluded it was too great to be explained by membrane potential alone (266). Y. Adachi returned to

Japan from the Arias lab and in 1991 reported that taurocholate was actively transported into canalicular

vesicles if an ATP generating system was present (267) . That same year, the Arias lab (Nishida et al) also

reported that rat canalicular membrane vesicles contain an ATP-dependent bile acid transport system

(268).

A year or so later, the Zurich group began to work on the ATP-dependent process of conjugated bile

acid extrusion by the canaliculus. They studied a variety of cell systems to find one which did not display

ATP-stimulated bile acid transport and finally ended up with Sf9 (insect) cells. They noted a paper from

the laboratory of Victor Ling in Vancouver, Canada that showed a partial sequence of a transport protein

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

46

named “sister of P-glycoprotein,” later named multidrug resistance 1 (MDR1) (269). Using this

sequence, an antibody to the putative canalicular transporter was generated. The Zurich group then used

phage libraries, their antibody, and Sf9 cells to monitor expression. They were ultimately successful in

cloning the bile salt export pump (BSEP) (protein, bile salt export pump; gene, Abc11A1) (270). Our

laboratory provided 3H-labeled bile acids with high specific activity (259) that were used to define its

substrate affinity.

It was not long until children with cholestatic liver disease were found to have mutations in Abcb11A1,

the gene encoding BSEP (271). This disease had already been named Progressive Familial Intrahepatic

cholestasis, type 2 (PFIC2), and as the defect is canalicular, markers of ductular injury such as γ-

glutamyl-transferase are not elevated in patients with this genetic defect. The name PFIC2 is widely

used, but it seems preferable to call the disease BSEP deficiency. There are other ABC-type canalicular

transporters such as MRP2, MDR1, and BCRP but these are not believed to be very important in the

canalicular transport of unsulfated bile acids in man, based on the severe cholestasis that occurs in

patients with BSEP deficiency as well as evidence from in vitro studies of MRP2 (272).

The laboratory of Victor Ling, in Vancouver, reported the creation of an Abcb11 knockout mouse,

which appeared to have only mild cholestasis. (273). The mildness of the mouse phenotype contrasted

strikingly with the severe cholestatic phenotype that occurs in infants with a genetic defect. This striking

difference between BSEP deficiency in humans and that in mice appears to have at least three

explanations. First, in mice, a major bile acid is β-muricholic acid, which is much less cytotoxic than

DCA and CDCA, these being major bile acids in man. Second, in mice, MDR1 is upregulated and is

likely to be an effective conjugated bile acid extruder (274). Third, BSEP deficient mice form tetra-

hydroxy and penta-hydroxy bile acids (275,276), some or all of which are likely to be transported by

MRP2 (277). Humans with cholestatic liver disease do not form tetra- or penta-hydroxy bile acids to any

extent. In mice, the defect in bile acid export from the hepatocyte is easily shown by feeding such mice

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

47

cholic acid (278), which in this condition is highly toxic. Impaired β-fatty acid oxidation is also present

in in such cholic acid-fed animals (279).

5. The basolateral transporter of the ileal enterocyte

There was one last carrier to be identified, the basolateral transporter of the ileal enterocyte. Also, if

conjugated bile acids were absorbed by cholangiocytes using ASBT and returned via the periductular

capillary plexus to the hepatocyte, there had to be a basolateral transporter in cholangiocytes. The

hepatocyte did not require a basolateral efflux transporter, as it already had one in MRP4, and most bile

acid efflux from the hepatocyte was across the canalicular membrane into bile mediated by BSEP.

In 2001, Wei Wang working with James Boyer and Nazzareno Ballatori at the Mount Desert Island

identified two genes encoding a Na+ independent bile alcohol sulfate transporter in the skate (280). The

genes were isolated using the Xenopus laevis technique of expression cloning, and they mediated

transport only when they were expressed simultaneously. They were named organic solute transporters A

and B (OSTα/OSTβ). Subsequently, Ballatori collaborated with the laboratory of Paul Dawson and

identified this dimeric organic solute transporter as the long sought ileal enterocyte basolateral transporter

(281). The two proteins were present predominantly in ileum and kidney, and immunohistochemistry

showed that the dimeric transporter was present on the basolateral membrane of the ileal enterocyte and

renal proximal tubular cell. When OSTα/OSTβ was expressed in polarized monolayers at the basolateral

domain and ASBT was expressed at the apical domain, bile acid transport from the apical side to the

basolateral side was observed (281). Further study of OSTα/OSTβ showed that it was a facilitated

diffusion carrier and not entirely specific for bile acids, in contrast to ASBT, which is highly specific for

bile acids. In cholestasis, hepatocyte OSTα/OSTβ is upregulated on the basolateral (sinusoidal)

membranes, and mediates bile acid efflux from the hepatocyte into sinusoidal plasma (282).

To date, no diseases caused by defective OSTα/OSTβ have been identified. A mouse knockout has

been created, and it differs from the ASBT knockout in that with the OSTα/OSTβ knockout, there is no

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

48

marked increase in bile acid biosynthesis despite bile acid malabsorption (283). In contrast, in the

Slc10A2 (ASBT) knockout, bile acid synthesis increases nearly twentyfold (284). This apparently

anomalous absence of a compensatory increase in bile acid synthesis is presumed to be the result of bile

acids accumulating in the ileal enterocyte and upregulating the nuclear receptor FXR, which in turn

stimulates the release of FGF15 into portal venous blood (285). Hepatocyte uptake of FGF15 acts to

downregulate bile acid synthesis, as noted in the Introduction. If there is no absorption of bile acids from

the ileum, yet synthesis does not increase, the net result should be a bile acid deficiency in the proximal

small intestine, as secretion will fall and become equal to synthesis. This in turn should cause

malabsorption of saturated fatty acids and fat soluble vitamins.

6. Imaging of bile acid transport

An 11C-labeled analogue of taurocholate (cholyltaurine) has been synthesized by the PET (positron

emission tomography) scan center of Aarhus University Hospital (286). The molecule, cholyl-N-11CH3 –

glycine (cholylsarcosine) permits the kinetics of conjugated bile acid hepatic uptake and excretion into the

biliary tract to be measured and visualized in real time. Because of the extremely short half-life of 11C (20

minutes), this compound can be used only in PET centers that have a reactor (needed for the production

of 11C) close to the patient. The clinical utility of this molecule in assessing cholestatic disease is under

active investigation.

While this review was being written, a book entitled, “Hepatobiliary Transport in Health and Disease”

was published (287). It should cover the topics discussed in our review in much more detail.

B. The enterohepatic circulation in health

1. History

The enterohepatic cycling of bile was clearly described by Borelli in 1681 (288). He wrote, “Bile is

collected in the portal vein [from the intestine] and separated from the blood again in the liver. It is then

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

49

sent into the bile vessels. It leaves these and repeats its circuit thought the intestine and the mesenteric

veins again and again.” Liebig, a prominent German chemist who worked in the mid-19th century, was

fully aware of the enterohepatic circulation of some or all of the constituents of bile (289). Lehmann in

his text book of physiological chemistry, published in 1853, wrote,”the greater part of the bile is again

resorbed as it passes through the intestine. The chief biliary constituents which are resorbed are the

soluble salts and cholic acid” (290). Moritz Schiff, working in Florence, Italy, prepared anesthetized dogs

with an “amphibolic” catheter. When bile was diverted to the outside, bile flow stopped; when bile was

allowed to enter the intestine, bile flow continued. Schiff concluded that bile secretion requires the

intestinal absorption of bile acids (291). (Schiff was put on trial by the English in Florence for

unnecessary cruelty to animals, as he used anesthetized dogs in his experiments. He was found guilty,

and forced to leave Italy. Schiff established his laboratory in Geneva, Switzerland, and became a leading

physiologist in Europe in the 19th century). Additional information on the history of knowledge about the

enterohepatic circulation is given in the detailed review of the enterohepatic circulation by M. Carey and

W. Duane published in 1994 (292).

The enterohepatic circulation of bile acids denotes a mass of bile acids that is discharged by gallbladder

contraction into the small intestine during digestion, is absorbed from the distal intestine, returns to the

liver, and may either be stored in the gallbladder or resecreted into the intestine. When a meal is ingested,

the hormone cholecystokinin is released from the small intestine. Cholecystokinin induces gallbladder

contraction as well as relaxation of the valve (sphincter of Oddi) at the end of the common bile duct

where it empties into the small intestine. Bile then enters the duodenum. Some of the bile acids are

absorbed in the jejunum, but most are transported by intestinal peristalsis to the distal ileum where they

are efficiently absorbed. The bile acid molecules pass through ileal enterocytes, and enter portal venous

blood to return to the liver. One of the early illustrations of the enterohepatic circulation of bile acids with

values for man was presented by Sune Bergström in 1959 (8) and is shown in Figure 8. Whether ancient

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

50

vertebrates, e.g. the coelacanth and the hagfish, have an enterohepatic circulation with their C27 bile

alcohol sulfates is not known.

2. Determinants, properties and measurement of the enterohepatic circulation

As noted previously, the enterohepatic circulation is the resultant of vectorial transport by the ileal

enterocyte and the hepatocyte, as well the flow in the portal venous system, the small intestine, and the

biliary tract. The only molecules that have an enterohepatic circulation are those that are transported by

the membrane transporters of the ileal enterocyte and the hepatocyte. Because only bile acids are

transported by specific transporters, only bile acids have an enterohepatic circulation. The possible

enterohepatic circulation of drugs is beyond the scope of this review.

The vast majority of the circulating bile acids are in the biliary tract and intestine, because plasma and

hepatocyte levels are very low. Plasma levels are low (< 10µM) because of efficient hepatic extraction as

well as dilution. Urinary levels are also low (< 10µM) because only a fraction of plasma bile acids enter

the glomerular filtrate, as bile acids are bound to plasma proteins, mostly to albumin (293). Moreover,

much of the fraction of bile acids that enters the glomerular filtrate is reabsorbed in the proximal tubules

via ASBT and OSTα/β. We found that cholyl conjugates were 70% bound, and dihydroxy conjugates

were >95% bound in human serum (251), so the glomerular filtrate should contain mostly conjugates of

cholic acid in man. The end result is that the concentration of bile acids in urine in healthy man is

extremely low, and urinary bile acids constitute a negligible fraction of bile acid excretion. .

The mass of cycling bile acids in man was first measured by Sven Lindstedt, a member of the

Bergström group, then in Lund (14). Lindstedt prepared cholic acid tagged with tritium and followed the

decay of bile acid specific activity (in bile) with time in human subjects. The specific activity of the

isolated cholic acid fell exponentially with time, indicating that loss of radioactivity followed first order

kinetics. The slope of the natural logarithm of the bile acid specific activity decay curve can be

extrapolated to zero time to permit calculation of the pool size. The slope of the specific activity decay

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

51

curve times the pool size gives the synthesis rate. The technique of isotope dilution described by

Lindstedt has been used to measure the pool size and synthesis rate of the two primary bile acids – cholic

and CDCA repeatedly (294). Such studies have shown that the rate of cholic acid synthesis is about twice

that of CDCA.

The technique can also be performed with stable isotopes, measuring the atoms % excess in plasma

bile acids by mass spectrometry, approach developed by Franz Stellaard and his colleagues in Groningen

(295). When radioactive bile acids are used in the usual amounts, the specific activity of plasma bile acids

is too low to measure accurately.

The isotope dilution technique has also been used for DCA (296), but input now represents absorption

of newly formed DCA rather than synthesis. The input of DCA is some fraction of cholic acid synthesis,

and could therefore vary from 0 to nearly 1.0. This fraction – DCA input/cholic acid synthesis has been

termed the fdehydrox and ranges in healthy adults from 0.2 to 0.5 (297).

In the past, there has been concern because values for bile acid synthesis obtained by isotope dilution

were often somewhat higher than those obtained by analysis of fecal bile acids. The belief at present, is

that the discrepancy between these two methods, which should give identical results, is attributable to the

difficulty of analyzing all fecal bile acids. Thus some fecal bile acids are not measured, and a higher result

for bile acid synthesis is obtained by the isotope dilution technique. Also, if a considerable volume of bile

is removed during the daily duodenal bile sampling used in the isotope dilution technique, this will

increase bile acid synthesis.

No isotope dilution studies have been performed with C27 bile acids or bile alcohols.

3. Synthesis of labeled bile acids for metabolic studies

In the laboratory of the senior author at the Mayo Clinic, better radioactive tags for bile acids were

sought in order to characterize bile acid metabolism. The Bergström group was the first to synthesize 24-

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

52 14C tagged bile acids (298), and this label was stable during enterohepatic cycling. In order to prepare 3H

tagged bile acids that would also be stable during enterohepatic cycling, we used a method developed by

Peter Klein to exchange 3H into the C-2 and C-4 positions (299). N. LaRusso, as his first research

project, examined the stability of this label, and found it was good, but not perfect (300). For labeling

CDCA and lithocholic acid, we synthesized their Δ11 derivatives, and reduced them with 3H. Again, this

label was good, but not perfect (301). Finally, in San Diego, in work led by our chemist, Claudio

Schteingart, we prepared bile acids with a double bond between C-22 and C-23, and then reductively

tritiated these molecules (259). The 22-23 3H label was shown to be completely stable during

enterohepatic cycling in a collaborative study performed with W. Duane at the University of Minnesota

(302). Isotope dilution studies have not been performed with β-muricholic acid because of the non-

availability of labeled material. However, the Δ22 compound, the precursor for reductive tritiation, has

been synthesized by the Iida group (137).

4. The enterohepatic circulation in man

Because of bacterial deconjugation in the intestine followed by absorption from the intestine and

reconjugation in the liver, the turnover rate of the steroid moiety of C24 bile acids is considerably slower

than that of its glycine moiety. As noted previously, this deconjugation-reconjugation pathway was

identified in Sweden by A. Norman in rats (252), and also somewhat later by clinical studies in the

laboratory of the senior author at the Mayo Clinic (253). Deconjugation-reconjugation also occurs with

taurine conjugated bile acids, but the difference in turnover rates between the steroid and amino acid

moiety is less, because taurine conjugates are deconjugated more slowly than glycine conjugates (162).

These studies, performed at the Mayo Clinic in the laboratory of the senior author, have never been

repeated, so confirmation of their findings is desirable.

No information is available on the turnover rates of either the steroid moiety or the taurine moiety of

C27 bile acids. For most C27 bile alcohols, the process of deconjugation – reconjugation is unlikely, as the

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

53

liberated bile alcohol, in contrast to that of unconjugated bile acids, is too polar to be passively absorbed

(190).

Early workers measured the bile acid pool by isotope dilution and guessed at the number of times that

the pool cycled, the product being bile acid secretion. In our view, this assumption is not valid, and is

analogous to trying to predict cardiac output from the measurement of blood volume. A flow can only be

estimated by indicator dilution techniques. Workers at the Mayo Clinic used such an indicator dilution

technique to measure bile acid secretion and found that it averaged about 12 g/day, rising during meal

digestion and falling during the interprandial interval, as well as during overnight fasting (304,305). Bile

acid secretion during digestion was about 0.3 µmol/kg-min, a value confirmed in a separate study of

gallbladder emptying by G. Van Berge Henegouwen and colleagues, also at the Mayo Clinic (306). Bile

acid secretion was also quantified in another intestinal perfusion study by T. Northfield and the senior

author, and similar results were obtained (307).

Portal venous blood delivers bile acids to the hepatic sinusoids, and bile acids are efficiently extracted,

first pass fractional extraction being 50-90%, depending on the bile acid. C. Einarsson, B. Angelin and

their colleagues performed experiments in which they measured bile acid concentrations in portal venous

blood and hepatic venous blood in patients at surgery and thus could measure directly hepatic extraction

(308). M. Korman and his colleagues found that hepatic extraction of glycocholate was identical during

both the fasting and postprandial state (309). This constancy of efficient hepatic extraction means that

hepatic extraction is blood-flow limited, and this view was confirmed in a careful study of hepatic bile

acid extraction in the dog from the laboratory of R. Hanson (310). First pass clearance for CDCA in

healthy volunteers was determined.by van Berge Henegouwen and colleagues. They compared the area

under the curve (AUC) after intravenous administration, with the AUC after oral administration. A value

of 62% was obtained (311). Unconjugated UDCA has a first pass extraction of about 50% (312). In

general, dihydroxy conjugates, i.e. the conjugates of DCA and CDCA have a lower first pass extraction

than cholyl conjugates, perhaps because the dihydroxy conjugates are bound much more tightly by

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

54

albumin. For a given steroid moiety, conjugated bile acids are extracted more efficiently than the

unconjugated bile acid.

5. Absorption of bile acids in the biliary ductules; cholehepatic shunting

The discovery that cholangiocytes contain ASBT (238,239) is present in the ileal enterocyte,

meant that a fraction of conjugated bile acids secreted into canalicular bile is absorbed during passage

through the biliary ductules. The biliary ductules are nurtured by a periductular capillary plexus that

empties into the portal region of the hepatic sinusoids. If conjugation is not complete, unconjugated bile

acids will enter into canalicular bile. In addition, we found in our laboratory that 24-nor C23 dihydroxy

bile acids (norCDCA, norDCA, and norUDCA), which are inefficiently conjugated, are absorbed

passively from the biliary ductules (163,164). The absorbed molecules return to the hepatocyte and are

once again resecreted into canalicular bile, thus undergoing a cholehepatic circulation. The absorption of

the unconjugated bile acid removes a proton from the canalicular bile, generating a bicarbonate anion, the

result being no change in bile osmolarity. Lamri et al provided additional evidence for the validity of

cholehepatic shunting using immunohistochemical techniques (313). The existence of a “cholehepatic

shunt” for unconjugated mono- and dihydroxy bile acids has now been accepted by most hepatologists.

When the unconjugated bile acid returns to the sinusoid, it is taken up by the hepatocyte and

resecreted into bile, thereby inducing additional canalicular bile flow, as evidenced by increased mannitol

clearance. Mannitol clearance is considered a measure of canalicular secretion (314). The increase in

bile flow enriched in bicarbonate is termed “hypercholeresis”. Hypercholeresis was originally described

by the laboratory of Serge Erlinger (315). In their experiments in biliary fistula rats, UDCA was infused

at a rate exceeding the hepatocyte’s capacity to conjugate it. As a result, unconjugated UDCA was

secreted into bile, underwent cholehepatic shunting and induced a bicarbonate rich hypercholeresis. This

report from the Erlinger laboratory gave rise to the erroneous belief that UDCA had unique choleretic

properties. In cholestatic disease, most patients appear to conjugate their ingested UDCA completely

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

55

based on analyses of biliary bile acids (316), but if conjugation were incomplete and unconjugated UDCA

were secreted into bile, it would most likely be absorbed in the biliary ductules, thereby undergoing

cholehepatic shunting. In further work from our laboratory, we showed that sulindac, but not other

NSAID’s, also underwent cholehepatic shunting (317).

6. Determinants of plasma bile acids

The plasma level of bile acids is determined by the instantaneous rates of intestinal absorption

and hepatic uptake. Dogs with a chronic biliary fistula have no detectable bile acids in their systemic

venous blood, thus confirming that intestinal absorption is the source of plasma bile acids, at least in

health (318). The profile of plasma bile acids is not identical to the profile of biliary bile acids. Plasma

bile acids are enriched in dihydroxy conjugates because of their lower hepatic extraction.

7. Regulation of the enterohepatic circulation

The enterohepatic circulation of bile acids must balance two conflicting physiological functions. On the

one hand, bile acids must serve as the end products of cholesterol metabolism and excretion is necessary

for cholesterol balance. For this function, enterohepatic cycling is unimportant. On the other hand, the

ileal conservation of bile acids is needed to provide ample bile acids for efficient lipid digestion, and bile

acid excretion is unimportant. Given these conflicting aims, it should be highly likely that the

enterohepatic circulation of bile acids would be tightly regulated.

The idea of regulation of the enterohepatic circulation is not new. Whipple in his 1922 review (91)

noted that there is little increase in total bile acids in dogs ingesting bile acids chronically, and suggested

that absorption must be regulated. Sten Eriksson, working in Lund, observed that bile acid synthesis

increased with time in the biliary fistula rat (17). Thus the working hypothesis was that bile acid

synthesis is determined by the hepatocyte concentration of bile acids. When this falls, bile acid synthesis

increases. When bile acids are fed, bile acid synthesis decreases. The senior investigator characterized

bile acid metabolism in patients with ileal resection. A marked increase – up to ten fold – was present in

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

56

such patients, again attributed to bile acid malabsorption lowering the concentration of bile acids in the

hepatocyte (319). A similar increase in bile acid synthesis was induced by cholestyramine administration

(320). Studies in the rhesus monkey with an exteriorized enterohepatic circulation of bile acids, also

provided convincing evidence that bile acid synthesis was regulated in a negative feedback manner (321).

In these studies performed by R.H. Dowling, D. Small, and E. Mack (the surgeon who prepared the

monkeys), the return of bile acids to the liver could be systematically decreased, and an increase in

synthesis quantified.

However, two observations did not seem to support this useful hypothesis. The first was that bile acid

synthesis increased at least acutely when the bile duct is ligated (115). The second, made independently

by the group of Reno Vlahcevic in Richmond, VA, (322) and the group of Chiijiwa in Japan (323), was

that upregulated synthesis in the biliary fistula rat could be returned to normal levels by intestinal, but not

by intravenous infusion of bile acids. Both types of infusion should lead to an increase in the

intracellular concentration of bile acids. The Richmond group proposed that a blood-borne signal from

the intestine to the liver might down regulate bile acid synthesis.

The breakthrough came with the identification of FGF15 (in the rat) and FGF19 (in man) as a protein

released by the ileal enterocyte in response to FXR induction by bile acids. This observation by Inagaki

and Kliewer and colleagues at UT Southwestern (58) was a major advance in our understanding of the

regulation of the enterohepatic circulation.

The present view is that the enterohepatic circulation is regulated in two aspects. Input, i.e. bile acid

synthesis is regulated at the level of the hepatocyte. Conservation, i.e. ileal transport, is regulated at the

level of the ileal enterocyte. Both synthesis as well as ileal transport are downregulated by FGF19 (53).

FGF19 enters the hepatocyte via a receptor (FGFR4) and directly or indirectly downregulates bile acid

synthesis. Synthesis of FGF19 in the ileal enterocyte is under the control of FXR, the nuclear receptor

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

57

activated by bile acids. Nonetheless, in some species, it remains possible that bile acid synthesis is

regulated by the bile acid concentration in the hepatocyte rather than by FGF19 (324).

The regulation of ileal conservation is poorly understood. J. Lillienau, working with the senior author,

found that bile salt feeding in the guinea pig downregulated ileal transport whereas administrations of a

bile acid sequestrant upregulated ileal transport (325). Upregulation of ileal bile acid transport was

associated with recruitment of more proximal enterocytes to transport bile acids. Nonetheless, with

complete cholestasis or with parenteral alimentation, bile acid transport by the ileal enterocyte decreases

(326,327). Thus there must be multiple mechanisms of control. It is likely that the down regulation of

the ileal bile acid transporter (ASBT) with bile acid feeding involves FXR agonism and FGF19 release.

C. Perturbations of the enterohepatic circulation in disease

Perturbations of the enterohepatic circulation result from defects in bile acid transport or flow,

assuming that bile acid synthesis and conjugation are normal. Bile flow begins at the canaliculus and

there can be ductular obstruction, as occurs in cholestatic liver disease such as primary biliary cirrhosis.

Physical obstruction can occur with choledocholithiasis or cholangiocarcinoma. Bile flow can be diverted

to the outside, as in patients with partial or complete biliary fistula. Bile acids can regurgitate into the

stomach and from there into the esophagus. In the small intestine, conjugated bile acids might leak across

the epithelium if increased paracellular permeability were present. (To date, such a defect has not been

identified). Bacterial overgrowth can be present, leading to deconjugation, and sometimes, to

dehydroxylation. Unconjugated bile acids are rapidly absorbed, leading to an intraluminal bile acid

deficiency, that in turn can cause lipid malabsorption.

Return of bile acids from the small intestine can be caused by defects in ileal transport, either at the

apical or basolateral domains of the ileal enterocyte, or by absence of the ileum. In addition, if rapid

intestinal transport propels bile acids past the terminal ileum at a rate exceeding its absorptive capacity,

spillover into the colon will occur, and such may induce diarrhea. Portal venous blood flow may be

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

58

obstructed or portal-caval shunting may occur. Finally hepatocyte uptake, transport through the

hepatocyte, or canalicular secretion may be impaired. It is beyond the scope of this historical review to

deal with each of these perturbations on bile acid metabolism.

V. Physicochemical Aspects of Bile Acids

A. Overview

The physical chemistry of biliary lipids and digestive lipids are similar, in that both involve

solubilization of polar lipids in the form of mixed micelles. Mixed micelle formation – whether during

biliary lipid secretion or triglyceride digestion - have received a great deal of attention from physicians

working together with physical chemists, in an attempt to unravel the complexities of these processes.

In 1983, the Kroc Foundation sponsored a small conference (9 physicians, 15 chemists) entitled, “The

Physical Chemistry of Bile in Health and Disease”. Many of the topics discussed below are treated in

more detail in the published proceedings of that meeting (328). Sadly, it was to be the last Kroc

Foundation conference. Ray Kroc, patron of the foundation (and founder of McDonald’s, a global fast

food chain) died shortly thereafter, and his widow, Joan Kroc, had other priorities for her generous

philanthropy. The foundation was dissolved.

In 1966, the senior author collaborated with Donald Small in the writing of a brief review on the

physicochemical properties of bile acids (329). In 1985, Martin Carey authored a systematic review of

the physicochemical properties of C24 bile acids (330). His review included tabulation of physical

constants, crystal structure as determined by X-ray diffraction, surface chemistry of bile acids, pH-

solubility profiles, ionization behavior, and solubilization properties of bile acid solutions. Together, these

two reviews summarized existing knowledge of the physicochemical properties of bile acids, as

envisioned by medically trained physician-scientists. In the discussion below, only selected aspects of the

physicochemical properties are considered.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

59

B. Bile acids in aqueous solution

1. Solubility of the protonated acid

The first detailed studies on the solubility of protonated, unconjugated bile acids were performed by A.

Roda and A. Fini at the University of Bologna, Italy (331). They reported solubility values at pH 3 for ten

common bile acids. Values varied enormously. That of lithocholic acid was 0.05 µM whereas that of the

7β- epimer of cholic acid (so called “ursocholic” acid) was 1670 µM. Another observation of clinical

relevance was that the two bile acids used for gallstone dissolution had quite different aqueous

solubilities: that of CDCA was 27 µM whereas that of UDCA was 9 µM. The greater aqueous solubility

of CDCA contributes to its excellent gastrointestinal absorption (210); UDCA, with a lower monomeric

solubility is absorbed less efficiently and its crystal dissolution is rate limiting in its absorption (332).

The solubility of several protonated glycine conjugated bile acids have also been determined by A.

Roda and his colleagues. They are quite low (Aldo Roda, personal communication). Taurine conjugated

bile acids, in contrast, even in the acid form, are water soluble.

As yet, we have no solubility values for any of the unconjugated C27 bile acids. The solubility of 5α-

cyprinol was found to be 360 µM, thus being similar to that of cholic acid (190). No other solubility data

are available for C27 bile alcohols.

2. Ionization

One of the purposes of the Kroc Foundation meeting was to reach a consensus on the pKa values of

bile acids, as titration studies performed by D. Small (333) and H. Igimi and M.C. Carey (334) gave

conflicting results, suggesting that the pKa of an unconjugated bile acid was influenced by the number of

hydroxyl groups. B. Josephson, working in Stockholm in the 1930’s as a predoctoral student, determined

that taurine conjugated bile acids were strong acids, that glycine conjugates had a pKa of 4-5, and that

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

60

unconjugated bile acids had a pKa of about 6 (335). A similar conclusion for unconjugated bile acids was

reached by the laboratory of Per Ekwall working in Turku, Finland (336).

At the Kroc meeting, Aldo Roda presented work performed by him and A. Fini which gave accurate

pKa values for unconjugated bile acids (331). Roda used a technique previously used to determine the

pKa of long chain fatty acids. One measures the apparent pKa in mixtures of increasing proportions of

water in methanol and extrapolates to 100% water. The values reported were about 5.1 for the common,

natural unconjugated bile acids with no substituents on the side chain. Later, Roda and Fini obtained

values of about 4 for glycine conjugates (337). The pKa of glycine conjugates is lower than that of

unconjugated bile acids, because the electron withdrawal effect of the amide bond enhances the loss of a

proton from the carboxyl group (338). The pKa of cholylsarcosine was determined by our laboratory to

be 3.7 (338), about 0.3 pKa units lower than that of cholylglycine.

For unconjugated bile acids, the pKa can be reduced by electronegative substituents on the side chain.

Roda et al found the pKa of 23R-hydroxy-CDCA to be 4.8, about 0.2 units lower than that of CDCA

(135). It should be possible to lower the pKa of unconjugated bile acids considerably by adding one or

two fluorine atoms to the C-23 carbon.

The physicochemical significance of conjugation is that it increases solubility markedly at intestinal

pH. Dihydroxy bile acids, which constitute 60% of human bile acids, are insoluble at the pH conditions

prevailing in the small intestine during digestion (pH 6-7). In vitro, the solubility of the bile acid ion

increases exponentially with increasing pH. When the concentration of bile acid reaches the critical

micellization concentration (CMC), solubility increases markedly, in that a crystalline excess becomes

transformed into micelles. There is thus a “critical micellization pH” (CMpH) which defines an

approximate concentration above which the bile acid is highly soluble and below which the bile acid is

insoluble (339). Lillienau et al, working in our laboratory, found the CMpH of glycocholate to be 4.8

(338). That of CDCA was found by van Berge Henegouwen to be about 6.8 (340) and that of UDCA has

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

61

been calculated to be 7.9 (339). The converse of CMpH has been termed “the pH of precipitation” (333)

and denotes the pH at which bile acids begin to precipitate from a true solution of the bile acid, the value

being dependent on the bile acid concentration. The matter is discussed extensively in the review of

Donald Small which gives values for the pH of precipitation of the CDC, DCA, and cholic acid (333).

K.J. Mysels derived the following equation:

CMpH = log [CMC] + pKa + BApS

Where BApS is the negative logarithm of the aqueous solubility of the protonated monomer. The high

CMpH for UDCA is thus explained by its low aqueous solubility and a relatively high CMC.

Despite their excellent pH-solubility profile, glycine conjugated bile acids will precipitate in vivo if

the pH is sufficiently acidic. This occurs in patients with gastrin-secreting tumors in whom markedly

increased gastric acid secretion overwhelms pancreatic bicarbonate secretion and the small intestinal

content becomes acidic (341). In the porcupine, glycine conjugated bile acids reflux from the duodenum

into the stomach and precipitate, forming gastric bezoars (342).

3.Solubility of Calcium (Ca2+) Salts of Bile Acids

Conjugation also greatly increases the resistance to precipitation by divalent cations such as Ca2+.

Studies from our laboratory (343) in collaboration with K. Mysels (a legendary physical chemist) showed

that the calcium salts of taurine conjugates of common natural bile acids are water soluble. Glycine

conjugation has a very small effect on the ion product of Ca2+ ion x [BA-]2, and the solubility products of

Ca2+ salts of glycine conjugated bile acids are quite low. However, the supersaturated solutions of the

calcium salts of glycine conjugated bile acids are metastable, resisting formation of the insoluble Ca2+ salt

for many weeks. The solubilities of Ca2+ salts of unconjugated bile acids, like those of glycine conjugated

bile acids, are also quite low (344). However, metastability of supersaturated solutions is not observed

with unconjugated bile acids.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

62

In the enterohepatic axis, the most dangerous site for possible insoluble calcium salt formation is the

gallbladder, where ionized Ca2+ concentrations rise during the concentration of bile by the gallbladder

epithelium (345). Stone formation remains rare, as the concentration of ionized Ca2+ is kept rather low in

the biliary tract and small intestine. The low concentration of ionized Ca2+ is explained by the high Na+

concentration, which decreases the activity coefficient of calcium (339).

Formation of Ca2+ bile salt gallstones occurs when “calcium-sensitive” bile acids are present in

appreciable proportions in biliary bile acids. In general, this has occurred because of the feeding of

unusual bile acids or their precursors (346). Examples are the feeding of cholestanol (the saturated

derivative of cholesterol with an A/B trans structure) to the rabbit. The saturated sterol is converted to

allocholic acid, that in turn is converted to allodeoxycholic acid by intestinal bacteria. The

allodeoxycholic acid is conjugated with glycine and the insoluble glycine conjugate

(glycoallodeoxycholate) precipitates from solution forming concretions in the gallbladder (107). Other

instances of the formation of insoluble Ca2+ salts are exemplified by the feeding of lithocholic acid to the

taurine deficient rat (347,348), as well as the feeding of murideoxycholic acid to the prairie dog (349).

The biological significance of conjugation is that it enhances ionization at the pH present in jejunal

content during digestion, preventing passive absorption across the apical membrane of the enterocyte. The

bile acid molecule, whether unconjugated or conjugated is too large to traverse the paracellular junctions

of the healthy intestine. This “non-absorbability” of conjugated bile acids permits high concentrations of

conjugated bile acids to be maintained in the biliary tract and proximal small intestine where the pH

ranges from pH 6 to pH 7, and allows them to function as biological solubilizers. Conjugation is also

likely to prevent bile acids partitioning into the endoplasmic reticulum during their transit through the

hepatocyte.

4. Temperature effects on solubility

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

63

Temperature effects are not very important in the biology of bile acids. Solutions of the common

primary bile acids are stable at 0o C. Friedrich Krafft, working in Heidelberg, Germany observed that

turbid dispersions of soaps became a clear solution over a very narrow temperature range, and his

observation has been immortalized in the term “Krafft point” which denotes this temperature of a phase

transition. One definition of the Krafft point, is that temperature when the concentration of the bile acid

monomer reaches the CMC. With a further increase in temperature, the excess solid phase disappears and

the suspension is transformed into a clear, micellar solution. A thorough discussion of the Krafft point

can be found in the monograph of Laughlin (350).

The term “critical micellization temperature” (CMT) was proposed, but has not been widely adapted.

Sodium cholanoate (no hydroxyl groups) and most mono-hydroxyl bile acids (including lithocholic acid)

have high CMT values, well above body temperature.

In contrast, primary dihydroxy- and trihydroxy-bile acid conjugates have very low CMT values. An

attempt was made by the senior author to define the CMT values of sodium taurodeoxycholate. This was

done by measuring the CMT of sodium taurodeoxycholate:sodium palmitate mixtures with a progressive

increase in the mole fraction of sodium taurodeoxycholate. One can then estimate the CMT value of pure

sodium taurodeoxycholate by extrapolation. The extrapolated values were well below 0oC (350). Thus,

because of their solubility at 0o C, the common natural polyhydroxy C24 bile acids may be considered

excellent “cold water detergents”.

The influence of the A/B ring juncture on the CMT of C24 bile acids has not been defined. There is also

no information on the CMT values of C27 bile acids as well as those of bile alcohol sulfates with the

single exception of 5α-cyprinol sulfate whose CMT was reported to be < 00 C (190).

5. Self-association in solution: Micelle formation

In 1936, G.S. Hartley, working in London, wrote a small lucid monograph entitled, “Aqueous

Solutions of Paraffin-Chain Salts: A study in micelle formation“(351). This work summarized the

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

64

evidence, mostly based on conductivity measurements, for micelle formation by long chain fatty acid

anions as well as alkyl sulfates, and proposed a structure for the spherical micelle. A.S.C. Lawrence, a

colleague of Hartley’s once said to the senior author, “Hartley wrote down what everyone else was

thinking!” Hartley notes in the final chapter, “The bile salts are obviously amphipathic”. At the end of

the Second World War, Hartley returned to academic life and decided to take up the problem of micelle

formation in bile acid solutions. However, his postdoctoral student developed mental problems, and the

work stopped. Hartley himself left the academic world and became a scientist at Fisons, where he

invented novel sprayers for crops as well as developing new pesticides.

The concept of micelle formation had been first advanced by James McBain in 1913, who presented

this idea at a meeting of the Royal Society. The chairman listened carefully, and began the discussion

with, “Nonsense, McBain.” McBain, who spent the latter part of his scientific career at Stanford, spent

much of his subsequent years, characterizing soap solutions, and undertook limited studies with bile acid

solutions. McBain also noted in 1941 that detergent solutions can solubilize insoluble dyes. His widow

and E. Hutchinson published a book entitled, “Solubilization and Related Phenomena” in 1955, which

summarized many of the observations on this topic in the McBain laboratory (352). J. Bashour and L.

Bauman, working in New York, developed a method for synthesizing conjugated bile acids, and in 1937

used their synthetically prepared conjugates to show that bile acid solutions solubilize cholesterol in a

concentration-dependent manner (353).

In 1942, O. Mellander and E. Stenhagen, working in Sweden, performed conductivity studies with

sodium taurocholate and found evidence of aggregate formation (354). Per Ekwall, working in Turku,

Finland, summarized the evidence for micelle formation in sodium cholate solutions in 1951 (355).

Ekwall also characterized surface properties of bile acids and made many contributions to the physical

chemistry of bile acid solutions. (356).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

65

As noted above, a program on the biology and chemistry of bile acids was initiated by Sune Bergström

in Lund after the war. Arne Norman synthesized the glycine and taurine conjugates of most of the major

bile acids. He then collaborated with Per Ekwall to study the solubilization of 20-methylcholanthrene and

thus define the critical micellization concentration (CMC) for dilute solutions of pure, conjugated bile

acids (357). Shortly thereafter, and independently, the senior author, working in Lund, Sweden,

synthesized and measured the CMC of the major bile acids present in human bile using trans-azobenzene

as the solubilizate (358).

In contrast to typical ionic surfactants, the aggregation of bile acid anions is gradual, and there is no

well-defined CMC. Thus, the CMC for bile acid solutions is often taken as the midpoint of a small

concentration range over which micelles form. K.J. Mysels suggested the term “uncritical micellization

concentration”. P. Mukerjee has pointed out that bile acids are rigid molecules in contrast to typical ionic

detergents which have a flexible alkyl chain. Therefore, bile acid molecules, just as dye molecules, stack

rather than form spherical micelles. The aggregates (from tetramers up to 20 mers, depending on bile

acid structure and concentration) may be termed multimers rather than micelles. The size of the

aggregates was estimated by light scattering, especially by D. Small (329,333) and J. Krahtohvil (359).

However, as noted below, aggregates of bile acid molecules alone, i.e. without an accompanying lipid, do

not occur in nature, except when phospholipid extrusion into bile is defective.

The most recent measurement of CMC values for a large number of unconjugated and conjugated

bile acids was made by Aldo Roda in our laboratory, in collaboration with K. J. Mysels (360). This study

was successful because of two major advances. The first was the synthesis of a large number of bile acid

epimers by Takashi Iida and Frederick Chang (361), and in a few cases, by our laboratory. The second

was the availability of a maximum bubble pressure device designed, constructed, and characterized by

K.J. Mysels. A key aspect of this technique for measuring surface tension in relation to concentration, is

that a new bubble surface is constantly being formed whose composition consists of the dominant

constituent of the solution. In contrast, with surface tension methods using a tensiometer, a more surface

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

66

active impurity may concentrate at the surface of the solution to be analyzed and lead to erroneous values

for the CMC. Roda validated his bubble pressure values by showing that identical values were obtained

with the technique of dye solubilization using Orange OT (1-O-tolyl-azo-2-naphthol) as the water-

insoluble dye. Values are reported in the absence and presence of 0.15 M Na+ concentration.

The senior author, when working in Sweden in the 1960’s observed that the aggregation of

monoglycerides and bile acids to form mixed micelles was cooperative, so that solubilization occurred at

a concentration well below the CMC of bile acids in the absence of such amphiphilic lipids (358). W.

Duane used dialysis to measure the intermicellar concentration of bile acids (monomers plus simple

micelles) in both model bile samples and human bile samples. He obtained values for the critical

micellization concentration as well as for the intermicellar concentration of bile acids which were quite

similar to the critical micellization concentration in bile acid-monoglyceride systems (261).

C. Solubilization of polar lipids by bile acid micelles.

1. Physical chemistry of biliary lipids

Bile acids are present at micellar concentrations only in bile and small intestinal content. In man, the

concentrations in plasma (2-10 µM) and in colonic content (400 µM) are well below the CMC. In health,

bile acids are always associated with a polar lipid. In bile, this is predominantly phosphatidylcholine

(PC). In small intestinal content, during digestion, the polar lipids are monoacyl glycerol and fatty acids

(partly ionized). The only circumstance in which bile acid micelles are present without an accompanying

polar lipid is in the Mdr2 knockout mouse and in patients with a defective Mdr3 protein. Such mice (and

patients) lack this ATP-activated PC extruder in the canaliculus of the hepatocyte and are believed to have

essentially no phospholipid in ductular bile. The micellar bile acids, if sufficiently hydrophobic, attack

the membranes of the cholangiocytes, leading to ductal inflammation in mice (362) and to a variety of

biliary problems in patients (363). Nonetheless, the bile of many vertebrate species has an extremely low

ratio of phospholipids to bile acids (181) without any sign of biliary ductule injury. In such species, it

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

67

may be that phospholipid, after secretion into canalicular bile, is absorbed in the biliary ductules with the

result that it is not present in gallbladder bile in appreciable proportions, or that the ductules are highly

resistant to attack by bile acids or both.

The recognition that lecithin was present in bile occurred early in the 20th century. G. Whipple in his

1922 review (91) tabulated the reported analyses of biliary lecithin. In 1952, Polonovski and Bourillon,

working in Paris, confirmed that the dominant phospholipid in bile was lecithin, later to be named PC

(364). G. Phillips separated biliary phospholipids chromatographically and confirmed that the dominant

phospholipid in bile was PC (365). Another article in this series gives the history of phospholipids in

more detail.

Franz Ingelfinger, one of the dominant figures in American Gastroenterology in the mid 1900’s was

unhappy with the level of ignorance of the pathogenesis of the common disease of gallstones. He

arranged for Donald Small, one of his brightest fellows, to spend two years in Paris working under the

direction of a scholarly colloid chemist, Digren Dervichian. During those two years, the behavior of

model systems simulating bile was described clearly using triangular coordinates. The ternary systems –

water-lecithin-bile acid, water-cholesterol-bile acid, water-cholesterol-lecithin – were carefully defined

using sodium cholate, egg lecithin, and cholesterol. The behavior of the three lipids and water could then

be described by a tetrahedron. A slice across the tetrahedron at 10% total lipid concentration gave a

triangle with three coordinates – cholesterol, lecithin (PC) and bile acid. A line defined the limits of the

micellar zone. Accordingly, points lying above the line would be supersaturated in cholesterol either in

liquid crystalline or crystalline form. This classic work is summarized in reference 333 and illustrated in

Figure 9. The stage was set for applying this pure exercise in model systems to the pathogenesis of

cholesterol gallstone formation (see later).

That phospholipids play a key role in the solubilization of cholesterol in bile was perhaps first

recognized in the mid 1950’s by B. Isaksson, working in Sweden (366). Work on the solubilization of

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

68

lecithin by bile acid solutions was carefully studied by S. Yoshimuta (367), a member of the Department

of Surgery at Kyushu University in Japan whose director, E. Miyake, had a longstanding interest in

gallstone formation. This work was published in the 1960’s about the same time that D. Small was

studying model systems simulating bile in Paris. Another physician with interest in this problem was

Kerrison Juniper who attributed cholesterol solubilization in bile to a macromolecular complex (368). A

review by D. Small and the senior author, published in 1967, advanced the idea that the physical

chemistry of micelle formation in bile and micelle formation in small intestinal content during triglyceride

digestion were similar physicochemical processes (329).

Thus by 1967, both bile and small intestinal content were considered micellar solutions, the only

difference being the nature of the accompanying lipids. Simple bile acid micelles are poor solubilizers for

cholesterol or non-polar dietary lipids. The mixed micelle provides a hydrocarbon interior in which

poorly soluble lipids can dissolve. In bile, this insoluble lipid is cholesterol; in small intestinal content,

insoluble lipids of physiological importance are the fat soluble vitamins. The remaining vexing problem

was the molecular arrangement of the mixed micelles present in bile and small intestinal content.

2. Molecular arrangement of mixed micelles

To solve the problem of the structural arrangement of the mixed micelles in bile, D. Small traveled to

England to the laboratory of Dennis Chapman (F.R.S.), a physical chemist with a deep interest in

membrane structure. They examined solutions by NMR and found that the angular carbons of the bile

salt molecule were in a hydrophobic environment. On the basis of the NMR spectra, they proposed the

“mixed disc” model in which a bilayer of PC molecules with a drum shaped perimeter was coated on its

surface with bile acid molecules (369). Later, dimers of bile acids interdigitating between the PC bilayer

were added (330). The model was widely accepted.

At the Kroc meeting in 1984, G. Lindblom of the University of Lund proposed that bile acids could

also interact with phospholipid bilayers by lying flat in the bilayer with the hydrophobic face of the bile

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

69

acid molecule pushing the charged heads of PC apart (370). W. Nichols and J. Ozarowski proposed that

bile acids lie flat on the surface of worm-shaped micelles (371) in contrast to the mixed disc model. Rex

Hjelm, a biophysicist working at the Los Alamos National Laboratory, examined bile acid-PC-water

systems using small angle neutron scattering. Hjelm concluded that as the PC/bile salt ratio increased, the

change in scattering indicated that PC-bile acid micelles increase in size by the elongation of constant

diameter rods (372), providing additional support for the “radial shell model” structure advanced by

Nichols and Ozarowski. . Hjelm also concluded that the disk-shaped micelle originally proposed by

Small et al. could not be correct. Nonetheless, the conclusions of Hjelm are not widely known in the

medical community, because much of his work was published in chemical journals that are not included

in the PubMed data base. A depiction of the original model for the mixed micelle proposed by Small and

Carey is compared with that of the model inferred from the small angle neuron studies of Hjelm and his

colleagues in Figure 10.

In 2002, S. Marrink and A. Mark, working in the Netherlands used the technique of molecular

dynamic simulations, in which molecules aggregate in water to give the lowest free energy (373). They

concluded that, “phospholipids are packed radially with their headgroups at the surface and the

hydrophobic tales [sic] point…. toward the micellar center. The bile salts act as wedges between the

phospholipid headgroups”. They concluded that the structure of the micelles generated in their system

resemble the radial shell model proposed by Nichols and Hjelm. Additional work by Long et al (374)

using small angle X-ray scattering and quasi-elastic light scattering were also in agreement with the

model proposed by Nichols.

Another approach to study the interaction of bile acids and phospholipids was taken by Heuman et al

(375). They prepared single layer vesicles and characterized the binding of bile acids to them. They also

determined at what phospholipid/bile acid ratios, the vesicles were transformed into micelles.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

70

For teaching purposes, it would be nice to have a video with realistic molecular models showing the

transformation of lipid bilayers into mixed micelles by bile acid anions.

D. Physical chemistry of bile and cholesterol gallstone disease

1. Supersaturated bile in bile from gallstone patients

D. Small returned from France to Boston in 1965, and began to apply his studies on model systems

simulating bile to human biliary lipid compositions. Bile from radiolucent gallstone patients and healthy

control subjects was collected. The proportions of bile acid, PC, and cholesterol were plotted using

triangular coordinates in a study in which W. Admirand did much of the leg work. A line denoting

equilibrium cholesterol saturation had been obtained by D. Small based on his studies in Paris: samples

above the line were supersaturated with cholesterol whereas those below the line were unsaturated in

cholesterol. When the data were plotted, bile from patients with gallstones was supersaturated with or

without cholesterol microcrystals, whereas bile from most non-gallstone patients was unsaturated (376).

[The paper also re-introduced triangular (or ternary) coordinates to the medical audience. Triangular

coordinates can be used when there are three variables adding up to a constant sum, i.e. there are two

degrees of freedom, permitting the constructions of graphs in two dimensions. They were originally

proposed in 1873 by the great physical chemist J. Willard Gibbs, of Yale University and two decades later

popularized by a Dutch chemist, H.W.B. Roozeboom (377). Probably, their first usage in medical

journals was in a paper by E.H. Ahrens (378) published in 1957 to describe the proportions of fats,

carbohydrates, and protein in diets].

Cholesterol gallstone disease could now be considered a disease of defective bile secretion by the liver,

rather than being the result of a diseased gallbladder, as had often been proposed in the past. The paper

also indicated that biliary lipid composition should be expressed as a ratio of solute (cholesterol) to

solvent (bile acid-PC micelles). The story of the rise and fall of medical dissolution of cholesterol

gallstones is discussed later.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

71

2. Vesicle formation and gallstone disease

In the original phase diagram of D. Small, the physical state of cholesterol in supersaturated systems

depended on the ratio of PC to bile acid. At low ratios, cholesterol was crystalline. At higher ratios, the

excess was liquid crystalline, i.e. in lamellae of PC and cholesterol. Norman Mazer and Martin Carey

used quasi-elastic light scattering to characterize model systems simulating bile. At higher PC to bile acid

ratios, they found evidence for “microprecipitates” although they also suggested that these

microprecipitates were vesicles enriched in cholesterol compared to mixed micelles (379,380). About the

same time Giora Somjen and Tuvia Gilat, working in Israel, used gel permeation chromatography to

separate mixed micelles from vesicles (381). They equilibrated their columns with 10 mM cholate prior

to chromatography and thus the vesicular phase remained unchanged during the chromatographic

separation. They also noted that if the columns were preequilibrated with buffer alone, a bile sample

would become entirely vesicular, as the bile salt molecules moved into the aqueous spaces in the gel

permeation column, thereby increasing the phospholipid/bile acid ratio. Thomas Holzbach, working in

Cleveland, also found vesicles in human bile samples and showed that nucleation of cholesterol crystals

from the cholesterol-rich vesicles occurred rapidly (382). William Higuchi, working in Salt Lake City,

showed using dialysis techniques that model bile systems contained four constituents: monomers, simple

bile acid micelles, mixed micelles (containing bile acids, PC, and cholesterol) and vesicles also containing

the three biliary lipids. The vesicles were relatively enriched in cholesterol (383).

Because of the complexity of the physicochemical aspects of bile and because of the widespread

interest in gallstone pathogenesis and treatment, in 1989, the NIH sponsored a workshop entitled, “Biliary

Cholesterol Transport and Precipitation”. The proceedings (including much animated discussion) were

published as a supplement to Hepatology the following year (with financial support from the Ciba-Geigy

Corporation) (384). The meeting included a wide range of scientists from pure membrane biophysicists to

gastroenterologists and surgeons, and the discussion often indicated problems in communication. A

summary of the meeting was authored by Steven Strasberg, a scholarly surgeon, and his associate Robert

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

72

Harvey. Their summary (385) presented a reasonably coherent view of bile supersaturation. Cholesterol

and phospholipids are secreted in vesicular form. Bile acids, initially present as simple bile acid micelles,

solubilize phospholipid and cholesterol. However, the transfer process, presumably occurring by

collision, solubilizes phospholipids preferentially, leaving behind vesicles containing an excess of

cholesterol (from which cholesterol crystallizes). Thus, there is a progressive change in the physical state

of bile as it moves from the canaliculus into the gallbladder.

At this meeting, J. Donovan, then working in the Carey laboratory, tabulated the existing clinical data

(386). The proportion of cholesterol in vesicles was higher in hepatic bile than in gallbladder bile in all

studies, indicating that the transformation of vesicles to mixed micelles continued during gallbladder

storage of bile. C.Schriever and D.Juengst showed, using bile samples from gallstone patients, that the

ratio of cholesterol to phospholipid in vesicles purified by gel filtration was 2.0 (387), a ratio that favors

formation of cholesterol crystals.

Since this meeting, especially because of the declining interest in medical dissolution of gallstones,

work on the physical chemistry of bile has waned. W. Higuchi continued his work on model systems,

emphasizing the “activity” of monomeric cholesterol (388). Cholesterol saturation continued to be

calculated from the original triangular figure of D. Small, even though the limit to the micellar zone was

moved downward, based on in vitro studies of model systems from the laboratories of Henrik Dam in

Copenhagen (389), and that of R.T. Holzbach in Cleveland (390). [Henrik Dam, a Dane, had a long

interest in cholesterol. He received the Nobel Prize in 1943 for his discovery of vitamin K. In the 1960’s,

he turned his enormous energy to the study of cholesterol gallstones and did both animal (391) and

clinical studies (392) of note. Sadly most of his publications are to be found in an obscure journal (Z. fur

Ernahrungswissenchaft) and are seldom cited]. Simple methods for calculating cholesterol saturation

based on the revised line of saturation were developed. M. Carey published “critical tables” that give a

value for cholesterol saturation for any molar ratio of phospholipids to cholesterol, as well as varying total

lipid concentration (393). The line of saturation is also influenced by bile acid type -- being lower when

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

73

bile acids are enriched in conjugates of UDCA. M. Carey has also published critical tables for this special

circumstance (394).

E. Physical chemistry of fat digestion

1. Studies on model systems simulating triglyceride digestion

The physical chemistry of model systems simulating fat digestion has received much less attention.

The senior author, when working in Sweden, defined the behavior of each of the relevant constituents in

dilute bile acid solutions. Fatty acids (oleic) were readily available, but monoacyl glycerides

(monoglycerides) were not. Fortunately, they were generously supplied by Fred Mattson, a chemist

working at the Miami Valley laboratories of Proctor and Gamble in Ohio. [Mattson, a gifted biochemist,

discovered the positional specificity of pancreatic lipase (395) and then showed that triglyceride was

absorbed from the intestine in the chemical form of monoacylglycerol and fatty acid (396).] The solubility

of fatty acids in bile acid micelles was pH dependent, as protonated fatty acid has a rather low solubility

in bile acid micelles, whereas fully ionized fatty acid (soap) is infinitely soluble (350). Simple titration

experiments (397) indicated the pKa of fatty acids in a taurine conjugated bile acid micelle was about 6.9,

indicating that fatty acids at proximal small intestinal pH (where most fat absorption occurs) are about

half ionized. Mono-olein, whether the 1- or 2-isomer, was highly soluble, with the ratio of solubilized

monoolein to micellar bile acid approaching 1.8 (358). In contrast, di- and triacyl glycerides had little

solubility in bile acid micelles.

In 1988, M. Svärd working in the group of Björn Lindman in Lund, investigated the taurocholate-

monoolein-water system by a great variety of techniques, and constructed the complete phase diagram

(398). In her paper, she also noted minor, but consistent differences between this system and the bile

acid-PC-water system elucidated by the pioneering work of D. Small two decades before. What both

diagrams share in common is a large micellar zone, as shown for the taurocholate-monolein-water system

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

74

illustrated in Figure 11. The paper of Svärd et al also proposes that bile acid molecules lie flat on the

surface of the bilayers.

2. Presence of a micellar phase in vivo during triglyceride digestion

The in vitro findings indicated that fatty acids and monoglycerides were highly soluble in bile acid

micelles, but that di- and triglycerides were not. These observations led to the prediction that during fat

digestion, glycerides and fatty acids would be in two phases. There would be an oil phase containing di-

and tri-acyl glycerides and a micellar phase consisting of bile acid micelles containing partly ionized fatty

acid and monoglyceride. The oil phase might also contain protonated fatty acids if these were generated

by pancreatic lipolysis at a rate exceeding that which could be solubilized in mixed micelles. Later,

healthy volunteers were intubated, fed a mixed meal, and intestinal content was collected. It was then

ultracentrifuged at body temperature, and both the oil phase and micellar phase analyzed. The results

confirmed the predictions derived from the in vitro experiments (399).

Some years later, a more detailed analysis of this system was performed by O. Hernell and J. Staggers

working in the M. Carey laboratory. They studied both model systems (400), as well as small intestinal

content obtained by intubation in healthy volunteers who had ingested a triglyceride rich meal (401).

They confirmed the older studies of the senior author, but noted in addition, that vesicles were present.

They proposed that vesicles form at the surface of the oil droplets, and that these are fairly rapidly

converted to mixed micelles when they encounter unsaturated bile acid micelles. In 1959, A.S.C.

Lawrence, a colloid chemist at Sheffield University, proposed that detergents adsorb to “dirt” and form

liquid crystalline myelin figures (multilayered vesicles). His analysis (402) is probably an apt description

of the formation of vesicles composed of fatty acid and monoacylglycerol at the surface of the triglyceride

droplet.

A closing chapter on this topic was authored by R. Hjelm, in which model mixtures simulating fat

digestion were prepared in our laboratory and examined by small angle neutron scattering. Hjelm

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

75

concluded that mixed micelles containing a bile acid and either monoglyceride (monoacylglycerol) or

monoglycerides plus fatty acids had essentially the same structure as the mixed micelles containing bile

acids and PC that he had studied previously (403,404). In both, the bile acid molecules lie flat on the

surface of the spherical or worm shaped micelles.

3. Necessity of bile acids for efficient absorption of triglycerides

Numerous studies dating back to the 19th century had shown that triglyceride absorption efficiency

was impaired in animals with a biliary fistula and that feeding of bile acids would correct fat

malabsorption (405). Nonetheless, there were efforts to show that bile acids were unnecessary for fatty

acid absorption (406). The view of the senior author is that bile acids are essential for the efficient

absorption of those fatty acids that have very low aqueous solubilities, i.e. saturated fatty acids of C16

length or longer (407). Fatty acids with appreciable aqueous solubility (unsaturated fatty acids and

saturated acids having a chain length ‹ C16) do not require bile acids for efficient absorption. This view

that bile acids facilitate the absorption of saturated fatty acids (≥ C16 in length) is based on two

observations. First, fecal fatty acid analyses indicate that exogenous bile acids cause a decrease in the

proportion of long chain saturated fatty acids in short bowel syndrome patients (408). Second,

administration of cholestyramine to rats causes selective malabsorption of long chain saturated fatty acids

(409). It is well established that bile acids are required for the absorption of fat soluble vitamins.

The lack of requirement of bile acids for efficient absorption of fatty acids with appreciable aqueous

solubility provides the rationale for using medium chain triglycerides for nutritional support in patients

with a deficiency of bile acids in the small intestine. The liberated medium chain fatty acids are quite

water soluble and are rapidly absorbed in the complete absence of bile acids.

VI. Dissolution of cholesterol gallstones by oral bile acids and topical solvents

A. Gallstone dissolution by oral bile acids.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

76

As noted in the introduction, the famous triangle of D. Small (and W. Admirand) indicated how biliary

lipid composition could be plotted. (The term “biliary lipids” to include bile acids, PC (lecithin), and

cholesterol was probably introduced by D. Small in the 1960’s. It did not include bilirubin diglucuronide

(which gives bile its color), as this pigment derived from heme is a very minor constituent of bile. Biliary

lipids differ from plasma lipids in that biliary lipids include bile acids, and do not include glycerides and

fatty acids).

In the 1950’s, Charles Johnston, a surgeon at Wayne State University, was interested in determining

what he termed the “cholesterol holding capacity” of bile, that is, how much additional cholesterol could

be dissolved when added to bile samples. He observed that T-tube bile from gallstone patients was

“saturated”, whereas bile samples obtained from healthy subjects undergoing surgery was unsaturated.

With a young Japanese surgeon, Fumio Nakayama, he fed cholic acid to gallstone patients, but found that

it did not increase the “cholesterol holding capacity”. In the discussion of his paper, he noted that he

hoped to try CDCA, but of course, at that time, there was no CDCA to be had (410). [Fumio Nakayama

continued to study gallstone pathogenesis using chemical techniques and had a long and distinguished

career as a Professor of Surgery in Kyushu, Japan. After he retired he summarized his lifelong interest in

gallstone formation and treatment in a monograph (411)].

After J. Thistle and L. Schoenfield made the seminal discovery that CDCA but not cholic acid changed

bile from supersaturated to unsaturated, it was logical to perform a clinical trial of CDCA for gallstone

dissolution. The effort was spearheaded by J. Thistle, as L. Schoenfield took a new position in Los

Angeles in 1970. In 1971, the first observations were made that gallstones were decreasing in size,

probably the first witnesses being J. Thistle (and his radiological colleague) and the senior author. In

1972, successful gallstone dissolution was reported in May at the annual meeting of the American

Gastroenterological Association and later that year in Europe at the 2nd International Bile Acid Meeting

sponsored by the Falk Foundation of Freiburg, Germany. The early Mayo experience with gallstone

dissolution was published in the New England Journal of Medicine in 1972 and became a “Citation

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

77

Classic” (37). Later that year, gallstone dissolution by CDCA was reported by the group of R.H.

Dowling, working at Guy’s Hospital in London (38). In 1973, Dr. Falk Pharma decided to market CDCA

for gallstone dissolution.

In February 1972, a small meeting was held at the NIH to discuss a national multicenter placebo-

controlled trial of CDCA. It was felt that the absence of patent protection would preclude development

by pharmaceutical companies. A “Request for Proposals” was issued in July 1972. L. Schoenfield

submitted a proposal which included committee structure for the proposed National Cooperative

Gallstone Study (NCGS). On the committees were most every American scientist or physician interested

in gallstone disease!

The study was delayed, because of the FDA concern that CDCA might cause liver damage. The

FDA demanded that liver biopsies be performed on the first 100 patients, including those on placebo.

Despite the ethical problems inherent in subjecting placebo group patients to liver biopsy, the study was

performed without any adverse events.

The concern of the FDA was based in part on two reports from the Huntington Laboratories in

England of severe toxicity induced by CDCA feeding in the rhesus monkey (412). As a result of these

reports, European studies were stopped in 1973. Report of the primate toxicity provided a justification for

liver biopsies in the patients when therapy was stopped (413). An international registry of biopsies was

set up, and a careful review of the biopsy histology showed no important histological changes. Trials in

Europe recommenced.

The answer to this remarkable species difference in CDCA toxicity came from work by the senior

investigator and his colleagues who showed that in man, lithocholic acid, the major bacterial metabolite

of CDCA, is sulfated and rapidly excreted from the body (414,415), whereas in the rhesus monkey,

lithocholic acid is extremely poorly sulfated, accumulates in the circulating bile acids, and induces liver

damage (416,417). Independent studies by the lab of E. Mosbach reported that CDCA also was toxic in

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

78

the rabbit, again because of lithocholic acid accumulation (418). Similar findings were observed when

rhesus monkeys (419) and baboons (420) were given CDCA.

The NCGS began in 1976 and its results were published in 1981 (421). The study was a double blind

study with three groups -- placebo, 350 mg CDCA/day and 750 mg CDCA/day. Dissolution occurred in

the placebo group (11%), was greater (24%) in the low dose group and highest (41%) in the high dose

group. During the study it was recognized that the high dose was probably still too low a dosage (422),

but the study could not be changed. In the high dose group, 8% of patients had to discontinue the study

because of an increase in serum aminotransferase levels. Toxicity was shown to be attributable to CDCA

and not to lithocholic acid (423). While the NCGS was proceeding, controlled studies were being

performed in Europe which showed that CDCA had unequivocal efficacy and minimal toxicity (424,425).

An analysis of the kinetics of gallstone dissolution by William Higuchi indicated that the reduction in

diameter induced by the presence of unsaturated bile occurred independently of size (426). Thus, the

bigger the stone, the longer the time needed for complete dissolution. This prediction was later confirmed

by Senior et al in a thoughtful analysis of x-ray changes of gallstone size during medical dissolution

(427). .

Just as the NCGS was beginning, word came from Japan (a careful study led by Isao Makino and his

colleagues) that ingestion of ursodeoxycholic acid, the 7β- epimer of CDCA, would also induce gallstone

dissolution (40). Earlier, there were several anecdotal cases in which subjects had taken large doses of

UDCA and induced their own gallstone dissolution, and this was done without knowledge of the

American studies with CDCA. Thus, medical dissolution of gallstones must be considered an American

and Japanese discovery.

In the United States, the NCGS drafted a protocol that would permit a study similar to the original

NCGS, but would compare UDCA with CDCA; however, the study was not funded by NIH. CDCA was

approved for marketing in the United States by the FDA in 1983, and UDCA was approved a few years

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

79

later. UDCA, like CDCA, was also toxic in the rhesus monkey (428) and rabbit (429) because of

lithocholic acid accumulation in the circulating bile acids.

Despite the safety and efficacy of CDCA and UDCA, there were some physicians who were

unimpressed by medical therapy as compared to surgical therapy. There were several problems with

medical therapy. First, it took a long time – months to years -- to obtain complete dissolution, especially

for larger stones. Second, studies by Wolpers suggested that with the first attack of biliary colic, the

resulting inflammation would cause a surface layer of non-cholesterol constituents on the gallstone,

slowing or precluding dissolution (430). Thus, there was no window of opportunity: when stones became

symptomatic, they had already become resistant to medical dissolution. Moreover, it took many months

of therapy before one could be sure that the stone was resistant. Finally, after stones dissolved, there was

recurrence (431). It was gallstone recurrence that led surgeons a half century before to switch from

cholecystotomy (removal of the stone but leaving the gallbladder in place) to cholecystectomy (432). A

thoughtful review by Gracie and Ransohoff concluded that it was better to “wait and cut” than to “screen

and treat” because of the benign natural history of most gallstones (433). An editorial by K. Isselbacher

was entitled “Disillusion with Dissolution” (434).

B. Acceleration of dissolution by extracorporeal shockwave lithotripsy

Attempts were made to speed up dissolution. One approach was to fragment the stones by using

extracorporeal shockwave lithotripsy, a technique that had been developed for the fragmentation of

kidney stones. For gallstones, the stone fragments would be smaller than the original stone(s), have

cholesterol surfaces exposed by the shattering of the stones, and thus, medical dissolution would occur

more rapidly. However, there was a major difference between kidney stones and gallbladder stones.

When kidney stones were fractured by this technique, the fragments passed spontaneously down the

urinary tract and were expelled during urination. With gallbladder stones, the fragments sunk to the

bottom of the gallbladder, so that dissolution by desaturating bile acids was needed. Another limitation of

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

80

extracorpeal shock wave lithotripsy was that only certain types of stones (one or two in number) could be

treated. Thorough clinical studies of the efficacy and safety of this technique for gallstone dissolution

were conducted by the Munich group led by Gustav Paumgartner (435). Leslie Schoenfield led one of

several efforts in the United States (436).

C. Topical dissolution with organic solvents

Another approach to non-surgical treatment of cholesterol gallstones was contact (topical) dissolution

with organic solvents. The Mayo group, led by J. Thistle, introduced the idea of placing a percutaneous

transhepatic catheter into the gallbladder and then lavaging the stone with methyl tert-butyl ether (MTBE)

a solvent that had become available as a gasoline additive (437) and was a superb cholesterol solvent.

When the solvent was pumped into and then aspirated from the gallbladder, stones would dissolve in a

matter of a few days. A spark-free pump was designed and patented by the Mayo Clinic. European trials

commenced in a few centers (436,437).

In our laboratory at UCSD, we had not planned to become involved in this approach. A patient with

symptomatic gallstones who was a poor operative risk presented with symptomatic gallstones, and we

reluctantly carried out successful dissolution by manual instillation and aspiration of MTBE. A “smart”

pump was designed by S. Zakko so that solvent could be pumped into and out of the gallbladder at a high

rate. The pump was programmed to continuously sense intraluminal pressure and aspirate gallbladder

contents whenever the gallbladder started to contract, thus avoiding spillage of the solvent into the

common bile duct (438). A new solvent – ethyl propionate – was tested and found to dissolve gallstones

as nearly as rapidly as MTBE (439). Ethyl propionate did not induce nausea in contrast to MTBE

because of rapid removal from plasma (440). Patients were treated successfully (441).

However, there were multiple problems with contact dissolution. The solvents used were commercial

samples and they were not manufactured according to FDA standards. MTBE was flammable and

considered toxic (442). Preclinical studies had not been done. The procedure required some hours for

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

81

complete dissolution, and as described, it required professional personnel to perform the infusion and

aspiration. In studies performed in pigs, the gallbladder epithelium was observed to be destroyed by

either solvent, although it recovered rapidly (443). There was a report from France of MTBE escaping

into the blood stream and causing severe renal damage (444). In the United States, two deaths occurred

unrelated to usage of the solvent, but nonetheless in association with solvent usage. A company was

formed to bring the pump and solvent into clinical practice. However, venture capital was never obtained,

and the approach slowly passed into oblivion. A review of topical dissolution by our group was published

(445).

D. Concurrent advances in gallstone management

Two other developments were occurring during this time that doomed widespread usage of medical

dissolution. The first was the development of laparoscopic cholecystectomy, a major surgical advance.

Laparoscopic surgery was an import from France, used first in gynecological procedures. Using this

technique, a gallbladder could be removed quickly, without a linear scar, and the patient could leave the

hospital in a few days. The gallbladder with its gallstones had been removed, and the patient was cured.

In experienced hands, the technique was as safe as the traditional open cholecystectomy which had been

performed widely for the past half century. The composition of the stone(s) was unimportant, whereas

with oral bile acid therapy only gallstones that had cholesterol as the major constituent could be

successfully treated...

The second advance was ultrasound imaging of the gallbladder and its contents. All types of stones

were detected, and there was no radiation exposure. In contrast, oral cholecystography required the

ingestion of iodine-rich contrast agents and subsequent X-ray to detect the presence of gallstones.

Thus in twenty years, medical dissolution had been discovered, prospered, and then slowly

disappeared. In the long history of biliary disease, medical dissolution was a “bubble”. Once again

symptomatic cholelithiasis became a surgical disease. Nonetheless, medical dissolution was “proof of

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

82

principle”, and medical therapy with UDCA can be used to prevent cholesterol gallstone formation.

Gallstone prophylaxis might be useful in some clinical situations, for example, in patients with recurrent

gallstone pancreatitis who did not wish to have surgery.

The availability of UDCA led to the accidental discovery of its utility in the treatment of primary

biliary cirrhosis and cholestasis of pregnancy, as noted in the introduction.

VII. Epilogue: omissions and apologies

Space limitations means that historical aspects of many facets of biology and chemistry have been

omitted. We have not discussed the history of inborn errors of bile acid synthesis and conjugation. Our

understanding of these genetic defects is quite recent, and an excellent review (446) by Clayton is

available. In the United States, Ken Setchell, a talented mass spectroscopist, has described several new

inborn errors of bile acid biosynthesis, as well as a defect in bile acid conjugation. Identification of the

exact enzyme defect requires mass spectrometry to detect the presence of bile acid precursors and gene

sequencing to identify the defect (156). The rewards of discovery are great for the patient, as bile acid

replacement may be life-saving. Some of the early history is given in J. Sjövall’s retrospective review

(10).

A second area not discussed is the utility of bile acids and bile alcohols to disclose evolutionary

relationships, an area pioneered by G.A.D. Haslewood (21). Often bile acids agree with the perceived

evolutionary relationships, but sometimes they do not. It would be of interest to explore these

nonconcordancies in detail.

A third area not discussed is the role of bile acids in colonic secretion (447,448) and absorption (449),

as well as recent work suggesting that bile acid activation of TGR5 is essential for normal defecation, at

least in the mouse (70) . Details of the biochemical mechanism(s) for bile acid-induced intestinal

secretion are being elucidated by several laboratories, among these being that of Stephen Keely, in

Dublin, (448) and of Meena Rao in Chicago (450). Bile acid malabsorption causes diarrhea, and this can

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

83

be treated by sequestrant administration (451,452). A new syndrome of defective FGF19 release causing

an inappropriate increase in synthesis of bile acids has been identified by Julian Walter and his colleagues

(453), and is considered to be present in many patients with irritable bowel syndrome of the diarrhea type

(454), as well as in patients with so-called primary bile acid malabsorption. Plasma levels of “C4” (7α-

hydroxy-cholest-4-ene-3-one), an intermediate in C24 bile acid biosynthesis, have been shown to correlate

highly with the rate of bile acid synthesis (455,456). The necessity of measuring C4 (as a marker of

increased bile acid synthesis) and FGF15/19 levels (to detect impaired FGF15/19 secretion from the ileal

enterocyte) in every patient with unexplained diarrhea is slowly being recognized by gastroenterologists

(457). The group of Michael Camilleri at Mayo is exploring the relationship of fecal bile acid profile to

constipation as well as diarrheal conditions (458). Our laboratory identified some children with

functional constipation whose fecal bile acids were predominantly the 3 sulfate of CDCA (459). The

absence of C-12 hydroxylation in these children could indicate a deficiency in cyp8B1, the enzyme that

hydroxylates C4 at the C-12 position.

A fourth area not discussed is the cellular toxicity of bile acids. Mechanisms of toxicity are multiple,

involving activation of the death receptor (460), as well as induction of reactive oxygen species (461).

UDCA is cytoprotective and mechanisms responsible for this effect are being studied (462). A new aspect

of bile acid toxicity is the proinflammatory action of retained bile acids (463).

One last area not discussed is the possible influence of bile acids on the microbiome. The presence of

secondary bile acids in bile has long been a sign that metabolic activity of the microbiome influences bile

acid metabolism. Indeed, characterization of bile acid metabolism in the germ free animal (464) by

Gustafsson and his colleagues in Lund aided in the initial distinction of primary and secondary bile acids

by the laboratory of Sune Bergström (8). There is still much is to be learned about the effect of individual

bile acids on the microbiome (465).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

84

The last two generations have had powerful tools to elucidate bile acid metabolism. One of these was

the discovery of 14C – a discovery that almost occurred by chance (466). The availability of 14C in

chemical form permitted 14C labeled bile acids to be prepared and their metabolism to be defined. A

second essential tool has been chromatography – adsorption, reversed phase (HPLC), and gas/liquid. A

final tool is mass spectrometry which when coupled with gas chromatography or HPLC provides both

qualitative and quantitative information on new molecules (9). For the assignment of structure to newly

discovered bile acids, proton and 13C nuclear magnetic resonance measurements are mandatory. Of

course, in today’s research, generation of specific knockouts, characterized by multisystem phenotyping,

combined with PCR, have enormous power in unraveling the recently discovered signaling properties of

bile acids.

At present (2014), at least two types of drugs based on natural bile acids are being tested for their

utility in the treatment of liver disease. The first, obeticholic acid, the 6α-ethyl derivative of CDCA, is a

potent FXR agonist. Its efficacy in the treatment of primary biliary cirrhosis (467) and nonalcoholic

steatohepatitis (468) is being tested in clinical trials. NorUDCA, the C23 homologue of UDCA, is also in

clinical trials for the treatment of cholestatic liver disease. In the bile duct ligated mouse, its feeding

causes less damage than UDCA feeding (469).

Inhibitors of ASBT, the sodium dependent bile acid transporter present in the apical domain of the ileal

enterocyte, are once again being developed. They were originally aimed at providing adjuvant therapy for

hypercholesterolemic patients who responded inadequately to statins (470). Now the target is cholestatic

liver disease or its symptom of pruritus as well as type 2 diabetes (471) and constipation (472).

Colesevelam is an improved bile acid sequestrant that should have a similar pharmacodynamic activity as

that of the ASBT inhibitors; however, it is only approved for type II diabetes, and its use to treat bile acid

diarrhea is off label. Bile acid derivatives that activate solely TGR5 or both TGR5 and FXR have been

synthesized (55) and may eventually enter clinical trials.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

85

In our opinion, the remarkable progress in our understanding of bile acid biology and chemistry is a

tribute to the hundreds of scientists who have worked with such diligence and talent throughout the world.

We have mentioned only a few in our brief history, and we have surely omitted some key advances.

There are many workers whose contributions have not been recognized because of space limitations. As

we noted in the introduction, this is a personal history, and other writers are likely to stress different

aspects of the historical record. We have probably overemphasized the contributions of our own

laboratory because we know them best. For those readers who would like more historical detail on some

of the topics discussed above, we refer them to a series of beautifully written historical reviews by Adrian

Reuben published in the journal Hepatology between 2002 and 2006.

Let us hope that future research in the chemistry and biology of bile acids will be as exciting as that of

the last 80 years!

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

86

Acknowledgments: We are grateful to many of our colleagues who helped us in this review. In

particular, we thank Takashi Iida, Bruno Stieger, and David Mangelsdorf, as well as the anonymous

reviewers. We also acknowledge a grant-in-aid from Intercept Pharma, Inc. In the past, our laboratory

has been supported by the National Institutes of Health as well as a grant-in-aid from the Falk Foundation,

e.V., Freiburg, Germany.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

87

REFERENCES

1. Thannhauser, S.J. and M. Lenke. 1928. Ueber die Herkunft der Gallensäuren, Cholesterin-

Gallensäurenbilanzen beim Hund mit totaler Gallenfistel. Arch. Exp. Pathol. Pharmakol. 1130:292-307.

2. Neubauer, E. 1924. Ueber die cholagoge Wirkung der Dehydrocholsäure beim Menschen. Klin.

Wochenschr. 3:883.

3. Shimizu, T. 1935. Ueber die Chemie and Physiologie der Gallensäuren. Verlag von M. Muramoto,

Japan.

4. Sarett, LH. 1946. Partial synthesis of pregnene 4 triol 17(β),20(β), 21 dione 3,11 and pregnene 4 diol

17(β), 21 trione 3,11,20 monoacetate. J. Biol. Chem. 162: 601-32.

5. Hillier, S.G. 2007. Diamonds are forever: the cortisone legacy. J. Endocrinol. 195: 1-6.

6. Hench, P.S. 1964. In Nobel Lectures. Physiology and Medicine. 1942-1962. Elsevier Publishing

Company, Amsterdam. 312-41.

7. Fieser.L.F., and M. Fieser. 1959. Steroids. Reinhold Publishing Corporation. New York, NY. 600-

83.

8. Bergström, S., H. Danielsson, and B. Samuelsson. 1960. Formation and metabolism of bile acids. In

Lipide Metabolism, K. Bloch, ed. John Wiley & Sons, New York, NY. 291-336.

9. Griffiths, W.J., and Sjövall, J. 2010. Bile acids: analysis in biological fluids and tissues. J. Lipid Res.

51:23-41.

10. Sjövall, J. 2004. Fifty years with bile acids and steroids in health and disease. Lipids 39:703-22.

11. Norman, A. 1955. Preparation of conjugated bile acids using mixed carboxylic acid anhydrides.

Arkiv. Kemi 8:331-42.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

88

12. Norman A., and J. Sjövall. 1960. On the transformation and enterohepatic circulation of cholic acid

in the rat. J. Biol. Chem. 233:872-85.

13. Norman, A., and Sjövall, J. 1960. Formation of lithocholic acid from chenodeoxycholic acid in the

rat. Acta Chem. Scand. 14: 1815-18.

14. Lindstedt, S., 1957. The turnover of cholic acid in man. Acta Physiol. Scand. 40:1-9.

15. Danielsson H., and J. Sjövall, 1975. Bile acid metabolism. Annu. Rev. Biochem. 44:233-53.

16. Samuelsson, B. 1959. On the metabolism of ursodeoxycholic acid in the rat. Acta Chem. Scand.

13:970-5.

17. Eriksson, S. 1957. Biliary excretion of bile acids and cholesterol in bile fistula rats. Proc. Soc. Exptl.

Biol. Med. 94:578-83.

18. Borgström, B., A. Dahlqvist, G. Lundh, and J. Sjövall. 1957. Studies of Intestinal digestion and

absorption in the human. J. Clin. Invest. 36:1521-36.

19. Lack, L., and I.M. Weiner. 1961. In vitro absorption of bile salts by small intestine of rats and guinea

pigs. Am. J. Physiol. 200:313-7

20. Borgström B, G. Lundh, and A.F. Hofmann. 1963. The site of absorption of conjugated bile salts in

man. Gastroenterology 45::229-38.

21. Haslewood, G.A.D. 1967. Bile salt evolution. J. Lipid Res. 8:535-50.

22. Hsia, S.L., W.H. Elliott, J.T. Matschiner, E.A. Doisy, Jr., S.A. Thayer, and E.A. Doisy. 1958. Bile

acids. XI. Structures of the isomeric 3 alpha,6.7-trihydroxycholanic acids. J. Biol. Chem. 233:1337-9.

23. Elliott, W.H. 1978. Bile acids LVII. Analysis of bile acids by high pressure liquid chromatography

and mass spectrometry. Lipids 13:971-5.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

89

24. Prof. T. Shimizu (obituary). 1958. Nature 181:880.

25. Hoshita, T. 1985. Bile alcohols and primitive bile acids. In Sterols and Bile Acids. H. Danielsson and

J. Sjövall, editors. Elsevier Science Publishers, Amsterdam. 279-302.

26. Une, M., and T. Hoshita. 1994. Natural occurrence and chemical synthesis of bile alcohols, higher

bile acids, and short side chain bile acids. Hiroshima J. Med. Sci. 43: 37-67.

27. Yamasaki, K. 1972. 40-year studies on bile acids. Yonago Acta Med. 15:171-87.

28. Carey, J.B., Jr. 1956. Chenodeoxycholic acid in human blood. Science 123:892

29. Carey, J.B., Jr., I.D. Wilson, F.G. Zaki, and R.F. Hanson. 1966. The metabolism of bile acids with

special reference to liver injury. Medicine 45:461-70.

30. Ahrens, E.H., Jr., and L.C. Craig. 1952. The extraction and separation of bile acids. J. Biol. Chem.

195:763-778.

31. Grundy, S.M., E.H. Ahrens, Jr., and T. A. Miettinen. 1965. Quantitative isolation and gas-liquid

chromatographic analysis of total fecal bile acids. J. Lipid Res. 6:397-410.

32. McGuire, D.K. and S.M. Grundy. 2011. Interview with Dr. Scott Grundy during AHA EPI/PAM

meeting in March 2011. J Clin. Lipidol. 5:371-2.

33. Rajaratnam, R.A., H. Gylling, and T.A. Miettinen. 2001. Cholesterol absorption, synthesis, and fecal

output in postmenopausal women with and without coronary artery disease. Arterioscler. Thromb. Vasc.

Biol. 21:1650-5.

34. Grundy, S.M., A.F. Hofmann, J. Davignon, and E.H. Ahrens, Jr. 1966. Human cholesterol synthesis

is regulated by bile acids. J. Clin Invest. 45:1018-19 (abstract).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

90

35. Thistle, J.L. and L.J. Schoenfield. 1971. Induced alterations in composition of bile of persons

having cholelithiasis. Gastroenterology 61:488-96.

36. Danzinger, R., A.F. Hofmann, L.J. Schoenfield, and J.L. Thistle. 1972. Dissolution of cholesterol

gallstones by chenodeoxycholic acid. N. Engl. J. Med. 286:1-8.

37. Bell, G.D., B. Whitney, and R.H. Dowling. 1972. Gallstone dissolution in man using

chenodeoxycholic acid. Lancet 2:1213-6.

38. Hench, P.S. 1938. Effect of jaundice on rheumatoid arthritis. Br. Med. J. 2:394-8.

39. Sugata, F., and M. Shimizu. 1974. Retrospective studies on gallstone disappearance. Jap. J.

Gastroenterol. 71:70-80.

40. Nakagawa, S., I. Makino, T. Ishazaki, and I. Dohi. 1977. Dissolution of cholesterol gallstones by

ursodeoxycholic acid. Lancet 2:367-9.

41. Makino, I., and H. Tanaka. 1998. From a choleretic to an immunomodulator: Historical review of

ursodeoxycholic acid as a medicament. J. Gastroenterol. Hepatol. 13:659-64

42. Leuschner, U., and W. Kurtz. 1987. Treatment of primary biliary cirrhosis and cholestatic disorders

with ursodeoxycholic acid. Lancet 2: 508.

43. Poupon, R., Y. Chretien, R.E. Poupon, F. Ballett, Y. Calmus, and F. Darnis. 1987. Is ursodeoxycholic

acid an effective treatment for primary biliary cirrhosis? Lancet 1:834-6.

44. Lindor, K.D., E.R. Dickson, W.P. Baldus, R.A. Jorgensen, J. Ludwig, P.A. Murtaugh, J.M. Harrison,

R.H. Wiesner, M.L. Anderson, S.M. Lange, G. LeSage, S.S. Rossi, and A.F. Hofmann. 1994.

Ursodeoxycholic acid in the treatment of primary biliary cirrhosis. Gastroenterology 106:1284-90.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

91

45. Palma, J., H. Reyes, J. Ribalta, I. Hernandez, L. Sandoval, R. Almuna, J. Liepins, F. Lira, M. Sadano,

O. Silva, D. Tohe, and J.J. Silva 1997. Ursodeoxycholic acid in the treatment of cholestasis of pregnancy:

a randomized, double-blind study controlled with placebo. J. Hepatol. 27:1022-8.

46. Nonappa, and U. Maitra. 2008. Unlocking the potentials of bile acids in synthesis,

supramolecular/materials chemistry and nanoscience. Org. Biomol. Chem. 6:657-69.

47. Allen, R.D., I.Y. Yan, G.M. Wallraff, R.A. DiPietro, D.C.Hofer, and R.R. Kunz. 1995. Resolution and

etch resistance of a family of 193nm positive resists. J. Photopolymer Sci. Technol. 8:623-6.

48. Steinberg, D. 2013. In celebration of the 100th anniversary of the lipid hypothesis of atherosclerosis.

J. Lipid Res. 54:2946-9.

49. Makishima M., A.Y. Okamoto, J.J. Repa, H. Tu, R.M. Learned, A. Luk, M.V. Hull, K.D. Lustig,

D.J.Mangelsdorf, and B. Shan. 1999. Identification of a nuclear receptor for bile acids. Science

284:1362-5.

50. Wang H, J. Chen, K. Hollister, L.C. Sowers, and B.M. Forman. 1999. Endogenous bile acids are

ligands for the nuclear receptor FXR/BAR. Mol. Cell. 3:543-553.

51. Parks, D.J. S.G. Blanchard, R.K.Bledsoe, G. Chandra, T.G. Consler, S.A. Kliewer, J.B. Stimmel, T.M.

Willson, A.M. Zavacki, D.D. Moore, and J.M. Lehmann. 1999. Bile acids: natural ligands for an orphan

nuclear receptor. Science 284:1365-8.

52. Matsubara, T., F. Lei, and F.J. Gonzalez. 2013. FXR signaling in the enterohepatic system. Mol.

Cell Endocrinol. 368:17-29.

53. Gonzalez, F.J. 2012. Nuclear Receptor Control of Enterohepatic circulation. Compr. Physiol.

2:2811-28.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

92

54. Porez, G., J. Prawitt, B. Gross, and B. Staels. 2012. Bile acid receptors as targets for the treatment of

dyslipidemia and cardiovascular disease. J. Lipid Res. 53:1723-37.

55. Inagaki, T., M. Choi, A. Moschetta, L. Peng, C.L. Cummins, J.G. McDonald, G. Luo, S.A. Jones, B.

Goodwin, J.A. Richardson, R.D. Gerard, J.J. Repa, D.J. Mangelsdorf, and S.A. Kliewer. 2005. Fibroblast

growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab.

2:217-25.

56. Jung, D., T. Inagaki, R.D. Gerard, P.A. Dawson, S.A. Kliewer, D.J. Mangelsdorf, and A. Moschetta.

2007. FXR agonists and FGF15 reduce fecal bile acid excretion in a mouse model of bile acid

malabsorption. J. Lipid Res. 48:2693-700.

57. Uriarte, I., M.G. Fernandez-Barrena, M.J. Monte, M.U. Latasa, H.C.Y. Chiang, S. Carotti, U.

Vespasiani-Gentilucci, S. Morini, E. Vicente, A.R. Concepcion, J.F. Medina, J.J.G. Marin, C. Berasain, J.

Prieto, and M.A. Avila. 2013. Identification of fibroblast growth factor 15. A novel mediator of liver

regeneration and its application in the prevention of post-resection liver failure in mice. Gut 62:899-910

58. Mudaliar, S., R.R. Henry, A.J. Sanyal, L. Marrow, H.U. Marschall, M. Kipnes, L. Adorini, C.I.

Sciacca, P. Clopton, E. Castelloe, P. Dillon, M. Pruzanski, and D. Shapiro. 2013. Efficacy and safety of

the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty

liver disease. Gastroenterology 145:574-82.

59. Gadaleta, R.M., K.J. van Erpecum, B. Oldenburg, E.C.L. Willemsen, W. Renooij, S. Murzilli, L.W.J.

Klomp, P.D. Siersema, M.E.I Schipper, S. Danese, G. Penna, G. Lavery, L. Adorini, A. Moschetta, and

S.W.C. van Mil. 2011. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal

barrier in inflammatory bowel disease. Gut 60:463-72.61.

60. Sato, H., A. Macchiarulo, C. Thomas, A. Gioiello, M. Une, A.F. Hofmann, R. Saladin, K. Schoonjans,

R. Pellicciari,and J. Auwerx. 2008. Novel potent and selective bile acid derivatives as TGR5 agonists:

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

93

Biological screening, Structure-Activity Relationships, and Molecular Modeling Studies. J. Med. Chem.

51:1831-41.

61. Pellicciari R., S. Fiorucci, E. Camaioni C. Clerici, G. Costantino, P.R. Maloney, A. Morelli, D.J. Parks,

and T.M. Willson. 2002. 6α-ethyl chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist

endowed with anticholestatic activity. J. Med. Chem. 45: 3569-72..

62. Adorini, L., M. Pruzanski, and D. Shapiro. 2012. Farnesoid X receptor targeting to treat nonalcoholic

steatohepatitis. Drug Discov. Today 17:988-97.

63. Maruyama, T., Y. Miyamoto, T. Nakamura, Y. Tamai, H. Okada, E. Sugiyama, T. Nakamura, H.

Itadani, and K. Tanaka. 2002. Identification of membrane-type receptor for bile acids (MBAR. Biochem.

Biophys. Res. Commun. 298:714-9.

64. Kawamata, Y., R. Fujii, M. Hosova, M. Harada, H. Yoshida, M. Miwa, S. Fukusumi, Y. Habata T.

Itoh, Y. Shintani, S. Hinuma, Y. Fujisawa, and M. Fujino. 2003. A G protein-coupled receptor responsive

to bile acids. J. Biol. Chem. 278:9435-40.

65. Poole, D.P., C. Godfrey, F. Cattanruzza, G.S. Cottrell, J.G. Kirkland, J.C. Pelayo, N.W. Bunnett, and

C.U. Corvera. 2010. Expression and function of the bile acid receptor GpBAR1 (TGR5) in the murine

enteric nervous system. Neurogastroenterol. Motil. 22:814-25.

66. Alemi, F., E. Kwon, D.P. Poole, T. Lieu, V. Lyo, F. Cattaruzzza, F. Cevikbas, M. Steinhoff, R. Nassini,

S. Materazzi, R. Guerrero-Alba, E. Valdez-Morales, G.S. Cottrell, K. Schoonjans, P. Geppetti, S.J. Vanner,

N.W. Bunnett, and C.U. Corvera. 2013. The TGR5 receptor mediates bile acid-induced itch and

analgesia. J. Clin. Invest. 123:1513-30.

67. Keitel, V., and D. Häussinger. 2013. TGR5 in cholangiocytes. Curr. Opin. Gastroenterol. 29:299-

304.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

94

68. Thomas, C., A. Gioiello, L. Noriega, A. Strehle, J. Oury, G. Rizzo, A. Macchiarulo, H. Yamamoto, C.

Mataki, M. Pruzanski, R. Pellicciari, J. Auwerx and K. Schoonjans. 2009. TGR5-mediated bile acid

sensing controls glucose homeostasis. Cell Metab. 10:167-77.

69. Keitel, V., R. Reinehr, P. Gatsios, C. Rupprecht, B. Görg, O. Selbach, D. Häussinger, and R. Kubitz.

2007. The G-protein coupled bile salt receptor TGR5 is expressed in liver sinusoidal endothelial cells.

Hepatology 45:695-704.

70. Alemi, F., D.P. Poole, J. Chiu, K. Schoonjans, F. Cattaruzza, J.R. Grider, N.W. Bunnett, and C.U.

Corvera. 2013. The Receptor TGR5 mediates the prokinetic actions of intestinal bile acids and is

required for normal defecation in mice. Gastroenterology 144:145-54.

71. Wang, Y-D, W-D Chen, D. Yu, B.M. Forman and W. Huang. 2011. The G-protein-coupled bile acid

receptor. Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing

nuclear factor Kappa light-chain enhancer of activated B cells (NF-κB) in mice. Hepatology 54:1421-32.

72. Sobotka H. 1938. The chemistry of the steroids. Williams and Wilkins, Baltimore, MD.

73. Sobotka, H. Physiological chemistry of the bile. 1937. Williams and Wilkins, Baltimore, MD.

74. Haslewood, G.A.D. 1967. Bile Salts. Methuen, London.

75. Haslewood, G.A.D. 1978. The Biological Importance of Bile Salts. North-Holland Publishing Co.,

Amsterdam, The Netherlands.

76. Elsevier’s Encyclopedia of Organic Chemistry. 1962. Amsterdam, The Netherlands

77. The Bile Acids. Volume 1, Chemistry, 1971; Volume 2, Physiology and metabolism, 1973; Volume 3,

Pathophysiology, 1976; Volume 4, Chemistry, Physiology, Metabolism, 1988. Plenum Press, New York

NY.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

95

78. New Comprehensive Biochemistry, volume 12. Sterols and Bile Acids. 1985. Elsevier, Amsterdam,

The Netherlands.

79. Roda, A., E. Roda, and A. F. Hofmann. 1999. Acidi biliari 2000: Aggiornamento per il future.

Masson, Milano, Italy.

80. Hofmann, A.F. 1985. Bile acid research: some major themes during the past 60 years. In Trends in

Hepatology. L. Bianchi, W. Gerok, and H. Popper, Eds. MTP Press Limited, Lancaster, England. 3-28.

81. Dowling, R.H. 1985. Bile acids in therapy – pre-1924, circa 1924, and in 1984. In Trends in

Hepatology. L. Bianchi, W. Gerok, and H. Popper, Eds. MTP Press Limited, Lancaster, England. 29-40.

82. Hofmann, A.F., L.R. Hagey, and M.D. Krasowski. 2010. Bile salts of vertebrates: structural

variation and possible evolutionary significance. J. Lipid Res. 51:226-46.

83. H. Wieland. 1928. The Chemistry of the Bile Acids. In Nobel Lectures Chemistry 1922-41. Elsevier

Publishing Company, Amsterdam. 94-102.

84. A, Windaus, 1928. Constitution of sterols and their connection with other substances occurring

nature. In Nobel Lectures Chemistry 1922-1941. Elsevier Publishing Company, Amsterdam, The

Netherlands. 105-121.

85. Bernal, J.D. 1932. Crystal structures of vitamin D and related compounds. Nature 129:277-78.

86. Rosenheim, O., and H. King. 1934. The chemistry of the sterols, bile acids, and other cyclic

constituents of natural fats and oils. Annu. Rev. Biochem.3:87-110.

87. Lindley, P.F., and M.C. Carey. 1987. Molecular packing of bile acids: structure of ursodeoxycholic

acid. J. Crystal. Spectros. Res. 17:231-49.

88. Kametani, T., K. Suzuki, and H. Nemoto. 1981. First total synthesis of (+) chenodeoxycholic acid. J.

Am. Chem. Soc. 103:2891-2.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

96

89. Swann, B., and F. Aprahamian, eds. 1999. J.D. Bernal: A life in science and politics. Verso, London,

UK.

90. Brown, A. 2005. J.D. Bernal. The Sage of Science. Oxford University Press, Oxford, U.K.

91.. Whipple, G.H. 1922. The origin and significance of the constituents of the bile. Physiol. Rev. 2:440-

59.

92. Ruzicka, L. 1973. In the borderland between bioorganic chemistry and biochemistry. Ann. Rev.

Biochem. 42:1-20.

93. Berthold, H.K. 1998. Rudolf Schoenheimer (1898-1941) Leben und Werk. 1998. (printed by Falk

Foundation, e.V., Freiburg, Germany)

94. Bloch, K., B.N. Berg, and D. Rittenberg. 1943. The biological conversion of cholesterol to cholic

acid. J. Biol. Chem. 149:511-7.

95. Shimizu, T., and T. Oda, 1934. Untersuchung der Krotengalle. II. Trioxy-bufo-sterocholensäure

C28H46O5 aus Wintergalle. Z. Physiol. Chem. 227:74-83.

96. Suganami, T., and K. Yamasaki. 1942. Uber die Alligatorschildkrotengalle. II. Trioxy-stero- and

trioxy-isostero-cholansäure. J. Biochem. (Tokyo) 35:155-172.

97. Tint, G.S. B. Dayal, A.K. Batta, S. Shefer, T. Joanen, L. McNease, and G. Salen. 1980. Biliary bile

acids, bile alcohols, and sterols of Alligator mississippiensis. J. Lipid Res. 21:110-7.

98. Bridgwater, R.J., and G.A.D. Haslewood. 1952. Comparative studies of bile salts. 6. Partial

synthesis of coprostanic acid, a ‘stem acid’ of primitive bile salts. Biochem. J. 52:588-590.

99. Ogawa, S., K. Mitamura, S. Ikegawa, M.D. Krasowski, L.R. Hagey, and A.F. Hofmann. 2011.

Chemical synthesis of the (25R)- and (25S)-epimers of 3α,7α,12α-trihydroxy-5β-cholestan-27-oic acid as

well as their corresponding glycine and taurine conjugates. Chem. Phys. Lipids 164:368-377.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

97

100. Hammarsten, O. 1898. Ueber eine neue Gruppe gepaarter Gallensäure. Z. Physiol. Chem. 24:322-

50.

101. Kazuno, T., T. Masui, and K. Okuda. 1965. Stero-bile acids and bile sterols. LXIV. The isolation

and the chemistry of a new bile alcohol, 3α,7α,12α,24ε,26-pentahydroxybishomocholane from Rana

catesbiana bile. J. Biochem. (Japan) 52:75-80.

102.. Haslewood, G.A.D. 1964. Comparative studies of ‘bile salts’. 19. The chemistry of ranol.

Biochem. J. 90:309-13.

103.. Haslewood, G.A., and L. Tokes. 1969. Comparative studies of bile salts. Bile salts of the lamprey

Petromyzon marinus L. Biochem. J. 114:179-84.

104. Hove, T.R., V. Dvomikovs, J.M. Fine, K.R. Anderson, C.S. Jeffrey, D.C. Muddiman, F. Shao, P.W.

Sorensen, and J. Wang. 2007. Details of the structure determination of the sulfated steroids PSDS and

PADS: new components of the sea lamprey (Petromyzon marinus) migratory pheromone. J. Org. Chem.

72:7544-50.

105. Hagey, L., R. K. Takagi, C.D. Schteingart, H-T. Ton-Nu, and A.F. Hofmann. 1994. C24 bile acid

reduction into bile alcohols: a novel, ubiquitous minor pathway of cholesterol elimination in vertebrates.

Hepatology 20:A665 (abstract).

106. Elliott, W.H. 1971. Allo bile acids In The Bile Acids: Chemistry, Physiology, Metabolism, Volume

1. Chemistry. P.P. Nair and D. Kritchevsky, eds. Plenum Press, New York, NY. 47-93.

107. Hofmann, A.F. and E.H. Mosbach. 1964. Identification of allodeoxycholic acid as the major

component of gallstones induced in the rabbit by 5α cholestan-3β-ol. J. Biol. Chem. 239:2813-21.

108. Mendoza, M.E., M.J. Monte, M.A. Serrano, M. Pastor-Anglada, B. Stieger, P.J. Meier, M. Medarde,

and J.J. Marin. 2003. Physiological characteristics of allo-cholic acid. J. Lipid Res. 44:84-92.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

98

109. Setchell, K.D.R., J.M. Street, and J. Sjövall. 1988. Fecal bile acids. In K.D.R. Setchell, D.

Kritchevsky, and P.P. Nair, eds. The Bile Acids: Chemistry, Physiology, Metabolism, vol. 4. Plenum

Press, New York, NY. 441-570.

110. Ridlon, J.M., D.J. Kang, and P.B. Hylemon. 2006. Bile salt biotransformation by human intestinal

bacteria. J. Lipid Res. 47:21-59.

111. Katona, B.W., N.P. Rath, S. Anant, W.F. Stenson, and D.F. Covey. 2007. Enantiomeric deoxycholic

acid: total synthesis, characterization, and preliminary toxicity toward colon cancer lines. J. Org. Chem.

72:9298-307.

112. Strecker, A. 1848. Untersuchung der Ochsengalle. Annalen der Chem. und Pharmacie 67:1-60.

113. Mylius, F. 1886. Ueber die Cholsäure. Ber. 19:374

114. Otto, R., 1869. Ueber die Gänsegalle und die Chenotaurocholsäure. Annalen der Chem. Und

Pharmacie. 149: 185-201.

115. Marschall H-U., M. Wagner, K. Bodin, G. Zöllner, P. Fickert, J. Gumhold, D. Silbert, A.

Fuchsbichler, J. Sjövall, and M. Trauner. 2006. FXR(-/-) mice adapt to biliary obstruction by enhanced

phase I detoxification and renal elimination of bile acids. J. Lipid Res. 47:582-92.

116. Wang R, L. Liu, J.A. Sheps, D. Forrest, A.F. Hofmann, L.R. Hagey, and V. Ling. 2013. Defective

canalicular transport and toxicity of dietary ursodeoxycholic acid in the abcb11-/- mouse: transport and

gene expression studies. Am. J. Physiol. Gastrointest. Liver Physiol. 305:G288-94.

117. Hofmann, A.F., J. Sjövall, G. Kurz, A. Radominska, C.D. Schteingart, G.S. Tint, Z.R. Vlahcevic, and

K.D.R. Setchell. 1992. A proposed nomenclature for bile acids. J. Lipid Res. 33:599-604.

118. Bokkenheuser, V.D., J. Winter, P.B. Hylemon, N.K.N. Ayengar, and E.H. Mosbach. 1981.

Dehydroxylation of 16α-hydroxyprogesterone by fecal flora of man and rat. J. Lipid Res. 22:95-102.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

99

119. Hofmann A.F., V. Loening-Baucke, J.E. Lavine, L.R. Hagey, J.H. Steinbach, C.A. Packard, T.L.

Griffin, and D.A. Chatfield, 2008. Altered bile acid metabolism in childhood functional constipation:

Inactivation of secretory bile acids by sulfation in a subset of patients. J. Ped. Gastroenterol. Nutr.

47:598-606.

120. Kelsey, M., J. Molina, S. Huang, and K. Hwang. 1980. The identification of microbial metabolites

of sulfolithocholic acid. J. Lipid Res. 21:751-9.

121. Cohen, B.I., A.K.Singhal, J. Mongelli, M.A. Rothschild, C.K. McSherry, and E.H. Mosbach. 1983.

Hydroxylation of secondary bile acids in the perfused prairie dog liver.Lipids 18:909-12.

122. Kakiyama G, H. Tamegai, T. Iida, K. Mitamura, S. Ikegawa, T. Goto, N. Mano, J. Goto, P. Holz,

L.R. Hagey, and A.F. Hofmann AF. 2007. Isolation of a major novel dominant biliary bile acid in the

wombat: 15α-hydroxylithocholic acid. J. Lipid Res. 48:2682-92.

123. Shefer, S., G. Salen, S. Hauser, B. Dayal, and A.K. Batta. 1982. Metabolism of iso-bile acids in the

rat. J. Biol. Chem. 257:1401-6.

124. Marschall H.U., U. Broome, C. Einarsson, G. Alvelius, H.G. Thomas, and S. Matern. 2001.

Isoursodeoxycholic acid: metabolism and therapeutic effects in primary biliary cirrhosis. J. Lipid Res.

42:735-42.

125. Hagey, L.R., T.Iida, S. Ogawa, Y. Adachi, M. Une, K. Mushiake,. M. Maekawa, M. Shimada, N.

Mano, and A.F. Hofmann. 2011. Biliary bile acids in birds of the Cotingidae family: Taurine-conjugated

(24R,25R)-3α,7α,24-trihydroxy-5β-cholestan-27-oic acid and two epimers (25R and 25S) of 3α,7α-

dihydroxy-5β-cholestan-27-oic acid. Steroids 76:1126-1135

126. . Haslewood, G.A.D., and L. Tokès. 1972. Comparative studies of bile salts. A new type of bile salt

from Arapaima gigas (Cuvier), (Family Osteoglossidae). Biochem. J. 126:1161-70.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

100

127. Kuroki, S., C.D. Schteingart, L.R. Hagey, B.I. Cohen, E.H.Mosbach, S.S. Rossi, A.F. Hofmann, N.

Matoba, M. Une, T. Hoshita, , and D.K. Odell. 1988. Bile salts of the West Indian manatee, Trichechus

manatus latirostris: novel bile alcohol sulfates and absence of bile acids. J. Lipid Res. 29:509-522.

128. Ishida, H., H. Miyamoto, T. Kajino, H. Nakayasu, H. Nukaya, and K. Tsuji. 1996. Study on the

bile salt from Megamouth shark. I. The structures of a new bile alcohol, 7-deoxyscymnol, and its new

sodium sulfates. Chem. Pharm. Bull. 44:1289-92.

129. Hammarsten, O. 1909. Untersuchung ϋber die Galle einiger Polartiere. III. Mitteilung ϋber die

Galle des Wallrosses. Hoppe-Seylers Z. Physiol. Chem. 61:454-96.

130. Windaus, A. and A. van Schoor. 1928. Uber die β-Phocae-cholsäure. Hoppe-Seylers Z.

Physiol.Chem. 143:312-20.

131. Pellicciari, R., B. Natalini, A. Roda, M. Iracema, L. Machdao, and M. Marinzzi 1989. Preparation

and physicochemical properties of natural (23R)-hydroxy trihydroxylated bile acids and their (23S)

epimers. J. Chem. Soc. Perkin Trans. 1:1289-96.

132. Merrill J.R., C.D. Schteingart, L.R. Hagey, Y. Peng, H-T. Ton-Nu, E. Frick, M. Jirsa and A.F.

Hofmann. 1996. Hepatic biotransformation in rodents and physicochemical properties of 23(R)-

hydroxychenodeoxycholic acid, a natural α-hydroxy bile acid. J. Lipid Res. 37: 98-112, 1996.

133. Hoshita, T., S. Hirofuji, T. Sasaki, and T. Kazuno. 1967, Stero-bile acids and bile alcohols.

LXXXVII. Isolation of a new bile acid, haemulcholic acid from the bile of Parapristipoma trilineatum.

J. Biochem. (Japan). 61:136-41.

134. Kihara, K., Y. Morioka, and T. Hoshita. 1981. Synthesis of (22R and 22S)-3 alpha, 7 alpha, 22-

trihydroxy-5-beta-cholan-24-oic acids and structure of haemulcholic acid, a unique bile acid isolated from

fish bile. J. Lipid Res. 22:1181-7.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

101

135. Roda, A., B. Grigolo, A. Minutello, R. Pellicciari, and B. Natalini. 1990. Physicochemical and

biological properties of natural and synthetic C-22 and C-23 hydroxylated bile acids. J. Lipid Res.

31:289-98.

136. Rodrigues, C.M.P., B. T. Kren, C.J. Steer, and K.D.R. Setchell. 1996. Formation of Δ22-bile acids in

rats is not gender specific and occurs in the peroxisome. J. Lipid Res. 37:540-50.

137. Kakiyama G, T.Iida, A. Yoshimuta, T. Goto, N. Mano, J. Goto, T. Nambara, L.R. Hagey, and A.F.

Hofmann. 2004. Chemical synthesis of (22E)-3α,6β.7β-trihydroxy-5β-chol-22-ene-24-oic acid and its

taurine and glycine conjugates: a major bile acid in the rat. J. Lipid Res. 45:567-73.

138. Une, M., F. Nagai, K. Kihira, T. Kuramoto, and T. Hoshita. 1983. Synthesis of four

diastereoisomers at carbons 24 and 26 of 3α,7α,12α.24-tetrahydroxy-5β-cholestan-26-oic acid,

intermediates of bile acid biosynthesis. J. Lipid Res. 24:924-9.

139. Hagey, L.R., S. Ogawa, N. Kato, R. Satoh, M. Une, K. Mitamura K, S. Ikegawa, A.F. Hofmann, and

T. Iida. 2012. A novel varanic acid epimer – (24R,25S)-3α,7α,12α,24-tetrahydroxy-5β-cholestan-27-oic

acid – is a major biliary bile acid in two varanid lizards and the Gila monster. Steroids 77:1510-20.

140. Windaus, A. W. Bergmann, and G. König. 1930. Uber einige Versuche mit Scymnol. Z. Physiol.

Chem. 189:148-54.

141. Asikari, H. 1939. Uber die Galle des “Akajei” Fisches (Dasyatis akajei) und die Konstitution des

Scymnols. J. Biochem (Tokyo) 29:319-324.

142. Bergmann, W., and W.T. Pace. 1943. Scymnol. J. Amer. Chem. Soc. 65:477.

143. Bridgwater, R.J., T. Briggs, and G.A.D. Haslewood, 1962. Comparative studies of ‘bile salts.’ 14.

Isolation from shark bile and partial synthesis of scymnol. Biochem. J. 82:285-290.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

102

144. Bridgwater, R.J., G.A.D. Haslewood, and J.R. Watt. 1963. Comparative studies of ‘bile salts’. 17.

A bile alcohol from Chimaera monstrosa. Biochem. J. 87:28-31.

145. Cross, A.D. 1961. Scymnol sulphate and anhydroscymnol. J. Chem. Soc. 2817-21.

146. Ishida, H., S. Kinoshita, R. Natsuyama, H. Nukaya, K. Tsuji, T. Kosuge, and K. Yamaguchi. 1991.

Study on the bile salt, sodium scymnol sulfate, from Rhizoprionodon acutus. II. The structures of

scymnol, anhydroscymnol, and sodium scymnol sulfate. Chem. Pharm. Bull. 39:3153-3156.

147. Ishida, H., H. Nakayasu, H. Miyamoto, H. Nukaya, and K. Tsuji. 1998. Study on the bile salts from

Sunfish, Mola mola L. I. The structures of sodium cyprinol sulfates, the sodium salt of a new bile acid

conjugated with taurine, and a new bile alcohol and its new sodium sulfates. Chem. Pharm. Bull. 46:12-

16.

148. Dayal, B., A.K. Batta, S. Shefer, G.S. Tint, G. Salen and E.H. Mosbach. 1978. Preparation of 24(R)-

and 24(S)-5beta-cholestane-3alpha,7alpha,24-triols and 25(R)- and 25(S)-5beta-cholestane-

3alpha,7alpha,26-triols by a hydroboration procedure. J. Lipid Res. 19:191-6.

149. Satoh, R., T. Saito, A. Ohsaki, T. Iida, K. Asahina, K. Mitamura, S. Ikegawa, A.F. Hofmann, and L.

R. Hagey. 20143. N-methyltaurine N-acyl amidated bile acids and deoxycholic acid in the bile of

angelfish (Pomacanthidae): a novel bile acid profile in Perciform fish. Steroids. 80:15-23

150. Une, M., T. Goto, K. Kihira, T. Kuramoto, K. Hagiwara, T. Nakajima,and T. Hoshita.1991. Isolation

and identification of bile salts conjugated with cysteinolic acid from bile of the red seabream,

Pagrosomus major. J. Lipid Res. 32:1619-23.

151. Hylemon, P.B., P.M. Bohdan, A.E. Sirica, D.M. Heuman, and Z.R. Vlahcevic. 1990. Chlesterol and

bile acid metabolism in cultures of primary rat bile ductular cells. Hepatology 11:982-8.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

103

152. Batta, S.K., G. Salen, and S. Shefer. 1984. Substrate specificity of cholyl glycine hydrolase for the

hydrolysis of bile acid conjugates. J. Biol. Chem. 259:15035-9.

153. Schmassmann. A., M. A. Angellotti, H-T. Ton Nu, C.D. Schteingart, S.N. Marcus, S.S. Rossi, and

A.F. Hofmann. 1990. Transport, metabolism and effect of chronic feeding of cholylsarcosine, a

conjugated bile acid resistant to deconjugation and dehydroxylation. Gastroenterology 98:163-174.

154. Lilienau, J., C.D. Schteingart, and A.F. Hofmann AF. 1992. Physicochemical and physiological

properties of cholylsarcosine: a potential replacement detergent for bile acid deficiency states in the small

intestine. J. Clin. Invest. 89:420-431.

155. Schmassmann, A., A.F. Hofmann, M.A. Angellotti, H-T. Ton Nu, C.D. Schteingart, C. Clerici, S.S.

Rossi, M.A. Rothschild, B.I. Cohen, R. J. Stenger RJ, and E.H. Mosbach. 1990. Prevention of

ursodeoxycholate hepatotoxicity in the rabbit by conjugation with N methyl amino acids. Hepatology

11:989-996.

156. Russell, D.W. 2003. The enzymes, regulation, and genetics of bile acid synthesis. Ann. Rev.

Biochem. 72:1137-74.

157. Ikegawa, S., H. Ishikawa, H. Oiwa, M. Nagata, J. Goto, T. Kozaki, M. Gotowda, and N. Asakawa.

1999. Characterization of cholyl-adenylate in rat liver microsomes by liquid

chromatography/electrospray ionization – mass spectrometry. Anal. Biochem. 266:125-32.

158. Falany, C.N., M.R. Johnson, S. Barnes, and R.B. Diasio. 1994. Glycine and taurine conjugation of

bile acids by a single enzyme. Molecular cloning and expression of human liver bile acid CoA:amino

acid N-acyltransferase. J. Biol. Chem. 269:19375-9.

159. Kase, B.F., and I. Björkhem. 1989. Peroxisomal bile acid-CoA:amino-acid N-acyltransferase in rat

liver. J. Biol. Chem. 264:9220-3.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

104

160. Rembaca, K.P., J. Woudenberg, M. Hoekstra, E.Z. Jonkers, F.A. van den Heuvel, M. Buist-Homan,

T.E. Woudenberg-Vrenken, J.Rohacova, M.L. Marin, M.A. Miranda, H. Hoshage, F. Stellaard, and K.N.

Faber. 2010. Unconjugated bile salts shuttle through hepatocyte peroxisomes for taurine conjugation.

Hepatology 52:2167-76.

161. Huijghebaert, S.M., and A.F. Hofmann. 1986. Influence of the amino acid moiety on

deconjugation of bile acid amidates by cholylglycine hydrolase or human fecal cultures. J. Lipid Res.

27:742- 752.

162. Hepner, G.W., J.A. Sturman, A.F. Hofmann and P.J. Thomas. 1973. Metabolism of steroid and

amino acid moieties of conjugated bile acids in man. III. Cholyltaurine (taurocholic acid). J. Clin.

Invest. 52:433-440.

163. Yoon, Y.B., L.R. Hagey, A.F. Hofmann, D. Gurantz D, E.L.Michelotti, and J.H. Steinbach. 1986.

Effect of side chain shortening on the physiological properties of bile acids: hepatic transport and effect

on biliary secretion of 23 nor ursodeoxycholate in rodents. Gastroenterology 90:837-852.

164. Yeh, H-Z., C.D. Schteingart, L.R. Hagey, H-T.Ton-Nu, U. Bolder, M.A. Gavrilkina, J.H. Steinbach,

and A.F. Hofmann. 1997. Effect of side chain length on biotransformation, hepatic transport, and

choleretic properties of chenodeoxycholyl homologues in the rodent: Studies with dinor- (C22), nor-

(C23) and chenodeoxycholic acid (C24). Hepatology 26: 374-385.

165. Rossi, S.S., J.L. Converse, and A.F. Hofmann. 1987. High pressure liquid chromatographic

analysis of conjugated bile acids in human bile: simultaneous resolution of sulfated and unsulfated

lithocholyl amidates and the common conjugated bile acids. J. Lipid Res. 28:589- 595.

166. Schwenk, M., A.F. Hofmann, G.L. Carlson, J.A. Carter, F. Coulston, and H. Greim. 1978. Bile acid

conjugation in the chimpanzee: effective sulfation of lithocholic acid. Arch. Toxicol. 40:109-118.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

105

167. Raedsch, R, B.H. Lauterburg, and A.F. Hofmann.1981. Altered bile acid metabolism in primary

biliary cirrhosis. Dig. Dis. Sci. 26:394-401.

168. Stiehl, A. 1977. Disturbances of bile acid metabolism in cholestasis. Clin. Gastroenterol. 6:45-67.

169. Barnes, S., P.G. Burhol, R. Zander, G, Haggstrom, R.L. Settine, and B.I. Hirschowitz. 1979.

Enzymatic sulfation of glycochenodeoxycholic acid by tissue fractions from adult hamsters. J. Lipid Res.

20:952-9.

170. Almè, B., A. Norden, and J. Sjövall. 1978. Glucuronides of unconjugated 6-hydroxylated bile acids

in urine of a patient with malabsorption. Clin. Chim. Acta 86:251-9.

171. Sacquet, E., M. Parquet, M. Riottot, A. Raizman, P. Jarrige, C. Huguet, and R. Infante. 1983.

Intestinal absorption, excretion, and biotransformation of hyodeoxycholic acid in man. J. Lipid Res.

24:604-13.

172. Radominska-Pyrek, A., P. Zimniak, Y.M. Irshaid, R. Lester, T.R. Tephly, and J.S. Pyrek. 1987.

Glucuronidation of 6 alpha-hydroxy bile acids by human liver microsomes. J. Clin. Invest. 60:234-41.

173. Matschiner, J.T., T.A. Mahowald, S.L. Hsia, E.A. Doisy Jr., W.H. Elliott, and E.A. Doisy. 1957.

Bile acids IV. The metabolism of hyodeoxycholic acid-24-C14. J. Biol. Chem. 225:803-10.

174. Kirkpatrick, R.B., M.D. Green, L.R. Hagey, A.F. Hofmann, and T.R. Tephly. 1988. Effect of side

chain length on bile acid conjugation: glucuronidation, sulfation, and CoA formation of nor bile acids

and their natural C24 homologues by human rat liver fractions. Hepatology 8:353-357.

175. Hofmann, A.F., S.F. Zakko, M. Lira, C. Clerici, L.R. Hagey, K.K. Lambert, J.H. Steinbach, C.D.

Schteingart, P. Olinga, G.M.M. Groothuis. 2005. Novel biotransformation and physiological properties of

norursodeoxycholic acid in man. Hepatology 42:1391-8.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

106

176. Borgström, B., J. Barrowman, L. Krabish, M. Lindstrom, and J. Lillienau. 1986. Effects of cholic

acid, 7 beta-hydroxy and 12 beta-hydroxy-isocholic acid on bile low, lipid secretion,and bile acid

synthesis in the rat. Scand. J. Clin. Lab. Invest. 46:167-75.

177. Marschall, H.U., W.J. Griffiths, U. Gotze, J. Zhang, H. Wietholtz, N. Busch, J. Sjövall, and S.

Matern. 1994. The major metabolites of ursodeoxycholic acid in human urine are conjugated with N-

acetylglucosamine. Hepatology 20: 845-53.

178. Iida, T., G. Kakiyama, Y. Hibiya, S. Miyata, T. Inoue, K. Ohno, T. Goto, N. Mano, J. Goto, T.

Nambara, and A. F. Hofmann. 2006. Chemical synthesis of the 3-sulfooxy-7-N-acetylglucosaminyl-24-

amidated conjugates of 3β,7β-dihydroxy-5-cholen-24-oic acid, and related compounds: unusual, major

metabolites of bile acid in a patient with Niemann-Pick disease type C1. Steroids 71: 18-29.

179. Mitamura K, N. Hori, T. Iida, M. Suzuki, T. Shimizu, H. Nittono, K. Takaori, A.F. Hofmann, and S.

Ikegawa. 2011. The identification of S-acyl glutathione conjugates of bile acids in human bile by means

of LC/ESI/MS. Steroids 76:1609-14.

180. Dietschy, J.M. and S.D. Turley 2002. Control of cholesterol turnover in the mouse. J. Biol. Chem.

277:3801-4.

181. Moschetta, A., F. Xu, L.R. Hagey, G.P. van Berge-Henegouwen, K.J. van Erpecum KJ,, F. Brouwers,

J.C. Cohen, M. Bierman, H.H. Hobbs, J.H. Steinbach, and A.F. Hofmann. 2005. A phylogenetic survey

of biliary lipids in vertebrates. J. Lipid Res. 46:2221-32.

182. Erb, W., and E. Böhle. 1969. Fecal excretion of lipids in infectious hepatitis, liver cirrhosis, and

obstructive jaundice. Klin. Wochenschr. 47:249-56.

183. van der Velde, A.E., C.L. Vrins, K. van den Oever, I. Seemann, R.P. Oude Elferink, M. van Eck, F.

Kuipers and A.K. Groen.2008. Regulation of direct transintestinal cholesterol excretion in mice. Am. J.

Physiol. Gastrointest. Liver Physiol. 295:G203-8.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

107

184. Fini, A., and A. Roda. 1967. Chemical properties of bile acids. IV. Acidity constants of glycine-

conjugated bile acids. J. Lipid Res. 28:755-9.

185. Huijghebaert, S.M. and A.F. Hofmann. 1986. Pancreatic carboxypeptidase hydrolysis of bile acid

amino acid conjugates: selective resistance of glycine and taurine amidates. Gastroenterology 90:306-

315.

186. Makino, I., S, Nakagawa, and K. Mashimo. 1969. Conjugated and unconjugated serum bile acid

levels in patients with hepatobiliary diseases. Gastroenterology 56:1033-9.

187. Setchell, K.D.R., A.M. Lawson, E.J. Blackstock, and G.P. Murphy. 1982. Diurnal changes in serum

unconjugated bile acids in normal man. Gut 23:637-42.

188. Hagey, L.R. and M.D. Krasowski. 2013. Microbial biotransformations of bile acids as detected by

electrospray mass spectrometry. Adv. Nutr. 4:29-35.

189. Hayakawa, S. 1982. Microbial transformation of bile acids. A unified scheme for bile acid

degradation and hydroxylation of bile acids. Z. Allg. Mikrobiol. 22:309-26.

190. Goto, T., F. Holzinger, L.R. Hagey, C. Cerrè, H.T.Ton-Nu, C.D. Schteingart, J.H. Steinbach, B.L.

Shneider, and A. F. Hofmann. 2003. Physicochemical and physiological properties of 5α-cyprinol

sulfate, the toxic bile salt of cyprinid fish. J. Lipid Res. 44:1643-51.

191. Sjövall, J. 1953. On the separation of bile acids by partition chromatography. Acta Physiol.

Scand. 29:232-40.

192. Haslewood G.A.D., and J. Sjövall. 1954. Comparative Studies of ‘Bile Salts’ 8. Preliminary

examination of bile salts by paper chromatography. Biochem J. 57:126-30.

193. Grundy, S.M., E.H. Ahrens, Jr., and T.A. Miettinen. 1965. Quantitative isolation and gas-liquid

chromatographic analysis of total fecal bile acids. J. Lipid Res. 6:397-410.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

108

194. Eneroth, P., G. Gordon, R. Ryhage, and J. Sjövall. 1966. Identification of mono- and dihydroxy bile

acids in human feces by gas-liquid chromatography and mass spectrometry. J. Lipid Res. 7:511-23.

195. Eneroth, P., and J. Sjövall. 1971. Extraction, purification, and chromatographic analysis of bile

acids in biological materials. In The Bile Acids. Chemistry, Physiology, and Metabolism. Vol.

1:Chemistry. P.P. Nair, and D. Kritchevsky, eds. Plenum Press, New York, NY. 121-68.

196. Stahl, E. Dϋnnschichtchromatographie. 1962. Springer Verlag, Berlin, Germany

197. Malins, D.C., and H. Mangold. 1960. Analysis of complex lipid mixtures by thin-layer

chromatography and complementary methods. J. Am. Oil Chem. Soc. 37:576-8.

198. Hofmann, A.F. 1962. Thin layer adsorption chromatography on microscope slides. Anal Biochem

3:145-149.

199. Hofmann AF. 1962. Thin layer adsorption chromatography of free and conjugated bile acids on

silicic acid. J. Lipid Res. 3:127 128.

200. Lepri, L., D. Heimler, and P.G. Desiderio. 1984. Reversed-phase high-performance thin-layer

chromatography of free and conjugated bile acids. J. Chromatogr. 288:461-8.

201. Momose, T., M. Mure, T. Iida, J. Goto, and T. Nambara. 1998. Method for the separation of the

unconjugates and conjugates of chenodeoxycholic acid and deoxycholic acid by two-dimensional

reversed-phase thin layer chromatography with methyl beta-cyclodextrin. J. Chromatogr A. 811:171-80.

202. Hofmann, A.F. 1964. Thin layer chromatographic mobilities of bile acids. In New Biochemical

Separations. L.J. Morris and A.T. James, Eds. Van Nostrand, London, 261-282.

203. Snyder, F., and H. Kimble. 1965. An automatic zonal scraper and sample collector for radioassay of

thin-layer chromatograms. Anal. Biochem. 11:510-8.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

109

204. Linnet, K., J.R. Andersen, and P. Hesselfeldt. 1984. Concentrations of glycine- and taurine-

conjugated bile acids in portal and systemic venous serum in man. Scand. J. Gastroenterol. 19:575-8.

205. Roda, A., A.M. Gioacchini, C. Cerrè, and M. Baraldini. 1995. High-performance liquid

chromatographic-electrospray mass spectrometric analysis of bile acids in biological fluids. J.

Chromatogr. B. Biomed. Appl. 665:281-94.

206. Kakiyama, G., A. Muto, H. Takei, H. Nittono, T. Murai, T. Kurosawa, A.F. Hofmann, W.M. Pandak,

and J.S. Bajaj. Development of a simple and accurate method for measurement of fecal bile acids:

validation by GC-MS and LC-MS and application in healthy and cirrhotic patients. J Lipid Res. (in press)

207. Iwata, T., and K. Yamasaki. 1964. Enzymatic determination and thin-layer chromatography of bile

acids in blood. J. Biochem (Japan) 56:424-31.

208. Hurlock, B. and P. Talalay. 1957. Principles of the enzymatic measurement of steroids. J. Biol.

Chem. 227:37-52.

209. Turley, S.D. and J.N. Dietschy. 1978. Re-evaluation of the 3α-hydroxysteroid dehydrogenase assay

for total bile acids in bile. J. Lipid Res. 19:924-8.

210. Beher, W.T., S. Stradnieks, and G.J. Lin. 1983. Rapid photometric determination of free and

esterified 3 alpha-hydroxy fecal bile acids. Steroids 41:729-40.

211.. Roda, A., L.J. Kricka, M. DeLuca, and A.F. Hofmann. 1982. Bioluminescent measurement of

primary bile acids using immobilized 7α-hydroxysteroid dehydrogenase: application to serum bile acids.

J. Lipid Res. 23:1354-361..

212. Rossi, S.S. M.A. Angellotti, K.D.R. Setchell, and A.F. Hofmann. 1991. Measurement of total

fasting-state serum bile acids: comparison of a solid-phase bioluminescence enzymatic assay, a

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

110

homogeneous fluorescence enzymatic assay, and isotope dilution gas chromatography-mass spectrometry.

Clin. Chim. Acta 200:63-66.

213. Simmonds, W.J., M.G. Korman, V.L.W. Go, and A.F. Hofmann. 1973. . Radioimmunoassay of

conjugated cholyl bile acids in serum. Gastroenterology 65:705- 711.

214. LaRusso, N.F. M.G. Korman, N.E. Hoffman, and A.F. Hofmann.1974. Dynamics of the

enterohepatic circulation of bile acids. Postprandial serum concentrations of conjugates of cholic acid in

health, cholecystectomized patients, and patients with bile acid malabsorption. N. Engl. J. Med. 291:689-

692.

215. Schalm, S.W. G.P. van Berge Henegouwen, A.F. Hofmann, A.E. Cowen and J. Turcotte. 1977.

Radioimmunoassay of bile acids. Development, validation and preliminary application of an assay for

conjugates of chenodeoxycholic acid. Gastroenterology 73:285-290.

216. Schalm, S.W., N.F. LaRusso, A.F. Hofmann , N.E. Hoffman, G.P. van Berge Henegouwen, and M.G.

Korman. 1978. Diurnal serum levels of primary conjugated bile acids. Assessment by specific

radioimmunoassays for conjugates of cholic and chenodeoxycholic acid. Gut 19:1006-1014.

217..van der Velden, L.M., M.V. Golynskiy, I.T.G.W. Bijmans, S.W.C. van Mil, L.W.J. Klomp, M. Merix,

and S.F.J. van deGraaf. 2013. Monitoring Bile Acid Transport in Single Living Cells using a genetically

encoded Förster Resonance Energy Transfer Sensor. Hepatology 57:740-52.

218. Cravetto, C., G. Molino, A.M. Biondi, A. Cavanne, P. Avagnina, and S. Frediani. 1985. Evaluation

of the diagnostic value of serum bile acid in the detection and functional assessment of liver diseases.

Ann. Clin. Biochem. 22:596-605.

219. Mishler, J.M. L. Barbosa, L.J. Mihalko, and H. McCarter. 1981. Serum bile acids and alanine

aminotransferase concentrations: comparison of efficacy as indirect means of identifying carriers of Non-

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

111

A. Non-B hepatitis agents and of onset, severity, and duration of posttransfusion Non-A, Non-B in

recipients. J.A.M.A. 246:2340-4.

220. Ennulat, D., M. Magid-Slav, S. Rehm, and K.S. Tatsuoka. 2010. Diagnostic performance of

traditional hepatobiliary biomarkers of drug-induced liver injury in the rat. Toxicol. Sci. 116:397-412,

221. Glantz, A., H.U. Marschall, and L.A. Mattsson. 2004. Intrahepatic cholestasis of pregnancy:

Relationships between bile acid levels and fetal complication rates. Hepatology 40:467-74.

222. Cowen, A.E., M.G. Korman, A.F. Hofmann, O.W. Cass, and S.B. Coffin. 1975. Metabolism of

lithocholate in healthy man. II. Enterohepatic circulation. Gastroenterology 69:67- 76.

223. Tappeiner, A.J.F.H. 1878. Ueber die Aufsaugung der Gallsäuren alkaline im Dunndarme. Wien.

Akad. Sitzber. 77:281-304

224. Verzar, F. 1936. Absorption from the intestine. Longmans, Green and Co. London.

225. Fröhlicher, E. 1936. Die Resorption von gallensäuren aus verschiedenen Dunndarmabschnitten.

Biochem. Z. 283:273-9.

226. Baker, R.D., and G.W. Searle. 1960. Bile salt absorption at various levels of rat small intestine.

Proc. Soc. Exp. Biol. Med. 105:521-3.

227. Wilson, T.H., and G. Wiseman. 1954. The use of sacs of everted small intestine for the study of the

transference of substances from the mucosal to the serosal surface. J. Physiol. 123:116-25.

228. Crane, R.K. 1960. Intestinal absorption of sugars. Physiol. Rev. 40:789-825.

229. Lack, L. and I.M. Weiner. 1961. In vitro absorption of bile salts by small intestine of rats and

guinea pigs. Am. J. Physiol. 200:313-7.

230. Holt, P.R. 1964. Intestinal absorption of bile salts in the rat. Amer. J. Physiol. 207:1-7.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

112

231. Lϋcke, H., G. Stange, R. Kinne, and H. Murer 1978. Taurocholate-sodium co-transport by brush-

border membrane vesicles isolated from rat ileum. Biochem. J. 174:951-8.

232. Kramer, W., and G. Kurz. 1983. Photolabile derivatives of bile salts. Synthesis and suitability for

photoaffinity labeling. J. Lipid Res. 24:910-23.

233. Kramer, W., S.B. Nicol, F. Girbig, U. Gutjahr, S.Kowalewski, and H. Fasold. 1992.

Characterization and chemical modification of the Na(+)-dependent bile-acid transport system in brush-

border membrane vesicles from rabbit ileum. Biochim. Biophys. Acta 1111:93-102.

234. Heidrich, H-G., R. Kinne, E. Kinne-Saffran, and K. Hannig. 1972. The polarity of the proximal

tubule cell in rat kidney. J. Cell Biol. 52:232-45.

235. Hediger, M.A., M. Coady, T.S. Ikeda, and E. Wright. 1987. Expression cloning and cDNA

sequencing of the Na+/glucose cotransporter. Nature 330: 379-81.

236. Wong, M.H., P. Oelkers, A.L. Craddock, and P.A. Dawson. 1994. Expression cloning and

characterization of the hamster ileal sodium-dependent bile acid transporter. J. Biol. Chem. 269:1340-7.

237. Wilson, F.A., G. Burckhardt, H. Murer, G. Rumrich, and K.J. Ulrich. 1981. Sodium-coupled

taurocholate transport in the proximal convolution of the rat kidney in vivo and in vitro. J. Clin. Invest.

67:1141-50.

238. Alpini, G., S.S. Glaser, R. Rodgers, J.L. Phinizy, W.E. Robertson, J. Lasater, A. Caligiuri, Z. Tretjak,

and G.D. LeSage. 1997. Functional expression of the apical Na+-dependent bile acid transporter in large,

but not small rat cholangiocytes. Gastroenterology 113:1734-40.

239. Lazaridis, K.N., L. Pham, P. Tietze, R.A. Marinelli, P.C. deGroen, S. Levine, P.A. Dawson, and N.F.

LaRusso. 1997. Rat cholangiocytes absorb bile acids at their apical domain via the ileal sodium-

dependent bile acid transporter. J. Clin. Invest. 100:2714-21.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

113

240. Kolhatkar, V., and J.E. Polli. 2012. Structural requirements of bile acid transporters: C-3 and C-7

modifications of steroidal hydroxyl groups. Eur. J. Pharm. Sci. 46:86-99.

241. Hu, N.J., S. Iwata, A.D. Cameron, and D. Drew. 2011. Crystal structure of a bacterial homologue

of the bile acid sodium symporter ASBT. Nature 478:408-11.

242. Doring, B., T. Lutteke, J. Geyer, and E. Petzinger. 2012. The SLC10 carrier family: transport

functions and molecular structure. Curr. Top. Membr. 70:105-68.

243. Hoffman, N.E., D.E. Donald, and A.F. Hofmann. 1975. Effect of primary bile acids on bile lipid

secretion from the perfused dog liver. Am. J. Physiol. 229:714 720.

244. Poupon R., R. Poupon, M. Dumont, and S. Erlinger. 1976. Hepatic storage and biliary transport

maximum of taurocholate and taurochenodeoxycholate in the dog. Eur. J. Clin. Invest. 30:431-7.

245. Van Dyke, R.W., J.E. Stephens, and B.F. Scharschmidt. 1982. Bile acid transport in cultured

hepatocytes. Am. J. Physiol. 243:G484-92.

246. Reichen, J., and G. Paumgartner. 1976. Uptake of bile acids by perfused rat liver. Am. J. Physiol.

231:734-42.

247. Hagenbuch, B., B. Stieger, M. Foguet, H. Lubbert, and P.J. Meier. 1991. Functional expression

cloning and characterization of the hepatocyte Na+/bile acid cotransport system. Proc. Natl. Acad. Sci.

USA 88:10629-33.

248. Geyer, J., T. Wilke, and E. Petzinger. 2006. The solute carrier family SLC10: more than a family of

bile acid transporters regarding function and phylogenetic relationships. Naynyn-Schmiedeberg’s Arch.

Pharmacol. 372:413-31.

249. Hagenbuch, B., and B. Stieger. 2013. The SLCO (former SLC21) superfamily of transporters.

Mol. Aspects Med. 34:396-412.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

114

250. Klaassen, C.D., and L. M. Aleksunes. 2010. Xenobiotic, bile acid and cholesterol transporters:

function and regulation. Pharm. Rev. 62:1-96.

251. Cowen, A.E., M.G. Korman, A.F. Hofmann, and P.J. Thomas. 1975. Plasma disappearance of

radioactivity after intravenous injection of labeled bile acids in man. Gastroenterology 68:1567-73.

252. Norman, A. 1970. 1970. Metabolism of glycocholic acid in man. Scand. J. Gastroenterol. 5:231-6.

253. Hepner, G.W., A.F. Hofmann, and P.J. Thomas. 1972. Metabolism of steroid and amino acid

moieties of conjugated bile acids in man. II. Glycine conjugated dihydroxy bile acids. J. Clin. Invest.

51:1898 1905.

254. Csanaky, I.L., H. Lu, Y. Zhang, K. Ogura, S. Choudhuri, and C.D. Klaassen. 2011. Organic anion-

transporting polypeptide 1b2 (Oatp1b2) is important for the hepatic uptake of unconjugated bile acids:

Studies in Oatp1b2-null mice. Hepatology 53: 272-81.

255. Hubbard, B., H. Doege, S. Punreddy, H. Wu, X. Huang, V.K. Kashik, R.L. Mozell, J.J. Bynes, A.

Stricker-Krongrad, C.J. Chou, L.A. Tartaglia, H.F. Lodish, A. Stahl, and R.E. Gimeno. 2006. Mice

depleted for fatty acid transport protein 5 have defective bile acid conjugation and are protected from

obesity. Gastroenterology 130:1259-69.

256. Steinberg, S.J., S.J. Mihalik, D.G. Kim, D.A. Cuebas, and P.A. Watkins. 2000. The human liver-

specific homolog of very long-chain acyl-CoA synthetase is cholate:CoA ligase. J. Biol. Chem.

275:15605-8.

257. Clayton, L.M., D. Gurantz, A.F. Hofmann, L.R. Hagey, and C.D. Schteingart. 1989. The role of

bile acid conjugation in hepatic transport of dihydroxy bile acids. J. Pharm. Exp. Therap. 248:1130

1137.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

115

258. Rius, M., J. Hummel-Eisenbeiss, A.F. Hofmann, and D. Keppler. 2005. Substrate specificity of

human ABCC4 (MRP4)-mediated cotransport of bile acids and reduced glutathione. Am. J. Physiol.

Gastrointest. Liver Physiol. 290:G640-9.

259. Sorscher, S., J. Lillienau, J.L. Meinkoth, J.H. Steinbach, C.D. Schteingart, J. Feramisco, and A.F.

Hofmann. 1992. Conjugated bile acid uptake by Xenopus Laevis oocytes induced by microinjection with

ileal Poly A+ mRNA. Biochem. Biophys. Res. Commun. 186:1455-1462.

260. Keppler, D. 2011. Cholestasis and the role of basolateral efflux pumps. Z. Gastroenterol.

49:1553-7.

261. Duane, W.C. 1975. The intermicellar bile salt concentration in equilibrium with the mixed micelles

of human bile. Biochim. Biophys. Acta 398:275-86.

262. Inoue, M., R. Kinne, T. Tran, L. Biempica, and I.M. Arias. 1983. Rat liver canalicular membrane

vesicles. Isolation and topological characterization. J. Biol. Chem. 258:5183-8.

263. Meier, P.J., E.S. Sztul, A. Reuben, and J.L. Boyer. 1984. Structural and functional polarity of

canalicular and basolateral plasma membrane vesicles isolated in high yield from rat liver. J. Cell. Biol.

98:991-1000.

264. Meier, P.J., A.S. Meier-Abt, C. Barrett, and J.L. Boyer. 1984. Mechanisms of taurocholate transport

in canalicular and basolateral rat liver plasma membrane vesicles. Evidence for an electrogenic

canalicular organic anion carrier. J. Biol. Chem. 259:10614-22.

265. Inoue, M., R. Kinne, T. Tran, and I.M. Arias. 1984. Taurocholate transport by rat liver canalicular

membrane vesicles. J. Clin. Invest. 73:659-663.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

116

266. O'Maille, E.R.L., and A.F. Hofmann. 1986. Relatively high biliary secretory maximum for non-

micelle forming bile acid: possible significance for mechanism of secretion. Quart . J. Exptl. Physiol.

71:475 482.

267. Adachi, Y., H. Kobayashi, Y. Kurumi, M. Shouji, M. Kitano, and T. Yamamoto. 1991. ATP-

dependent taurocholate transport by rat liver canalicular vesicles. Hepatology 14:655-9.

268. Nishida, T., Z. Gatmaitan, M. Che, and I.M. Arias 1991. Rat liver canalicular membrane vesicles

contain an ATP-dependent bile acid transport system. Proc. Natl. Acad. Sci. U.S.A. 88:6590-4.

269. Childs, S., R.L. Yeh, E. Georges, and V. Ling. 1995. Identification of a sister gene to P-

glycoprotein. Cancer Res. 55:2029-34.

270. Gerloff, T., B. Stieger, B. Hagenbuch, J. Madon, L. Landmann, J. Roth, A.F. Hofmann and P.J.

Meier. 1998, The sister of P-glycoprotein represents the canalicular bile salt export pump of mammalian

liver. J. Biol. Chem. 273:10046-10050.

271. Strautnieks, S.S., L.N. Bull, A.S. Knisely, S.A. Kocoshis, N. Dahl, H. Arnell, E. Sokal, K. Dahan, S.

Childs, V. Ling, M.S. Tanner, A.F. Kagalwalla, A. Nemeth, J. Pawlowska, A. Baker, G. Mieli-Vergani,

N.B. Freimer, R.M. Gardiner, and R. J. Thompson. 1998. A gene encoding a liver-specific ABC

transporter is mutated in progressive familial intrahepatic cholestasis. Nat. Genet. 20:233-8.

272. Akita, H., H. Suzuki, K. Ito, S. Kinoshita, N. Sato, H. Takikawa, and Y. Sugiyama. 2001.

Characterization of bile acid transport mediated by multidrug resistance associated protein 2 and bile salt

export pump. Biochim. Biophys. Acta 1511:7-16.

273. Wang, R., M. Salem, I.M. Yousef, B. Tuchweber, P. Lam, S.J. Childs, C.D. Helgason, C. Ackerley,

M.J. Phillips, and V. Ling. 2001. Targeted inactivation of sister of P-glycoprotein gene (spgp) in mice

results in nonprogressive but persistent intrahepatic cholestasis. Proc. Natl. Acad. Sci. U.S.A. 98: 2011-8.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

117

274. Wang, R., H.I. Chen, L. Liu, J.A. Sheps, M.J. Phillips, and V. Ling. 2009. Compensatory role of P-

glycoproteins in knockout mice lacking the bile salt export pump. Hepatology 50:948-56.

275. Perwaiz, S., D. Forrest, D. Magnault, B. Tuchweber, M.J. Phillips, R. Wang, V. Ling and I.M.

Yousef. 2003. Appearance of atypical 3 alpha, 6 beta, 7 beta, 12 alpha-tetrahydroxy-5 beta-cholan-24-

oic acid in spgp knockout mice. J. Lipid Res. 44:494-502.

276. Wang, R., L. Liu, J. Sheps, D. Forrest, A.F. Hofmann, L.R. Hagey, and V. Ling. 2013. Defective

canalicular transport and toxicity of dietary ursodeoxycholic acid in the Abcb11-/- mouse: transport and

gene expression studies. Am. J. Physiol. 305:G286-94.

277. Megaraj, V., T. Iida, P. Jungsuwadee, A.F. Hofmann, and M. Vore. 2010. Hepatobiliary disposition

of 3α,6α,7α.12α-tetrahydroxy-cholanoyltaurine: a substrate for multiple canalicular transporters. Drug

Metab. Dispos. 38:1723-30.

278. Wang, R., P. Lam, L. Liu, D. Forrest, I.M. Yousef, D. Mignault, M.J. Phillips, and V. Ling. 2003.

Severe cholestasis induced by cholic acid feeding in knockout mice of sister of P-glycoprotein.

Hepatology 38:1489-99.

279. Zhang, Y., F. Li, A.D. Patterson, Y. Wang, K.W. Krausz, G. Neale, S. Thomas, D. Nachagari, P.

Vogel, M. Vore, F.J. Gonzalez, and J.D. Schuetz. 2012. Abcb11 deficiency induces cholestasis coupled to

impaired β-fatty acid oxidation in mice. J. Biol. Chem. 287:24784-94.

280. Wang, W., D.J. Seward, L. Li, J.L. Boyer, and N. Ballatori. 2001. Expression cloning of two genes

that together mediate organic solute and steroid transport in the liver of a marine vertebrate. Proc. Nat.

Acad. Sci. U.S.A. 98:9431-6.

281. Dawson, P.A., M. Hubbert, J. Haywood, A.L. Craddock, N. Zerangue, W.V. Christian, and N.

Ballatori. 2005. The heteromeric organic solute transporter α-β, Ostα-Ostβ, is an ileal basolateral bile

acid transporter. J. Biol. Chem. 280:6960-6968.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

118

282. Soroka, C.J., N. Ballatori, and J.L. Boyer. 2010. Organic solute transporter, OST alpha-OST beta:

its role In bile acid transport and cholestasis. Semin. Liver Dis. 30:175-85.

283. Rao, A., J. Haywood, A.L. Craddock, M.G. Belinsky, G.D. Kruh, and P.A. Dawson. 2008. The

organic solute transporter alpha-beta, Ostalpha-Ostbeta, is essential for intestinal bile acid transport and

homeostasis. Proc. Natl. Acad. Sci. U.S.A. 105:3891-6.

284. Dawson, P.A., J. Haywood, A.L. Craddock, M. Wilson, M. Tietjent, K. Kluckman, N. Maeda, and

J.S. Parks. 2003. Targeted deletion of the ileal bile acid transporter eliminates enterohepatic cycling of

bile acids in mice. J. Biol. Chem. 278:33920-7.

285. Lan, T., J. Haywood, A. Rao, and P.A. Dawson. 2011. Molecular mechanisms of altered bile acid

homeostasis in organic solute transporter-alpha knockout mice. Dig. Dis. 29:18-22.

286. Frisch, K., S.Jakobsen, M. Sǿrenson, O. Lajord Munk, A.K.O. Alstrup, P. Ott, A.F. Hofmann, and S.

Keiding. 2012. [N-methyl-11C]Cholylsarcosine, a novel bile acid tracer for PET/CT of hepatic

excretory function: radiosynthesis and proof-of-concept studies in pigs. J. Nucl. Med. 53:1-7.

287. Hepatobiliary Transport in Health and Disease. 2012. D. Häussinger, V. Keitel, and R. Kubitz, ed.

De Gruyter, Boston. 292 pp.

288. Borelli, G. De Motu Animalium, Pars altera. Rome: Angeli Bernabo, 1681; 298-317. [On the

movement of animals, 5th ed. The Hague: Peter Gosse, 1743]. Berlin: Springer; 1989, 354-64.

289. Liebig, J. 1843. Animal chemistry, or chemistry in its applications to physiology and pathology.

2nd ed. London. Taylor and Watson.

290. Lehmann, C.G. 1855. Physiological chemistry. 2nd ed. Vol. 1. Philadelphia: Blanchard and Lea.

291. Schiff, M. 1870. Bericht ϋber einige Versuchsreihen. I. Gallenbildung abhängig von der

Aufsäugung der Gallstoffe. Pflugers Arch. [Physiol. 3:598-613.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

119

292. Carey, M.C. and W.C. Duane. 1994. Enterohepatic Circulation. In The Liver: Pathobiology. Third

Edition. Ed. I.M. Arias, J.L. Boyer, N. Fausto, W.B. Jakoby, D.A. Schacter, and D.A. Shafritrz. Raven

Press, Ltd. New York.

293. Back, P. 1988, Urinary bile acids. In Setchell, K.D.R., D. Kritchevsky, and P.P. Nair, eds. The Bile

Acids., vol. 4. Plenum Press 1988; 405-40.

294. Hofmann, A.F. and N.E. Hoffman. 1974. Measurement of bile acid kinetics by isotope dilution in

man. Gastroenterology 67:314-323.

295. Stellaard, F., G. Brufau, R. Boverhof, E.Z. Jonkers, T. Boer, and F. Kuipers. 2009. Developments in

bile acid kinetic measurements using (13)C and (2)H: 10(5) times improved sensitivity during the last 40

years. Isotopes Environ. Health Stud. 45:275-88.

296. LaRusso, N.F., P.A. Szczepanik, A.F. Hofmann, and S.B. Coffin. 1977. Effect of deoxycholic acid

ingestion on bile acid metabolism and biliary lipid secretion in normal subjects. Gastroenterology

72:132-140.

297. Van der Werf, S.D., F.M. Nagengast, G.P. van Berge Henegouwen, A.W. Huijbregts, and J.H. van

Tongeren. 1982. Colonic absorption of secondary bile-acids in patients with adenomatous polyps and in

matched controls. Lancet 1:759-62.

298. Bergstrom, S., M. Rottenberg, and J. Voltz. 1953. The preparation of some carboxyl-labeled bile

acids. Acta Chem. Scand. 7:481-4.

299. Hofmann, A.F. P.A. Szczepanik, and P.D. Klein. 1968. Rapid preparation of tritium labeled bile

acids by enolic exchange on basic alumina containing tritiated water. J. Lipid Res. 9:707 713.

300. LaRusso, N.F., N.E. Hoffman, and A.F. Hofmann. 1974. Validity of using 2,4-3H labeled bile acids

to study bile acid kinetics in man. J. Lab. Clin. Med. 84:759 765.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

120

301. Ng, P.Y., R.N. Allan, and A.F. Hofmann AF. 1977. Suitability of 11,12 3H- chenodeoxycholic acid

and 11,12 3H-lithocholic acid for isotope dilution studies of bile acid metabolism in man. J. Lipid Res.

18:753 758.

302. Duane, W.C., C.D. Schteingart, H-T. Ton-Nu, and A.F. Hofmann. 1996. Validation of [22,23-3H]-

cholic acid as a stable tracer through conversion to deoxycholic acid in human subjects. J. Lipid Res. 37:

431-436, 1996.

303. Kakiyama, G., T. Iida, A. Yoshimuta, T. Goto, N. Mano, J. Goto, T. Nambara, L. R. Hagey, and A.F.

Hofmann. 2004. Chemical synthesis of (22E)-3α,6β,7β-trihydroxy-5β-chol-22-en-24-oic acid and its

taurine and glycine conjugates: a major bile acid in the rat. J. Lipid Res. 45:567-73.

304. Go, V.L.W., A.F. Hofmann, and W.H.J. Summerskill. 1970. Simultaneous measurements of total

pancreatic, biliary, and gastric outputs in man using a perfusion technique. Gastroenterology 58:321-328.

305. Brunner, H., T.C. Northfield, V.L.W. Go, and W.H.J. Summerskill. 1974. Gastric emptying and

secretion of bile acids, cholesterol, and pancreatic enzymes during digestion: duodenal perfusion studies

in healthy subjects. Mayo Clin. Proc. 49:851-860.

306. Van Berge Henegouwen, G.P., and A.F. Hofmann. 1978. Nocturnal gallbladder storage and emptying

in gallstone patients and healthy subjects. Gastroenterology 75:879-885.

307. Northfield T.C., and A.F. Hofmann. 1973. Biliary lipid secretion in gallstone patients. Lancet 1:747

748, 1973.

308. Angelin, B., I. Björkhem, K. Einarsson, and S. Ewerth. 1982. Hepatic uptake of bile acids in man.

Fasting and postprandial concentrations of individual bile acids in portal venous and systemic blood

serum. J Clin. Invest. 70 724-31.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

121

309. Korman, M.G., N.F. LaRusso, N.E. Hoffman, and A.F. Hofmann. 1975. Development of an

intravenous bile acid tolerance test: plasma disappearance of cholylglycine in health. N. Engl. J. Med.

292:1205 1209.

310. Pries, J.M., C.A. Sherman, G.C. Williams, and R.F. Hanson. 1979. Hepatic extraction of bile salts in

conscious dog. Am. J. Physiol. 236:E191-97.

311. Van Berge Henegouwen, G.P., and Hofmann, A.F. 1977. Pharmacology of chenodeoxycholic acid.

II. Absorption and metabolism. Gastroenterology 73:300-309.

312. Marigold, J.H., H.J. Bull, I.T. Gilmore, D.J. Coltart, and R.P.H. Thompson. 1982. Direct

measurement of hepatic extraction of chenodeoxycholic acid and ursodeoxycholic acid in man. Clin. Sci.

63:197-203.

313. Lamri, Y., S. Erlinger, M. Dumont, A. Roda, and G. Feldmann. 1992. Immunoperoxidase

localization of ursodeoxycholic acid in rat biliary epithelial cells. Evidence for a cholehepatic circulation.

Liver 12:351-4.

314. Wheeler, H.O., E.D. Ross, and S.E. Bradley. 1968. Canalicular bile production in dogs. Am. J.

Physiol. 214:866-74.

315. Dumont, M., S. Erlinger, and S. Uchman. 1980. Hypercholeresis induced by ursodeoxycholic acid

and 7-ketolithocholic acid in the rat: possible role of bicarbonate transport. Gastroenterology 79:82-9.

316. Fracchia, M., K.D.R. Setchell, A. Crosignani, M. Podda, N. O'Connell, R. Ferraris, A.F. Hofmann,

and G. Galatola. 1996. Bile acid conjugation in cholestatic liver disease before and during treatment with

ursodeoxycholic acid. Clin. Chim. Acta 248:175-185. 1996.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

122

317. Bolder, U., N.V. Trang, L.R. Hagey, C.D. Schteingart, H-T. Ton-Nu, C. Cerrè, R.O. Elferink, and

A.F. Hofmann. 1999. Sulindac is excreted into bile by a canalicular bile salt pump and undergoes a

cholehepatic circulation in rats. Gastroenterology 117:962-71.

318. LaRusso, N.F., N.E. Hoffman, M.G. Korman, A.F. Hofmann, and A.E. Cowen. 1978. Determinants

of fasting and postprandial serum bile acid levels in healthy man. Am. J. Dig. Dis. 23:385-391.

319. Hofmann, A.F., and J.R. Poley. 1972. Role of bile acid malabsorption in pathogenesis of diarrhea

and steatorrhea in patients with ileal resection. I. Response to cholestyramine or replacement of dietary

long chain triglyceride by medium chain triglyceride. Gastroenterology 62:918-934.

320. Grundy, S.M., E.H. Ahrens, and G. Salen. 1971. Interruption of the enterohepatic circulation of bile

acids in man: comparative effects of cholestyramine and ileal exclusion on cholesterol metabolism. J.

Lab. Clin. Med. 78:94-121.

321. Dowling, R.H., E. Mack, and D.M. Small. 1970. Effects of controlled interruption of the

enterohepatic circulation of bile salts by biliary diversion and by ileal resection on bile salt secretion,

synthesis, and pool size in the Rhesus monkey. J. Clin. Invest. 49:232-242.

322. Pandak, W.M., D.M. Heuman, P.B. Hylemon, J.Y. Chiang, and Z.R. Vlahcevic. 1995. Failure of

intravenous infusion of taurocholate to downregulate cholesterol 7α hydroxylase in rats with biliary

fistulas. Gastroenterology 108:533-44.

323. Nagano, M., S. Kuroki, A. Mizuta, M. Furukawa, M. Noshiro, K. Chiijiwa, and M. Tanaka. 2004.

Regulation of bile acid synthesis under reconstructed enterohepatic circulation in rats. Steroids 69:701-9.

324. Shang, Q., G.L. Guo, A. Honda, M. Saumoy, G. Salen, and G. Xu. 2013. FGF15/19 protein levels in

the portal blood do not reflect changes in the ileal FGF 15/19 or hepatic Cyp7A1 mRNA levels. J. Lipid.

Res. 54:2606-14.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

123

325. Lillienau J. Munoz, S.J. Longmire-Cook, L.R. Hagey, D.L. Crombie, and A.F. Hofmann. 1993.

Negative feedback regulation of the ileal bile acid transport system in rodents. Gastroenterology 104:38-

46, 1993.

326. Sauer, P., A. Stiehl, B.A. Fitscher, H.D. Riedel, C. Benz, P. Klöters-Platchky, S. Stengelin, W.

Stremmel, and W. Kramer. 2000. Downregulation of ileal bile acid absorption in bile-duct-ligated rats.

J. Hepatol. 33:2-8.

327. Matsumura, J., M.A. Greiner, D.L. Nahrwold, and L.G. Dawes. 1993. Reduced ileal taurocholate

absorption with total parenteral nutrition. J. Surg. Res. 54:517-22.

328. The physical chemistry of bile acids in health and disease. 1984. Hepatology 4:1S-252S.

(supplemental issue).

329. Hofmann, A.F. and D.M. Small. 1967. Detergent properties of bile salts: correlation with

physiological function. Ann. Rev. Med. 18:333-376.

330. Carey, M.C. Physical-chemical properties of bile acids and their salts. 1985. In Sterols and Bile

Acids. H. Danielsson and J. Sjövall, eds. Elsevier, Amsterdam. 345-404.

331. Roda, A., and A. Fini. 1984. Effect of nuclear hydroxyl substituents on aqueous solubility and acidic

strength of bile acids. Hepatology (suppl.) 4:72S-76S.

332. Hofmann, A.F. 1994. Pharmacology of ursodeoxycholic acid, an enterohepatic drug. Scand. J.

Gastroenterol. 29 (suppl. 204):1-15.

333. Small, D.M. 1971. The physical chemistry of cholanic acids. In The Bile Acids: Chemistry,

Physiology, and Metabolism. Ed. P.P. Nair and D. Kritchevsky. Plenum Press, New York. 249-356.

334. Igimi, H., and M.C. Carey. 1980. pH-solubility relations of chenodeoxycholic and ursodeoxycholic

acids: physical-chemical basis for dissimilar solution and membrane phenomena. J. Lipid Res. 21:72-90.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

124

335. Josephson, B.A. 1933. Studien ϋber die gallensäuren und angehorige verbindungen.

Centraltryckeriet, Stockholm. (doctoral thesis).

336. Ekwall, P., T. Rosendahl, and B. Löfman. 1957. Studies on bile salt solutions. I. The dissociation

constants of the cholic and deoxycholic acids. Acta Chem. Scand. 11:590-9.

337. Fini, A., and A. Roda. 1987. Chemical properties of bile acids. IV. Acidity constants of glycine-

conjugated bile acids. J. Lipid Res. 28:755-9.

338. Lillienau, J., C.D. Schteingart, and A.F. Hofmann. 1992. Physicochemical and physiological

properties of cholylsarcosine: a potential replacement detergent for bile acid deficiency states in the small

intestine. J. Clin. Invest. 89:420-431.

339. Hofmann, A.F. and K.J. Mysels. 1992. Bile acid solubility and precipitation in vitro and in vivo:

the role of conjugation, pH and Ca2+ ions. J. Lipid Res. 33:617-626.

340. van Berge Henegouwen, G.P., A.F. Hofmann, and T.S. Gaginella. 1977. Pharmacology of

chenodeoxycholic acid. I. Pharmaceutical properties. Gastroenterology 73:291- 299.

341. Go, V. L. W., J.R. Poley, A.F. Hofmann, and W.H.J. Summerskill. 1970. Disturbances of fat

digestion induced by acid jejunal pH due to gastric hypersecretion in man. Gastroenterology 58:638-646.

342. Spriggs, M., K.A. Thompson, K. Volle, I. Stasiak, L. Beyea, A. Guthrie, D. Barton, J. Talley, A.

Roda, C. Camborata, A.F. Hofmann, and L.R. Hagey. 2014. Gastrolithiasis in prehensile-tailed

porcupines (Coendou Prehensilis): 9 cases and pathogenesis of stone formation. Am. J. Vet. Med.

(submitted)

343. Gu, J-J., A.F. Hofmann, H-T. Ton-Nu, C.D. Schteingart, and K.J. Mysels. 1992. Solubility of

calcium salts of unconjugated and conjugated natural bile acids. J. Lipid Res. 33:635-46.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

125

344. Jones, C., A.F. Hofmann, K.J. Mysels, and A. Roda. 1986. The effect of calcium and sodium ion

concentration on the properties of dilute aqueous solutions of glycine conjugated bile salts. J. Colloid

Interface Sci. 114:452-470.

345. Shiffman, M., H.J. Sugermen, J.M. Kellum, and E.W. Moore. 1992. Calcium in human gallbladder

bile. J. Lab. Clin. Med. 120:875-84.

346. Hofmann, A.F. 1984. Animal models of calcium cholelithiasis. Hepatology 4:209S-11S.

347. Palmer, R.H., and Z. Hruban. 1966. Production of bile duct hyperplasia and gallstones by

lithocholic acid. J. Clin. Invest. 45:1255-67.

348. Zaki, F.G., J.B. Carey, F.W. Hofbauer, and C. Nokolo. 1967. Biliary reaction and

choledocholithiasis induced in the rat by lithocholic acid. 1967. J. Lab. Clin. Med. 69:737-48.

349. Cohen, B.I., N. Ayyad, E.H. Mosbach, C.K. McSherry, N. Matoba, A.F. Hofmann, H-T. Ton-Nu, Y.

Peng, C.D. Schteingart, and R.J. Stenger. 1981. Replacement of cholesterol gallstones by

murideoxycholyl taurine gallstones in prairie dogs fed murideoxycholic acid. Hepatology 14:158-168.

350. Hofmann, A.F., and H.S. Mekhjian. 1973. Bile acids and the intestinal absorption of fat and

electrolytes in health and disease. In The Bile Acids. Ed. P.P. Nair and D. Kritchevsky New York:

Plenum Press, vol II, pp 103-152.

351, Hartley, G.S. 1936. Aqueous solutions of paraffin-chain salts. A study in micelle formation. In

Actualites Scientifiques et Industrielles. 387. Hermann and Co., Editeurs, Paris.

352. McBain, E.L. and E. Hutchinson. Solubilization and related phenomena: Physical Chemistry. 1955.

New York, Academic Press.

353. Bashour, J.T., and L. Bauman. 1937. The solubility of cholesterol in bile salt solutions. J. Biol.

Chem. 121:1-3.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

126

354. Mellander, S., and E. Stenhagen. 1942. The state of bile salt solutions. II. Conductivity studies on

dilute solutions of sodium taurocholate at 25o C. Acta Physiol. Scand. 4:349-61.

355. Ekwall, P. Micelle formation in sodium cholate solutions. 1951. Acta Acad. Aboensis Math. Phys.

17:3-10.

356. In memoriam: P. Ekwall. 1991. Colloid. Polym. Sci. 289:419-20.

357. Norman, A. 1960. The beginning solubilization of 20-methylcholanthrene in aqueous solutions of

conjugated and unconjugated bile salts. Acta Chem. Scand. 14:1300-9.

358. Hofmann, A.F. 1963. The function of bile salts in fat absorption: the solvent properties of dilute

micellar solutions of conjugated bile salts. Biochem. J. 89:57-68.

359. Kratohvil, J. 1986. Size of bile salt micelles: techniques, problems, and results. Adv. Colloid

Interface Sci. 26:131-54.

360. Roda, A., A.F. Hofmann, and K.J. Mysels. 1983. The influence of bile salt structure on self

association in aqueous solutions. J. Biol. Chem. 258:6362-6370.

361. Iida, T., and F.C. Chang. 1982. Potential bile acid metabolites. 6. Stereoisomeric 3,7-dihydroxy-5β-

cholanoic acids. J. Org. Chem. 47:2966-72.

362. Fickert, P., A. Fuchsbichler, M. Wagner, G. Zollner, A. Kaser, H. Tilg, R. Krause, F. Lammert, C.

Langner, K. Zatloukal, H-U. Marschall, H. Denk, and M. Trauner. 2004. Regurgitation of bile acids from

leaky bile ducts causes sclerosing cholangitis in Mdr2 (Abcb4) knockout mice. 2004. Gastroenterology

127:261-74.

363. Poupon, R., O. Rosmorduc, P.Y. Boelle, Y. Chretien, C. Corbechot, O. Chazouilleres, C. Housset,

and V. Barbu. 2013. Genotype-phenotype relationships in the low-phospholipid associated cholelithiasis

syndrome: a study of 156 consecutive patients. Hepatology 58:1105-10.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

127

364. Polonovski, M., and R. Bourillon. 1952. Les phospholipids de la bile. Bull. Str. Chim. Biol. 34:712-

9.

365. Phillips, G.B. 1960. The lipid composition of human bile. Biochim. Biophys. Acta. 41:361-3.

366. Isaksson, B. 1954. On the dissolving power of lecithin and bile salts for cholesterol in human

bladder bile. Act Soc. Med. Ups. 59:296-306.

367. Yoshimuta, S. 1960. Factors affecting solubility of cholesterol in bile. Fukuoka Acta Med. 51:510-

28.

368. Tamasue, N., T. Inoue, and K. Juniper. 1973. Solubility of cholesterol in bile salt-lecithin model

systems. Am. J. Dig. Dis. 18:670-8.

369. Small, D.M., S.A. Penkett, and D. Chapman. 1969. Studies on simple and mixed bile salt micelles

by nuclear magnetic resonance spectroscopy. Biochim. Biophys. Acta 176:178-89.

370. Lindblom, G., P-O. Eriksson and G. Arvidson. 1984. Molecular organization in phases of lecithin-

cholate-water as studied by nuclear magnetic resonance. Hepatology 4:129S-138S.

371. Nichols, J.W., and J. Ozarowski. 1990. Sizing of lecithin-bile salt mixed micelles by size-exclusion

high performance liquid chromatography. Biochemistry 29:4600-6.

372, Hjelm, P., P. Thiyagarajan, and H. Alkan. 1992. Organization of phosphatidylcholine and bile salt in

rodlike mixed micelles. J. Phys. Chem. 96:8653-61.

373. Marrink, S.J. and A.E. Mark. 2002. Molecular dynamics simulation of mixed micelles modeling

human bile. Biochemistry 41:5375-82.

374. Long M.A., E.W. Kaler, and S.P. Lee. 1994. Structural characterization of the micelle-vesicle

transition in lecithin-bile salt solutions. Biophys. J. 67:1733-42.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

128

375. Heuman, D., R.S. Bajaj, and Q. Lin. 1996. Adsorption of mixtures of bile salt taurine conjugates to

lecithin-cholesterol membranes: implications for bile salt toxicity and cytoprotection. J. Lipid Res.

37:562-73.

376. Admirand, W.H., and D.M. Small. 1968. The physicochemical basis of cholesterol gallstone

formation in man. J. Clin. Invest. 47:1043-52.

377. Rozeboom, H.W.B. 1894. Graphische darstellung der heterogenen systeme aus ein bis vier stoffen,

mit einschluss der chemischen umsetzung. Ztrschr. F. Physik. Chemie 15:145-58.

378. Ahrens, E.H. 1957. Seminar on atherosclerosis: nutritional factors and serum lipid levels. Am. J.

Med. 23:928-52.

379. Mazer, N.A., G.B. Benedek, and M.C. Carey. 1980. Quasieleastic light-scattering studies of

aqueous biliary lipid systems. Mixed micelle formation in bile salt-lecithin solutions. Biochemistry

19:601-15.

380. Mazer, N.A. and M.C. Carey. 1983. Quasi-elastic light scattering studies of aqueous biliary lipid

systems. Cholesterol solubilization and precipitation in model bile solutions. Biochemistry 22:426-42.

381. Somjen, G.J., R. Rosenberg, and T. Gilat. 1990. Gel filtration and quasielestic light scattering

studies of human bile. Hepatology 12:123S-29S.

382. Halpern, Z., M.A. Dudley, M.P. Lynn, J.M. Nader, A.C. Breuer, and R.T. Holzbach. 1986. Vesicle

aggregation in model systems of supersaturated bile: relation to crystal nucleation and lipid composition

of the vesicular phase. J. Lipid Res. 27:295-306.

383. Higuchi, W.I., C-L. Liu, Y. Adachi, N.A. Mazer, and P.H. Lee. 1990. Equilibrium dialysis studies

on aqueous taurocholate-lecithin solutions: further validation of the method. Hepatology 12:45S-50S.

384. Biliary Cholesterol Transport and Precipitation. 1989. Hepatology 12:1S-244S. Supplement.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

129

385. Strasberg, S.M. and P.R.C. Harvey. 1990. Biliary cholesterol transport and precipitation:

Introduction and overview of conference. Hepatology 12:1S-5S.

386. Donovan, J.M., and M.C. Carey. 1990. Separation and quantitation of cholesterol carriers in bile.

Hepatology 12:94S-105S.

387. Schriever C.E., and D. Jϋngst. 1989. Association between cholesterol-phospholipid vesicles and

cholesterol crystals in human bile. Hepatology 9:541-6.

388. Lee, P.H., W.I. Higuchi, Y. Adachi, and N.A. Mazer. 1992 . Mechanisms of cholesterol

monohydrate dissolution in aqueous taurocholate, taurochenodeoxycholate, and tauroursodeoxycholate-

lecithin solutions: correlation between micellar species and dissolution rates. J. Colloid. Interface Sci.

154:35-50.

389. Hegardt, F.G. and H. Dam. 1971. The solubility of cholesterol in aqueous solutions of bile salts and

lecithin. Z. Ernahrungswiss. 10:223-33.

390. Holzbach, R.T., M. Marsh, M. Olszewski, and K. Holan. 1973. Cholesterol solubility in bile:

evidence that supersaturated bile is frequent in healthy man. J. Clin. Invest. 52:1467-79.

391. Dam, H., I. Prange, and E. Sondergaard. 1972. Alimentary production of gallstones in hamsters.

XXIV. Influence of orally ingested chenodeoxycholic acid and hyodeoxycholic acid on formation of

gallstones. Z. Ernahrungswiss. 11:80-94.

392, Dam, H., I. Kruse, I. Prange, H.E. Kallehauge, H.J. Fenger, and M. Krogh Jensen. 1971. Studies on

human bile. III. Composition of duodenal bile from healthy young volunteers compared with

composition of bladder bile from surgical patients with and without uncomplicated gallstone disease. Z.

Ernahrungswiss. 10:160-77.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

130

393. Carey, M.C. 1978. Critical tables for calculating the cholesterol saturation of native bile. J. Lipid

Res. 19:945-55.

394. Carey, M.C. and G. Ko. 1979, The importance of total lipid concentration in determining cholesterol

solubility in bile and the development of critical tables for calculating “% cholesterol saturation” with a

correction factor for ursodeoxycholate-rich bile. In Biological Effects of bile Acids, G. Paumgartner, A.

Stiehl, and W. Gerok, eds. MTP Press, Lancaster. 299-308.

395. Mattson, F.H., and L.W. Beck. 1956. The specificity of pancreatic lipase for the primary hydroxyl

groups of glycerides. J. Biol. Chem. 219:735-40.

396. Mattson, F.H., and R.A. Volpenhein. 1964. The digestion and absorption of triglycerides. J. Biol.

Chem. 239:2772-7.

397. Hofmann A.F., and B. Borgström. 1963. Hydrolysis of long chain monoglycerides in micellar

solution by pancreatic lipase. Biochim Biophys Acta 70:317-331.

398. Svärd, M., P. Schurtenberger, K. Fontell, B. Jonsson, B. Lindman. 1988. Micelles, vesicles, and

liquid crystals in the monolein-sodium taurocholate-water system: phase behavior, NMR, self-diffusion,

and quasi-elastic light scattering studies. J. Phys. Chem. 92:2261-70.

399. Hofmann, A.F. and B. Borgström. 1964. The intraluminal phase of fat digestion in man: the lipid

content of the micellar and oil phases of intestinal content obtained during fat digestion and absorption. J.

Clin. Invest. 43:247-257.

400. Staggers, J.E., O. Hernell, R.J. Stafford, and M.C. Carey. 1990. Physical-chemical behavior of

dietary and biliary lipids during intestinal digestion and absorption: 1. Phase behavior and aggregation

states of model lipid systems patterned after aqueous duodenal contents of healthy adult human beings.

Biochemistry 29:2028-40.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

131

401. Hernell, O, J.E. Staggers, and M.C. Carey. 1990. Physical-chemical behavior of dietary and biliary

lipids during intestinal digestion and absorption.2. Phase analysis and aggregation states of luminal lipids

during duodenal fat digestion in healthy adult human beings. Biochemistry 29:2041-56.

402. Lawrence, A.S.C. 1959. The mechanism of detergence. Nature 103:1491-4.

403. Hjelm R.P., C.D. Schteingart, A.F. Hofmann, and D.S. Silvia. 1995. Form and structure of self-

assembling particles in monoolein-bile salt mixtures. J. Phys. Chem. 99:16395-16400.

404. Hjelm, R.P., C.D. Schteingart, A.F. Hofmann, and P. Thiyagarajan. 2000. The structure of

conjugated bile salt-fatty acid-monoglyceride mixed colloids: studies by small-angle neutron scattering. J.

Phys. Chem.104:197-211.

405. Annegers, J.H. 1954. Function of pancreatic juice and bile in assimilation of dietary triglyceride.

A.M.A. Arch. Intern. Med. 93:9-22.

406. Porter, H.P., D.R. Saunders, G. Tytgat, O. Brunser, and C.D. Rubin. 1971. Fat absorption in bile

fistula man. A morphological and biochemical study. Gastroenterology 60:1008-19.

407. Lucassen, J. 1966. Hydrolysis and precipitates in carboxylate soap solutions. J. Phys. Chem.

70:1824-30.

408. Heydorn, S., P.B. Jeppesen, and P.B. Mortensen. 1999. Bile acid replacement therapy with

cholylsarcosine for short-bowel syndrome. Scand. J. Gastroenterol. 34:818-23.

409. Harkins, R.W. C.H. Whiteside, H.B. Fluckiger, and H.P. Sarett. 1965. Fat utilization in rats fed

cholestyramine, a bile acid sequestrant. Proc. Soc. Exp. Biol. Med. 118:399-402.

410. Johnston, C.G., and F. Nakayama. 1957. Solubility of cholesterol and gallstones in metabolic

material. Arch. Surg. 75:436-42.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

132

411. Nakayama, F. 1997. Cholelithiasis: Causes and Treatment. Igaku-Shoin. Tokyo. ISBN:

4260143344.

412. Heywood, R., A.K. Palmer, C.V. Foll, and M.R. Lee. 1973. Pathological changes in fetal Rhesus

monkey induced by oral chenodeoxycholic acid. Lancet 2:1021 (Letter to the Editor).

413. Bell, G.D., H.Y.I. Mok, M. Thwe, G.M. Murphy, K. Henry, and R.H. Dowling. 1974. Liver structure

and function in cholelithiasis: effect of chenodeoxycholic acid. Gut 15:165-72.

414. Cowen A.E. M.G. Korman, A.F. Hofmann, and O.W. Cass. 1975. Metabolism of lithocholate in

healthy man. I. Biotransformation and biliary excretion of intravenously administered lithocholate,

lithocholylglycine, and their sulfates. Gastroenterology 69:59- 66.

415. Allan, R.N., J.L. Thistle, and A.F. Hofmann. 1976. Lithocholate metabolism during chemotherapy

for gallstone dissolution. II. Absorption and sulphation. Gut 17:413-19.

416. Gadacz, T.R., R.N. Allan, E. Mack, and A.F. Hofmann. Impaired lithocholate sulfation in the rhesus

monkey: a possible mechanism for chenodeoxycholate toxicity. Gastroenterology 70:1125 1129, 1976.

417. Webster, K.H., M.C. Lancaster, A.F. Hofmann, D.R. Wease, and A.H. Baggenstoss. 1975. Influence

on primary bile acid feeding on cholesterol metabolism and hepatic function in the rhesus monkey. Mayo

Clin. Proc. 50:134-138.

418. Fischer, C.D., N.S. Cooper, M.A. Rothschild, and E.H. Mosbach. 1974. Effect of dietary

chenodeoxycholic acid and lithocholic acid in the rabbit. Am. J. Dig. Dis. 18:877-86.

419. Dyrska, H., T. Chen, G. Salen, and E.H. Mosbach. 1975. Toxicity of chenodeoxycholic acid in the

Rhesus monkey. Gastroenterology 69:333-7.

420. Morrisey, K.P., C.K. McSherry, R.L. Swarm, W.H. Nieman, and J.E. Deitrick. 1975. Toxicity of

chenodeoxycholic acid in the nonhuman primate. Surgery 77:851-60.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

133

421. Schoenfield, L.J., J.M. Lachin, the Steering Committee, and the National Cooperative Gallstone

Study Group. 1981. Chenodiol (chenodeoxycholic acid) for dissolution of gallstones: The National

Cooperative Gallstone Study. Ann. Intern. Med. 95:257-282, 1981.

422. Palmer, R.H. and M.C. Carey. 1982. Sounding Board. An optimistic view of the National

Cooperative Gallstone Study. N. Engl. J. Med. 306:1171-4.

423. Fisher, R.L. A.F. Hofmann, J.L. Converse, S.S. Rossi, and S.P. Lan. 1991. Lack of relationship

between hepatotoxicity and lithocholic acid sulfation during chenodiol therapy in the National

Cooperative Gallstone Study. Hepatology 14:454-463.

424. Erlinger, S., A. Le Go, J.M. Husson, and J. Fevery. 1984. Franco-Belgians cooperative study if

ursodeoxycholic acid in the medical dissolution of gallstones: a double-blind randomized dose-response

study and comparison with chenodeoxycholic acid. Hepatology 4:308-14.

425. Roda, E., D. Festi, A. Roda, C. Sama, R. Aldini, and G. Mazzella. 1977. Results of chenic acid

therapy in patients with cholesterol calculi. Min. Med. 68:3027-9.

426. Higuchi, W.I., F. Sjuib, D. Mufson, A.P. Simonelli, and A.F. Hofmann.1973. Dissolution kinetics of

gallstones. Physical model approach. J. Pharm. Sci. 62:942-45.

427. Senior, J.R., M.F. Johnson, D.M. DeTurck, F. Bazzoli, and E. Roda. 1990. In vivo kinetics of

radiolucent gallstones by oral dihydroxy bile acids. Gastroenterology 99:243-51.

428. Sarva, R.P., H. Fromm, S. Farivar, R.F. Sembrat, H. Mendelow, H. Shinozuka, and S.K. Wolfson.

1980. Comparison of the effects between ursodeoxycholic and chenodeoxycholic acid on liver function

and structure and on bile composition in the rhesus monkey. Gastroenterology 79:629-36.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

134

429. Schmassmann, A., A.F. Hofmann, M.A. Angellotti, H-T. Ton-Nu, C.D. Schteingart, C. Clerici, M.A.

Rothschild, B.I. Cohen, R.J. Stenger, and E.H. Mosbach. 1990. Prevention of ursodeoxycholate

hepatotoxicity in the rabbit by conjugation with N- methyl amino acids. Hepatology 11:989-996.

430. Wolpers, C., and A.F. Hofmann. 1993. Solitary versus multiple cholesterol gallbladder stones. The

Clinical Investigator 71:423-434.

431. Petroni, M.L., R.P. Jazrawi, P. Pazzi, M. Zuin, A. Lanzini, M. Fracchia, D. Facchinetti, V. Alvisi, R.

Ferraris, J.M. Bland, K.W. Heaton, M. Podda, and T.C. Northfield. 2000. Risk factors for the

development of gallstone recurrence following medical dissolution. The British-Italian Gallstone Study

Group. Eur. J. Gastroenterol. Hepatol. 12:695-700.

432. Ammon, H.V., and A.F. Hofmann. 1983. The Langenbuch paper. 1. An historical perspective and

comments of the translators. II. A translation. Gastroenterology 85:1426-33.

433. Ransohoff, D.F., and W.A. Gracie. 1990. Management of patients with symptomatic gallstones: a

quantitative analysis. 1990. Am. J. Med. 88:154-60.

434. Isselbacher, K.J. 1981. Chenodiol for gallstones: dissolution or disillusion? Ann. Intern. Med.

95:377-9.

435. Sackmann, M., J. Pauletski, T. Sauerbruch, T. Holl, G. Schelling, and G. Paumgartner. 1991. The

Munich gallbladder lithotripsy Study: results of the first five years with 711 patients. Ann. Intern. Med.

114:290-6.

436. Schoenfield, L.J., G. Berci, R.L. Carnovale, W. Casarella, P. Caslowitz, D. Chumley, R.C. Davis,

J.Y. Gillenwater, A.C. Johnson, and R.S. Jones. (and additional authors). 1990. The effect of ursodiol on

the efficacy and safety of extracorporeal shock-wave lithotripsy of gallstones. The Dornier National

Biliary Lithotripsy Study. N. Engl. J. Med. 323:1239-45.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

135

437. Thistle, J.L., G.R. May, C.E. Bender, H.J. Williams, A.J. LeRoy, P.E. Nelson, C.J. Peine, B.T.

Petersen, and J.E. McCullough. 1989. Dissolution of cholesterol gallbladder stones by methyl tert-butyl

ether administered by percutaneous transhepatic catheter. N. Engl J. Med. 320:633-9.

436. Cheslyn-Curtis, S., W.R. Lees, A.R. Hatfield, and R.C. Russell. 1992. Percutaneous techniques for

the management of symptomatic gallbladder stones. Dig. Dis. 10:208-17.

437. Holl, J., T. Sauerbruch, M. Sackmann, J. Pauletzski, and G. Paumgartner. 1991. Combined

treatment pf symptomatic gallbladder stones by extracorporeal shock-wave lithotripsy (ESWL) and

instillation of methyl tert-butyl ether. Dig. Dis. Sci. 36:1097-1101.

438. Zakko, S.F. and A.F. Hofmann. 1990. Microprocessor-assisted solvent transfer system for effective

contact dissolution of gallbladder stones. IEEE Trans. Biomed. Eng. 37:410-416.

439. Long, C.A., S.K. Teplick, M.L. Baker, and J.C. Brandon. 1991. In vitro cholesterol gallstone

dissolution: Comparison of methyl tert-butyl ether with three new ester solvents. Radiology 180:47-9.

440. Esch, O., C.D. Schteingart, D. Pappert, D. Kirby, R. Streich, and A.F. Hofmann. Increased blood

levels of methyl tert-butyl ether but not of ethyl propionate during gallbladder instillation with contact

gallstone dissolution agents in the pig. Hepatology 18:373-379, 1993.

441. Hofmann, A.F. C.D. Schteingart, K. Lyche, O. Esch, A. Amelsberg, E. VanSonnenbereg, and H.B.

D’Agostino. 1997. Topical dissolution of cholesterol gallbladder stones using ethyl propionate: initial

clinical experience. Dig. Dis. Sci. 42: 1274-1282, 1997.

442. Phillips, S., R.B. Palmer, and A. Brody. 2008. Epidemiology, toxicokinetics, and health effects of

methyltert-butyl ether (MTBE). J.Med. Toxicol. 4:115-26.

443. Esch, O. J.C. Spinosa, R.L. Hamilton, D.L. Crombie DL, C.D. Schteingart, J.F. Rondinone, H.B.

D'Agostino, J. Lillienau, and A.F. Hofmann. 1992. Acute effects on gallbladder histology of topical

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

136

methyl tert-butyl ether or ethyl propionate in animals: A comparison of two solvents for contact

dissolution of cholesterol gallstones. Hepatology 16:984-991.

444. Ponchon, T., J. Baroud, B. Pujol, P.J. Valette, and D. Perrot. 1988. Renal failure during dissolution

of gallstones by methyl-tert-butyl ether. Lancet 2:276-7.

445. Hofmann, A.F., C.D. Schteingart, E. vanSonnenberg, O. Esch, and S.F. Zakko. 1991. Contact

dissolution of cholesterol gallstones with organic solvents. Gastroenterol. Clin. N. Am. 20:183-99.

446. Clayton, P.T. 2011. Disorders of bile acid synthesis. J. Inherit. Metab. Dis. 35:521-30.

447. Mekhjian, H.S., S.F. Phillips, and A.F. Hofmann. 1971. Colonic secretion of water and electrolytes

induced by bile acids: perfusion studies in man. J. Clin. Invest. 50:1569-77.

448. Keating, N., and S.J. Keely. 2009. Bile acids in regulation of intestinal physiology. Curr.

Gastroenterol. Rep. 11:375-82.

449. Kelly, O.B., M.S. Mroz, J.B. Ward, C. Colliva, M. Scharl, R. Pellicciari, J.F. Gilmer, P.G. Fallon,

A.F. Hofmann, A. Roda, F.E. Murray, and S.J. Keely. 2013. Ursodeoxycholic acid attenuates colonic

epithelial secretory function. J. Physiol. 591:2307-18.

450. Ao, M., J. Sarathy, J. Dominique, W.A. Airefai, and M.C. Rao. 2013. Chenodeoxycholic acid

stimulates Cl(-) secretion via cAMP signaling and increases cystic fibrosis transmembrane conductance

regulator phosphorylation in T84 cells. Am. J. Physiol. Cell Physiol. 305:C447-56.

451. Hofmann, A.F. and J.R. Poley. 1969. Cholestyramine treatment of diarrhea associated with ileal

resection. N. Engl..J. Med. 281:397-402.

452. Walters, J.R., and S.S. Pattni. 2010. Managing bile acid diarrhea. Therap. Adv. Gastoenterol.

3:349-57.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

137

453. Walters, J.R., A.M. Tasleern, O.S. Omer, W.G. Brydon, T. Dew, and C.W. Le Roux. 2009. A new

mechanism for bile salt diarrhea: defective feedback inhibition of bile acid biosynthesis. Clin.

Gastroenterol. Hepatol. 7:1189-94.

454. Wedlake, L., R. A’Hern, D. Russell, K. Thomas, and J.R. Walters. 2009. Systematic review: the

prevalence of idiopathic bile acid malabsorption as diagnosed by SeHCAT scanning in patients with

diarrhea predominant irritable bowel syndrome. Aliment. Pharmacol. Ther. 30:707-17.

455. Axelson, M, A. Aly, and J. Sjövall. 1988. Levels of 7 alpha-hydroxy-4-choleten-3-one in plasma

reflect rates of bile acid synthesis in man. FEBS Lett. 239:324-8.

456. Sauter, G., F. Berr, U. Beuers, S. Fischer, and G. Paumgartner. 1996. Serum concentrations of

7alpha-hydroxy-4-cholesten-3-one reflect bile acid synthesis in humans. Hepatology 24:123-6.

457. Vijayvargiya, P., M. Camilleri, A. Shin, and A. Saenger. 2013. Methods for diagnosis of bile acid

malabsorption in clinical practice. Clin. Gastroenterol. Hepatol. 11:1232-9.

458. Shin, A., M. Camilleri, P. Vijayvargiya, I. Busciglio, D. Burton, M. Ryks, D. Rhoten, A. Lueke, A.

Saenger, A. Girtman, and A.R. Zinsmeister. 2013. Bowel functions, fecal unconjugated primary and

secondary bile acids and colonic transit in patients with irritable bowel syndrome. Clin. Gastroenterol.

Hepatol. 11:1270-5.

459. Hofmann, A.F. V. Loening-Baucke, J.E. Lavine, L.R. Hagey, J.H. Steinbach, C.A. Packard, T.L.

Griffin, and D.A. Chatfield. 2008. Altered Bile Acid Metabolism in Childhood Functional Constipation:

Inactivation of Secretory Bile Acids by Sulfation in a Subset of Patients. J. Ped. Gastroenterol. Nutr.

47:598-606.

460. Higuchi, H., and G.J. Gores. 2003. Bile acid regulation of hepatic physiology. IV. Bile acids and

death receptors. Am. J. Physiol. Gastrointest. Liver Physiol. 284:G734-8.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

138

461. Sokol, R.J., M.S. Straka, R. Dahl, M.W. Devereaux, and B. Yerushalmi. 2001. Role of oxidant

stress in the permeability transition induced in rat hepatic mitochondria by hydrophobic bile acids.

Pediatr. Res. 49:519-31.

462. Amaral, J.D., R.J. Viana, R.M. Ramalho, C.J. Steer, and C.M. Rodrigues. 2009. Bile acids:

regulation of apoptosis by ursodeoxycholic acid. J. Lipid Res. 50:1721-34.

463. Allen, K., H. Jaeschke, and B.L. Kopple. 2010. Bile acids induce inflammatory genes in

hepatocytes: a novel mechanism of inflammation during obstructive cholestasis. Am. J. Pathol. 178:175-

86.

464. Gustafsson, B.E., J.A. Gustafsson, and J. Sjövall. 1966. Intestinal and fecal sterols in germfree and

conventional rats. Acta Chem. Scand. 20:1827-35.

465. Devkota, S., Y. Wang, M.W. Musch, V. Leone, H. Fehlner-Peach, A. Nadimpalli, D.A.

Antonopoulos, B. Jabri, and E.B. Chang. 2012. Dietary-fat-induced taurocholic acid promotes pathobiont

expansion and colitis in IL10-/- mice. Nature 487:104-8.

466. Kamen, M.D. 1963. The early history of carbon-14. J. Chem. Educ. 40:234-42.

467. Lindor, K.D. 2011. Farnesoid X receptor agonists for primary biliary cirrhosis. Curr. Opin.

Gastroenterol. 27:285-8.

468. Adorini, L., M. Pruzanski, and D. Shapiro. 2012. Farnesoid X receptor targeting to treat

nonalcoholic steatohepatitis. Drug. Discov. Today 17:988-97.

469. Fickert, P, M.J. Pollheimer, D. Silbert, T. Moustafa, E. Halilbasic, E. Krones, F. Durchschein, A.

Thϋringer, G. Zollner, H. Denk, and M. Trauner. 2013. Differential effects of norUDCA and UDCA in

obstructive cholestasis in mice. J. Hepatol. 58:1201-8.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

139

470. Huff, M.W., D.E. Telford, J.Y. Edwards, J.R. Burnett, P.H. Barrett, S.R. Rapp, N. Napawan, and

B.T. Keller. 2002. Inhibition of the apical sodium-dependent bile acid transporter reduces LDL

cholesterol and apoB by enhanced plasma clearance of LDL apoB. Arterioscler. Thromb. Vsc. Biol.

22:1884-91.

471. Chen, L, , X. Yao, A. Young, J. McNulty, D. Anderson, Y. Liu, C. Nystrom, D. Croom, S. Ross, J.

Collins, D. Rajpal, K. Hamlet, C. Smith, and B. Gedulin . 2012. Inhibition of apical sodium-dependent

bile acid transporter as a novel treatment for diabetes. Am. J. Physiol. Endocrinol. Metab. 302:E68-76.

472. M. Simren, A. Bajor, P.G. Gillberg, M. Rudling, and H. Abrahamsson. 2011. Randomised clinical

trial: the ileal bile acid transporter inhibitor A3309 vs. placebo in patients with chronic idiopathic

constipation – a double blind study. Aliment. Pharmacol. Ther. 34:41-50.

473. Hagey, L.R., N. Vidal, A.F. Hofmann AF, and M.D. Krasowski. 2010. Complex Evolution of Bile

Salts in Birds. The Auk. 127:820-31.

474. Lee, S.P., R. Lester, and J.S. Pyrek. 1987. Vulpecholic acid (1 alpha, 3 alpha, 7 alpha-trihydroxy-5-

beta-cholan-24-oic acid): a novel bile acid of a marsupial, Trichosurus vulpecula (Lesson). J. Lipid Res.

28:19-31.

475. Hagey, L.R., C.D. Schteingart, H-T. Ton-Nu, and A.F. Hofmann. 1994. Biliary bile acids of fruit

pigeons and doves (columbiformes): presence of 1β-hydroxychenodeoxycholic acid and conjugation with

glycine as well as taurine. J. Lipid Res. 35:2041-2048.

476. Murai, T., R. Mahara, T. Kurosawa, A. Kimura, and M. Tohma. 1997. Determination of fetal bile

acids in biological fluids from neonates by gas chromatography-negative ion chemical ionization mass

spectrometry. J. Chromatogr. B Biomed. Sci. Appl. 691:13-22

477. Bi D., X.Y. Chai, Y.L. Song, Y. Lei and P.F. Tu. 2009. Novel bile acids frombear bile power and

bile of geese. Chem. Pharm. Bull. (Tokyo). 57:528-31.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

140

478. Kakiyama, G., T. Iida, T. Goto, N. Mano, J. Goto, T. Nambara, L.R. Hagey, and A.F. Hofmann.

2006. Identification of a novel bile acid in swans, tree ducks, and geese: 3α,7α,15α-trihydroxy-5β-

cholan-24-oic acid. J. Lipid Res. 47:1551-8.

479. Kurosawa, T., Y. Nomura, R. Mahara, T. Yoshimura, A. Kimura, S. Ikegawa, and M. Tohma. 1995.

Synthesis of 19-hydroxylated bile acids and identification of 3 alpha, 7 alpha, 12 alpha, 19-tetrahydroxy-

5-beta-cholan-24-oic acid in human neonatal urine. Chem. Pharm. Bull. (Tokyo). 43:1551-7.

480. Schiff, L., J.B. Carey, and J. Dietschy. 1969. Bile salt metabolism. Thomas, Springfield,IL.

481. Schteingart, C.D., L.R. Hagey, K.D.R. Setchell, and A.F. Hofmann. 1993. 5β-Hydroxylation by the

liver: Identification of 3,5,7-trihydroxy nor-bile acids as new major biotrans-formation products of 3,7-

dihydroxy nor-bile acids in rodents. J. Biol. Chem. 268:11239-11246.

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

141

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

142

LEGENDS TO FIGURES

Figure 1. Participants in the first symposium in the United States dealing with bile acid metabolism. The

meeting was organized by Leon Schiff, a clinical hepatologist and took place in September, 1967. First

row, left to right. M. Shiner, L. Schiff (organizer), G. Haslewood, M. Siperstein, S. Tabaqchali, D. Small.

Second row, left to right. L. Lack, L. Schoenfield, P. Nair, J. Carey, A. Hofmann. Third row, left to right.

M. Stanley, S. Hsia, unidentified, N. Javitt, H. Danielsson, J. Dietschy, S. Emerman. Fourth row, left to

right. F. Kern, J. Sjövall, B. Drasar, R. Palmer, J. Senior, N. Myant, J. Wilson, E. Staple. Proceedings of

the meeting were published in 1969 (480).

Figure 2. Numbering system of bile acids and bile alcohols, and frontal view of CDCA. The numbering

system was developed in the 1930’s when the steroid structure was finally established.

Figure 3. Molecular structure of cholesterol (above) and CDCA (below) showing the changes in the

cholesterol molecule when a C24 bile acid is formed. The changes are (1) reduction of the double bond to

give a 5β (A/B cis) A/B ring juncture (2) a modified β- oxidation of the side chain to remove three carbon

atoms and convert the terminal carbon to a carboxyl group; and 3) epimerization of the 3β- hydroxy group

to a 3α- hydroxy group. For a discussion of the enzymes involved, see reference 155.

Figure 4. Depictions of the two epimers of decalin, a saturated C10 hydrocarbon that is used to illustrate

the two A/B ring juncture epimers of bile acids. When the two bridgehead hydrogen atoms are across

from each other, the juncture is called trans; when they are on the same side of the ring juncture, the

juncture is called cis. When bile acid structure is shown in frontal view (Figure 2), the only indication of

the stereochemistry of the A/B ring juncture is the orientation of the hydrogen atom, i.e. whether alpha or

beta. However, in bile acids, the substituent at the upper bridgehead carbon atom is the C-19 methyl

group. Often bile acid structure is shown without any indication as to the orientation of the hydrogen

atom at C-5. When this is done, it is understood that the molecule is A/B cis, as 5β- (A/B cis) bile acids

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

143

are much more common in nature than 5α (A/B trans) bile acids. A/B trans bile acids are termed “allo”

bile acids.

Figure 5. Silhouette of a C24 conjugated bile acid molecule emphasizing its possession of a hydrophobic

side (beta face) and a hydrophilic side (alpha face). Common sites of hydroxylation in addition to the

default structure (3α, 7α) are at C-6, C-12, and C-16; all of these are on the hydrophilic side of the

molecule. Modified from Roda et al (360).

Figure 6. Time course in healthy subjects of levels in peripheral venous plasma of immunoreactive

conjugates of cholic acid (cholyl conjugates) and those of CDCA (here termed “chenyl” conjugates) in

response to meals; meals are indicated by the thick, vertical arrows. Levels of CDCA conjugates rise

sooner than those of cholyl conjugates indicating more proximal absorption. Levels of CDCA conjugates

are also higher than those of cholyl conjugates despite similar proportions in bile because of lower

fractional extraction by the liver. From reference 216.

Figure 7. Schematic depiction of the work of L. Lack and I. Weiner on conjugated bile acid (here,

taurocholate) absorption by everted sacs of rat intestine, showing preferential and active absorption by the

distal ileum. At time zero, bile acid concentrations are identical on both sides of the intestine. Modified

from reference 229.

Figure 8. Depiction of cholesterol and bile acid metabolism as well as the enterohepatic circulation by

Bergström, Danielsson, and Samuelsson in 1959 (8). In the accompanying text, they note that the mass of

cholesterol entering the small intestine in bile is greater than that of dietary cholesterol. The figure shows

neither the spillover of absorbed bile acids into the systemic circulation nor selective absorption of

conjugated bile acids by the terminal ileum. Values for biliary bile acid secretion and fecal bile acid

excretion are about two fold higher than those obtained in more recent studies.

Figure 9. Depiction of the use of triangular coordinates to plot biliary lipid composition and thereby to

define the saturation of cholesterol in bile. The triangle is derived from a plane parallel to the base

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

144

indicating a constant water composition (10% solids, 90% water) in the tetrahedron consisting of the four

biliary components (water, lecithin, cholesterol, and bile acids). The micellar zone is shown in black.

Bile having a lipid composition falling within in the micellar zone is unsaturated. If the composition is

above the upper limit of the micellar zone, the sample is supersaturated in cholesterol and at risk for

cholesterol gallstone formation (376). If biliary lipids are known, the per cent saturation can also be

obtained from tables published by Carey (393,394).

Figure 10. Molecular arrangement of the drum shaped mixed micelle micelle originally proposed by

Small (369) and modified by Carey (330), as compared to the radial shell model proposed by Nichols and

Ozarowski (371), The radial shell model was confirmed by Hjelm et al using small angle neutron

scattering (372,403,404) and also by molecular dynamic studies of Marrink and Mark (373).

Figure 11. Phase diagram of the water-monoolein (monooleylglycerol)-taurocholate system, showing the

large micellar area (cross-hatched), thus depicting the excellent solubilizing potency of bile acids for

polar lipids. Liquid crystal states are indicated by vertical and horizontal cross hatching. Modified from

Svärd et al. (398).

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

145

Table. Common and uncommon sites of additional nuclear hydroxylation (in addition to the default

structure of the 3α,7α-dihydroxy compound) in primary C24 bile acids, C27 bile acids, and C27 bile alcohols

+ Hydroxylation occurs at C-5 with 24-nor (C23) UDCA in hamsters (481).

*The bile acid described is a tetrahydroxy bile acid (3α,7α,12α,19-tetrahydroxy-)

Class Common Uncommon+

Site Natural occurrence References

C24 bile acids 6α,6β,12α,16α 1α Australian opossum 474 1β Pigeons, neonates 475, 476 2β Neonates 477 4β Chinese pheasant 473 9α Bear 478 11α Sunfish 472 15α Swan, goose 478,479 19* Neonates 480 C27 bile acids 12α 1α Tinamou 125 16α Ancient birds 473 C27 bile alcohols 12α 2α Arapaima 126 6α and 6β Elephants, hyrax, manatee 127 11α Sunfish 473 by guest, on F

ebruary 12, 2018w

ww

.jlr.orgD

ownloaded from

Figure 1

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 2

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 3

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 4

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 5

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 6

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 7

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 8

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 9

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 10

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from

Figure 11

by guest, on February 12, 2018

ww

w.jlr.org

Dow

nloaded from


Top Related