Sei sulla pagina 1di 43

thematic review

Thematic Review Series: Living History of Lipids

Key discoveries in bile acid chemistry and biology and


their clinical applications: history of the last
eight decades
Alan F. Hofmann1 and Lee R. Hagey1
Department of Medicine, University of California, San Diego, San Diego, CA

Abstract During the last 80 years there have been extraor- conceptions are rare, and provocative judgments stand
dinary advances in our knowledge of the chemistry and biol- the test of time. Let us also hope that this effort will be
ogy of bile acids. We present here a brief history of the both useful and entertaining. Each of the topics discussed
major achievements as we perceive them. Bernal, a physi- merits at least a full length article, so there will be, of ne-
cist, determined the X-ray structure of cholesterol crystals,
cessity in many instances, consideration of only what we
and his data together with the vast chemical studies of
Wieland and Windaus enabled the correct structure of the perceive to be the highlights.
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 conju- THE EBB AND FLOW OF MEDICAL INTEREST IN
gation are summarized. Bile acid measurement encom- BILE ACIDS
passes the techniques of GC, HPLC, and MS, as well as
enzymatic, bioluminescent, and competitive binding meth- After the elucidation of the true chemical structure of
ods. The enterohepatic circulation of bile acids results from bile acids in 1932 (see below), there was little interest in
vectorial transport of bile acids by the ileal enterocyte and bile acids in the Western world. One exception to this
hepatocyte; the key transporters have been cloned. Bile ac- statement was the laboratory of Siegfried Thannhauser,
ids are amphipathic, self-associate in solution, and form
mixed micelles with polar lipids, phosphatidylcholine in who wrote the first textbook of metabolic biochemistry in
bile, and fatty acids in intestinal content during triglyceride Germany. He studied cholesterol and bile acid balance in
digestion. The rise and decline of dissolution of cholesterol the biliary fistula dog (1). During this time, bile acids were
gallstones by the ingestion of 3,7-dihydroxy bile acids is sold as liver tonics and laxatives, but there were no pla-
chronicled. Scientists from throughout the world have cebo-controlled studies showing efficacy. Indeed, bile ac-
contributed to these achievements.—Hofmann, A. F., and L. ids were considered by the medical profession to have no
R. Hagey. Key discoveries in bile acid chemistry and biology useful therapeutic properties. The tri-oxo derivative of
and their clinical applications: history of the last eight de- cholic acid (called “dehydrocholic acid”) was known to in-
cades. J. Lipid Res. 2014. 55: 1553–1595.
duce bile flow in animals (2), and was occasionally used to
stimulate bile flow in patients; but again there were no con-
Supplementary key words bile acid transport • enterohepatic circula-
tion • bile acid physical chemistry • bile acid analysis • bile acid
trolled studies showing efficacy in hepatobiliary disease.
metabolism

Abbreviations: ASBT, apical sodium-codependent bile acid trans-


This article contains aspects of the history of bile acid porter; AUC, area under the curve; BSEP, bile salt export pump; CDCA,
research during the last eight decades. The authors, now chenodeoxycholic acid; CMC, critical micellization concentration;
in the twilight of their careers, are not trained historians, CMT, critical micellization temperature; CMpH, critical micellization
pH; DCA, deoxycholic acid; FDA, Food and Drug Administration; FGF,
and there will be omissions, misconceptions, and provoca- fibroblast growth factor; FXR, farnesoid X receptor; IUPAC, Interna-
tive judgments. Let us hope the omissions are minor, mis- tional Union of Pure and Applied Chemistry; MDR, multidrug resis-
tance (protein); MRP, multidrug resistant protein; MTBE, methyl
tert-butyl ether; NCGS, National Cooperative Gallstone Study; NIH,
National Institutes of Health; NTCP, Na+ taurocholate cotransporting
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, Ger- polypeptide; OATP, organic anion transporting polypeptide; OST, or-
many. The authors acknowledge a grant-in-aid from Intercept Pharma, Inc. ganic solute transporter; PC, phosphatidylcholine; TGR5, transmem-
brane G protein-coupled receptor 5; UDCA, ursodeoxycholic acid.
Manuscript received 24 March 2014 and in revised form 2 May 2014. 1
To whom correspondence should be addressed.
Published, JLR Papers in Press, May 17, 2014 e-mail: AHofmann@ucsd.edu (A.F.H.); LHagey@ucsd.edu
DOI 10.1194/jlr.R049437 (L.R.H.)

Copyright © 2014 by the American Society for Biochemistry and Molecular Biology, Inc.

This article is available online at http://www.jlr.org Journal of Lipid Research Volume 55, 2014 1553
Albert Boehringer founded a pharmaceutical company said that Bergström chose his graduate students on the
in Ingelheim, Germany. His wife was a cousin of Heinrich golf course. It is also said that the lights never went off at
Wieland, then a professor in Munich. Wieland was success- night in the Bergström laboratory.)
ful in persuading the company to begin the isolation and Sjövall developed GC and then later, after he had moved
marketing of cholic acid from ox bile in 1917. Thus, cholic to the Karolinska Institute in Stockholm, developed MS
acid of high purity soon became available to the scientific for the measurement of bile acids (9). He used these tech-
community. During this time, Japanese students came to niques and others to define important aspects of bile acid
laboratories such as that of Wieland to pursue their doc- metabolism over the next four decades (10). Norman de-
toral studies. When they returned to Japan, they contin- veloped a simple synthesis of conjugated bile acids, pre-
ued their studies on bile acid and bile alcohol structure. pared the conjugates of all the major bile acids known at
Their work, as well as that occurring in Europe is summa- that time (11), and with Sjövall performed metabolic stud-
rized in the book Ueber die Chemie and Physiologie der Gallen- ies in animals which distinguished primary bile acids
säuren authored by Shimizu in 1935 (3). (made in the liver from cholesterol) from secondary bile
During the 1930s and 1940s, an enormous international acids (made from primary bile acids by intestinal bacte-
effort went into defining the structure of the major hor- ria). Their work proved that DCA was a secondary bile acid
monal steroids. When it was recognized that cortisone had (12, 13). Lindstedt measured bile acid kinetics (pool size,
a C-11 oxygen atom, it was logical to use deoxycholic acid turnover rate, and synthesis) in man (14). Danielsson
(DCA) (possessing a C-12 hydroxyl group) as a chemical identified key intermediates in bile acid biosynthesis (15).
precursor for the synthesis of corticosteroids. DCA was eas- Samuelsson synthesized ursodeoxycholic acid (UDCA)
ily isolated from bovine bile or synthesized from cholic and defined its metabolism (16). Eriksson showed that
acid, and it was not too difficult for a talented chemist to bile acid synthesis in the rat was under negative feedback
move the oxygen from C-12 to C-11. In 1946, H. Sarett (at control (17). Borgström, with his colleagues Sjövall,
the Merck Company) reported a complex synthesis (37 Lundh, and Dahlquist, intubated healthy volunteers and
steps!) of cortisone from DCA (4), and this led to its com- defined the site at which individual nutrients are absorbed
mercial production on a small scale. Two years later, (18). This study proposed that the bile acid pool circulates
Hench, a rheumatologist at the Mayo Clinic, who worked about twice per meal. Borgström became director of the
closely with his colleague Kendall, an able steroid chemist, Department of Physiological Chemistry in Lund in 1958,
obtained a small supply of cortisone supplied by Merck replacing Bergström who had moved earlier that year to
(5). Hench showed that this compound caused a strik- the Karolinska Institute in Stockholm with the bile acid
ing symptomatic improvement in patients with rheuma- group. Bergström and his pupil Samuelsson then left the
toid arthritis. The translational research of Hench and bile acid field and turned their attention to prostaglan-
Kendall resulted in their being awarded the Nobel Prize dins. Their outstanding work in this field led to their re-
in 1950 (6). ceiving the Nobel Prize in 1982. However, at the Karolinska
As a result of the truly exciting advent of cortisone in Hospital, Swedish scientists continued to work on bile ac-
the treatment of rheumatoid arthritis, there was an enor- ids, clinical and biochemical studies being conducted by
mous effort to develop a simple, efficient synthesis of this Kurt Einarsson, Bo Angelin, Ingemar Björkhem, and Kjell
hormone from DCA. Soon, there was concern that the Wikvall, among others.
world’s supply of DCA (derived from cow and sheep bile) In 1961, news of the work of Lack and Weiner (19),
would be insufficient to meet the medical demand. Then, which showed unequivocally that the ileum was the site of
workers at Upjohn discovered that hydroxylation at C-11 conjugated bile acid absorption, reached Sweden. Borg-
could be achieved using a fungus. Other workers found a ström directed the senior author, to perform studies show-
plant saponin that could be used as a substrate for the fun- ing that in man, conjugated bile acids are absorbed mainly
gal hydroxylation at C-11, after which the side chain was in the ileum (20).
easily altered to that of cortisone [for details, see Fieser In London, Geoffrey Haslewood pursued the structure
and Fieser (7)]. DCA was no longer needed, and chemical of bile acids in different species, based on his belief that
interest in bile acids collapsed. bile acid composition was a powerful phenotype that
After the Second World War, a few laboratories pursued would aid in the identification of evolutionary relation-
the search for new bile acids, as well as defining the me- ships (21). Edward Doisy, chairman of the Department of
tabolism of bile acids in mammals. The availability of 14C Biochemistry at St. Louis University, having already won
and 3H which could be incorporated into the bile acid the Nobel prize for his work on vitamin K, pursued the
molecule, together with the development of automatic liq- identity of the 3,6,7-trihydroxy bile acids in rats and identi-
uid scintillation counters and chromatography enabled fied the ␣-, ␤-, and ␻-muricholic acids (22). His student,
biotransformations to be identified and measured. Sune William Elliott synthesized numerous bile acids and de-
Bergström, then working in Lund, Sweden, recruited a scribed their chemical and chromatographic properties
highly talented group of doctoral students, Jan Sjövall, (23).
Henry Danielsson, Arne Norman, Sven Lindstedt, Bengt In Japan, a school of bile acid studies was established by
Samuelsson, Sven Eriksson, Bengt Borgström, among oth- Tayei Shimizu (24), who returned to his native country af-
ers, who carried out fundamental studies of bile acid me- ter having spent three years in the laboratory of Heinrich
tabolism in a variety of species including man (8). (It is Wieland. Japanese investigators pursued the structure of

1554 Journal of Lipid Research Volume 55, 2014


natural bile acids, the work continuing with Taro Kazuno In 1965, the senior author, working in the laboratory
and his pupil Takahiko Hoshita (25) and, in turn, with of E. H. Ahrens, began feeding studies with cholic acid in
Hoshita’s student, Mizuho Une (25, 26). Japanese workers a patient with severe hypercholesterolemia, and showed
also attempted to define the early steps in bile acid biosyn- that cholic acid feeding was a potent suppressor of bile
thesis from cholesterol (27). acid and cholesterol biosynthesis (34), based on measure-
In the United States, James Carey, working at the Uni- ment of fecal bile acids and sterols, using the newly devel-
versity of Minnesota, was directed by his mentor, Cecil oped gas chromatographic method for fecal bile acids that
Watson, to pursue bile acid metabolism in man. He identi- had been developed in this laboratory (32). It was logical
fied chenodeoxycholic acid (CDCA) as a major biliary bile to test CDCA, the other primary bile acid, but at that time,
acid (28) and proposed that lithocholic acid, its bacte- the world’s supply of pure CDCA was thought to be less
rial metabolite, caused liver injury in man (29). Edward than 10 g, and the synthesis from cholic acid was difficult.
(“Pete”) Ahrens, working with Lyman Craig at the Rocke- However, in the 1960s, a small English pharmaceutical
feller University, showed that countercurrent distribution company (Weddell Pharmaceuticals) began the manufac-
could be used to separate conjugated and unconjugated ture of CDCA for unknown reasons. A kilogram was pur-
bile acids (30). Later, two of his postdoctoral fellows, Scott chased for the senior author by the Mayo Clinic in 1967.
Grundy and Tatu Miettinen, developed a gas chromato- Leslie Schoenfield returned to the Mayo Clinic in 1966
graphic method for measurement of fecal bile acids, after having spent a year in the laboratory of Sjövall, and
whose output, in the steady state, is equivalent to bile acid initiated a clinical trial with his fellow, Johnson Thistle, to
synthesis (31). Grundy went on to a distinguished career in test whether oral cholic acid or hyodeoxycholic acid would
human nutrition (32). Miettinen continued to do sterol bal- lower cholesterol in bile and ultimately induce cholesterol
ance studies for much of his very productive career (33). gallstone dissolution. The senior author persuaded
The first symposium devoted solely to bile acids was or- Schoenfield to add CDCA to his protocol, and this study of
ganized by Leon Schiff, a clinical hepatologist, who was
Thistle and Schoenfield showed that CDCA feeding de-
one of the founders of the American Association for the
creased biliary cholesterol saturation, whereas neither
Study of Liver Diseases. This symposium, held in 1967, was
cholic acid nor hyodeoxycholic acid had any effect (35).
quite exciting for its participants who are shown in Fig. 1.
In 1972, the first gallstone dissolution induced by the in-
However, it is safe to say that the study of bile acids was
gestion of CDCA was observed, initially at the Mayo Clinic
pursued by only a small number of laboratories, some in
(36), and later in London by a group led by Hermon
Departments of Biochemistry and some in Departments of
Dowling (37). [This was not the first time that the efficacy
Medicine. Erwin Mosbach, one of the early workers in bile
of oral bile acids had been tested at the Mayo Clinic. In
acid metabolism, once stated to his wife, “Whenever I go
to the podium to give a paper on bile acids, everyone 1938, Philip Hench had fed a mixture of conjugated bile
leaves the room”. salts in an unsuccessful attempt to treat rheumatoid arthri-
tis (38).]
The discovery that CDCA would induce gradual dissolu-
tion 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.
In Japan, UDCA had been brought to market in the
1950s by Tokyo Tanabe as a liver tonic, based on the leg-
endary 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 hepatotox-
icity. To provide UDCA for therapeutic trials performed
Fig. 1. Participants in the first symposium in the United States around the world, UDCA was synthesized in kilogram
dealing with bile acid metabolism. The meeting was organized by
Leon Schiff, a clinical hepatologist and took place in September quantities and became the fourth bile acid to become
1967. First row, left to right: M. Shiner, L. Schiff (organizer), G. available as a fine chemical. A Japanese perspective on the
Haslewood, M. Siperstein, S. Tabaqchali, D. Small. Second row, left discovery of UDCA as a therapeutic agent is available (41).
to right: L. Lack, L. Schoenfield, P. Nair, J. Carey, A. Hofmann. As discussed later, dissolution of cholesterol gallstones
Third row, left to right: M. Stanley, S. Hsia, unidentified, N. Javitt, by ingestion of UDCA began to decline in the 1990s be-
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,
cause of the development of laparoscopic cholecystectomy,
E. Staple. Proceedings of the meeting were published in 1969 which promised a rapid cure for the problems of gallstone
(480). disease, irrespective of gallstone type. Once again, bile

History of bile acid research 1555


acids seemed to have little medical value, and interest in activities (59). Several potent FXR agonists have been syn-
their biology and chemistry declined. thesized (53, 60), and one of these, obeticholic acid (61),
Nonetheless, the ready availability of UDCA meant that is now being tested for its efficacy in primary biliary cirrho-
it continued to be used for gallstone patients who were sis and nonalcoholic steatohepatitis (62).
poor operative candidates. Ulrich Leuschner, a hepatolo- Two Japanese groups discovered transmembrane G-
gist based in Frankfurt, Germany, noted that UDCA in- coupled protein receptor 5 (TGR5) activated by bile acids
gestion improved biochemical parameters in patients with (63, 64). TGR5 has a wide tissue distribution, being most
primary biliary cirrhosis (42). UDCA was subsequently heavily expressed in the enteric nervous system (65); it is
shown to slow disease progression in primary biliary cir- also present in spinal cord neurons (66) and the primary
rhosis, initially by Raoul Poupon in Paris (43), and later by cilia of cholangiocytes (67). The consequences of this
Keith Lindor and his colleagues at the Mayo Clinic (44). activation include release of the hormone glucagon-like
UDCA treatment of primary biliary cirrhosis is now stan- peptide 1 (GLP-1), which has appetite-suppressing and an-
dard of care. UDCA was also shown to be effective in cho- tidiabetic effects (68). TGR5 also has effects on hepatic
lestasis of pregnancy (45). UDCA has been widely used blood flow (69), distal intestinal motility (70), and in addi-
for other cholestatic diseases and to restore bile flow fol- tion, has anti-inflammatory effects (71) that are under ac-
lowing liver transplantation, but proof of efficacy in these tive investigation.
conditions based on placebo-controlled trials has been Discovery of the signaling properties of bile acids is cer-
uncommon. tainly a new paradigm in the bile acid field. Bile acids have
At present, it is estimated that about 1,000 metric tons, become hormones. Now highly talented molecular biolo-
or about 1,000,000 kg, of cholic acid are produced globally gists in both academe and the biotechnology industry are
from bovine bile, the majority of which is used for the pro- pursuing bile acid biology. Bile acid signaling has been
duction of UDCA (S. Yorke, New Zealand Pharmaceuti- discussed only briefly in this review, as its aim is to focus on
cals, Ltd.; personal observation). Bile acids are also used as events in the past.
precursors for the synthesis of nuclear receptor agonists As bile acids have been the subject of scientific study
such as obeticholic acid (the 6␣-ethyl derivative of CDCA), for more than 150 years, the bile acid literature is vast.
as building blocks for other molecules (46), and as key The early history of bile acid discovery is available in the
constituents of some bacterial media. Bile acids have even two monographs by Harry Sobotka (72, 73), as well as
been used in the electronics industry as precursors of pho- the masterly text on steroids by the Fiesers published
toresist molecules in integrated circuits (47). (This un- in 1955 (7). A small pharmaceutical company, Dr. Falk
likely technology transfer from a biological laboratory to Pharma, based in Freiburg, Germany, was the first Euro-
the electronic world resulted from one of the early chem- pean company to bring CDCA to market for gallstone
ists in our laboratory, Rick DiPietro, taking a position in dissolution. Since 1972, the Falk Foundation, funded by
the electronics industry and envisioning a truly novel use Dr. Falk Pharma, has hosted biennial meetings on bile
for bile acids.) acid biology, chemistry, and therapeutics. The proceed-
In 1984, the results of the Coronary Primary Prevention ings of these meetings, usually published in book form,
Trial showed that the ingestion of cholestyramine, the first serve to document the advances that have occurred in
bile acid sequestrant to be marketed, lowered plasma cho- the last four decades. G. A. D. Haslewood’s slim mono-
lesterol levels and reduced cardiovascular events. This was graph summarizes current knowledge in 1966 (74) and
of considerable interest to the cardiovascular community, Haslewood later authored a monograph on the biologi-
as the study provided the first convincing evidence for the cal importance of bile salts (75). Elsevier’s Encyclopedia of
validity of the cholesterol hypothesis (48). Nonetheless, Organic Chemistry (76) contains detailed information
the demonstrated efficacy of treating hypercholesterol- on the chemistry of bile acids and their derivatives. A
emia with a bile acid sequestrant had a rather modest ef- four volume series, The Bile Acids, edited by Nair and
fect on clinical practice because of the poor tolerability of Kritchevsky (77), treats many of the topics discussed in
cholestyramine and the emergence of the statins, which this article in much greater detail. A book entitled Ste-
were still more potent and usually well tolerated. rols and Bile Acids is volume 12 of the series, New Compre-
The current rebirth of interest in bile acids arises from hensive Biochemistry. This volume, published in 1985, has
the astonishing discovery of their signaling properties. six chapters dealing with varying aspects of bile acids
Bile acids were found to be the ligand for the orphan nu- and bile alcohols (78). A multiauthor book describing
clear receptor farnesoid X receptor (FXR) (49–51), mean- many aspects of bile acid research with particular em-
ing that bile acids regulate the activity of many genes phasis on Italian contributions was published in 2000
(52–54). Bile acid derivatives that are potent FXR agonists (79). The senior author (80) and Hermon Dowling (81)
have been shown to induce the synthesis and basolateral have both written overviews of the field from a medical
release of the protein fibroblast growth factor (FGF)15/19 standpoint in 1985. The Journal of Lipid Research pub-
from the ileal enterocyte (55). FGF19 travels in portal ve- lished a number of excellent reviews on the biology and
nous blood to the liver and downregulates bile acid biosyn- chemistry of bile acids a few years ago.
thesis (55, 56). FGF19 promotes hepatocyte growth (57), The remainder of this historical review will discuss se-
and FXR agonism also has antidiabetic effects mediated by lected topics in more detail with an emphasis on struc-
multiple mechanisms (58), as well as anti-inflammatory ture-activity relationships. The review will not summarize

1556 Journal of Lipid Research Volume 55, 2014


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.

BILE ACID STRUCTURE: HISTORY AND CURRENT


CONCEPTS

The carbon skeleton


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 fol-
lowing 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 labo-
rious attempts to define the structure of bile acids and pre-
sented his current vision of the structure of cholic acid
(83). Windaus presented his proposed structure of choles-
terol as well as his brilliant studies on the formation of Fig. 2. Numbering system of bile acids and bile alcohols, and
vitamin D2 from cholesterol by radiation (84). These two frontal view of CDCA. The numbering system was developed in the
extraordinarily talented chemists, using the most primitive 1930s when the steroid structure was finally established.
of chemical techniques, had slowly developed the idea
that both cholesterol and bile acids share a four ring struc- being the helical structure of DNA and RNA by Watson
ture with a side chain of five carbon atoms for bile acids and Crick. Bernal’s left wing political views and his Bohe-
and eight carbons for cholesterol. However, the structures mian life style probably kept him from being awarded a
of the steroid nucleus shown in their Nobel lectures were Nobel Prize (89, 90).
not correct, and each of these giants described their struc-
tures as “probable”. The side chain. The structure of the C5 side chain of the
The path to the correct structure came from an unlikely common natural C24 bile acids, isopentanoic acid or more
origin. The polymath Desmond Bernal, working in the specifically ␥-methylbutanoic acid, was elucidated by
Mineralogical Museum in Cambridge, England decided, Wieland, and like the structure of the steroid nucleus, has
together with his colleague William Astbury, to undertake also been confirmed by X-ray diffraction (87) and total
X-ray diffraction studies of organic molecules. Bernal, synthesis (88).
nominally a physicist, began his studies in 1931. He stud- The structure of the C8 side chain in C27 bile alcohols
ied ergosterol, cholesterol, and vitamin D, and for all three and bile acids was more problematic. It was not clear ini-
of these compounds obtained a probable structure of a tially that these molecules possessed the side chain of cho-
flat molecule. He published his findings as a note in Nature lesterol because it was quite uncertain whether these
in 1932 (85). This report caught the attention of Rosen- compounds and even the common natural C24 bile acids
heim and King in London, and Wieland and Dane in Mu- were derived from cholesterol. The uncertainty arose from
nich. Later, in that same year, Wieland and Dane followed the failure of a dietary cholesterol load to increase bile
by Rosenheim and King published the application of Ber- acid output in the biliary fistula dog. George Whipple,
nal’s pioneering studies to the extensive literature on bile a leading physiologist, wrote in his 1922 review on the
acid structure (86). Each group proposed the cyclopen- constituents of bile (91): “Cholesterol … has often been
tanoperhydrophenanthrene structure for the steroid nu- suspected as the precursor of cholic acid, … but careful
cleus. Their final structure has stood the test of time with experiments negative [sic] this interesting suggestion.”
confirmation by both X-ray diffraction (87) and a total Nonetheless, Shimizu in his book Ueber die Chemie und Phys-
synthesis of CDCA (88). Figure 2 shows the structure of iologie der Gallensäuren, published in 1935 (3), noted that:
CDCA and the numbering system for the steroid nucleus “Interesting intermediates in the biological catabolism of
and side chain that was proposed in the 1930s and has cholesterol to bile acids can be found in scymnol and te-
been used ever since. traoxybufostan [both C27 bile alcohols].” Even earlier, in
Bernal’s X-ray diffraction studies were the first bio- 1923, Ruzicka had proposed that bile acids are formed
logical application of this revolutionary technique. Bernal from cholesterol (92).
later used X-ray diffraction to deduce the structure of Proof that cholesterol was converted to bile acids had to
other organic molecules, and his pupils such as Dorothy await the introduction of isotopes into biochemistry by Ru-
Crowfoot Hodgkin extended the work, its ultimate triumph dolph Schoenheimer (93). Bloch, Berg, and Rittenberg

History of bile acid research 1557


prepared cholesterol labeled with deuterium, gave it to a Takashi Iida reported the synthesis of the 25R and 25S ste-
dog with a surgical connection between the biliary tract reoisomers of this compound (99).
and the renal pelvis; in this chronic preparation, bile is The first C27 bile alcohol to be isolated was scymnol, ob-
excreted in the urine. Cholic acid was isolated from urine tained from the bile of the Greenland shark (Scymnus bo-
and shown to have incorporated deuterium into its struc- realis) by Olof Hammarsten, working in Sweden in 1898
ture, thus establishing the conversion of cholesterol to C24 (100). The finding that the molecule had 27 carbon atoms
bile acids (94). Figure 3 shows the structure of cholesterol is a tribute to the precision and accuracy of the combus-
and the changes that occur as it is converted to CDCA. tion techniques that were used at that time to determine
(Although Konrad Bloch received the Nobel Prize in 1964 elemental analysis. A half century later, another C27 bile
for his extraordinary work on fatty acid and sterol synthe- alcohol was isolated from the bile of the American bull-
sis, his Nobel lecture does not even mention this key frog (Rana catesbeiana) by Kazuno, Masui, and Okuda
experiment.) (101). Subsequently, Haslewood (102) found the same
The first bile acid with a longer side chain than the com- bile alcohol in the European common frog (Rana tempo-
mon natural C24 bile acids was isolated from the bile of the ria), and gave it the common name “ranol”. Our extensive
toad Bufo vulgaris formosus by Shimizu and Oda (95) in survey of vertebrate bile acids (82) has established that
1934. The structure of this bile acid was established by most bile alcohols have an iso-octanoic acid (␣,␧-dimethyl-
Takahiko Hoshita as a trihydroxy bile acid with a double hexanoic acid) side chain. The exceptions identified to
bond at C-22 and a carboxyl group at C-24 (25). Thus this date are C26 and C25 alcohols that are present in the bile of
bile acid is a C28 bile acid having one more carbon than its some frog species (25, 26), as well as a C24 bile alcohol,
precursor cholesterol. Other C28 bile acids with a methyl petromyzonol that occurs in the lamprey as a disulfate
group on the side chain have been reported (26), but are (103, 104). Trace proportions of C24 bile alcohols with the
quite uncommon in nature. nuclear structure of cholic acid and CDCA are found in
C27 bile acids are much more common (82). They were the bile of many species (105).
isolated from the bile of a number of early evolving verte-
brates. In particular, bile from the alligator, which was The A/B ring junction
readily available in large amounts (studies at that time be- In most natural bile acids, whether C24 or C27, the junc-
gan with up to 5 l of bile), became the object of intense tion of the A and B rings is cis. This steric conformation is
study as it contains a great variety of C27 bile acids (96, 97). indicated by depicting the bond connecting the C-5 hydro-
The carbon skeleton of the side chain of C27 bile acids gen atom to the A/B ring juncture as a solid line or wedge.
was assumed to have remained unchanged from that of The much rarer “allo” bile acids (A/B ring junction is
the parent compound, cholesterol, but the location of the trans) are usually denoted by a dashed line coming from
carboxyl group was unclear. In 1952, Bridgwater and the ring junction to the hydrogen atom. For the nonste-
Haslewood (98) synthesized a defined side chain starting roid chemist, it is not easy to grasp that the orientation of
from the steroid nucleus. They then showed that their prod- the C-5 hydrogen atom indicates the type of A/B ring
uct was identical to 3␣,7␣,12␣-trihydroxy-5␤-cholestan-27- junction, and thus determines the shape of the two mark-
oic acid, the major bile acid isolated from the bile edly different bile acid epimers. The structures of cis- and
of crocodiles and alligators, and they confirmed its struc- trans-decalin, model compounds for the A and B rings of
ture by X-ray diffraction. Subsequently, the laboratory of bile acids are shown in Fig. 4. Every bile acid and bile alco-
hol has, in principle, a 5␣ and a 5␤ 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 Fie-
ser’s comprehensive text on steroids (7). Probably, it was
Windaus who introduced the prefix allo to denote 5␣ (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 5␣ bile acids from
5␤ bile acids is to prepare their per-oxo derivatives, which
are readily distinguished using optical rotatory dispersion
Fig. 3. Molecular structure of cholesterol (above) and CDCA (107).
(below) showing the changes in the cholesterol molecule when a C24 allo bile acids are the dominant primary bile acids
C24 bile acid is formed. The changes are: 1) reduction of the dou- only in lizards, although they are present in trace propor-
ble bond to give a 5␤ (A/B cis) A/B ring juncture; 2) a modified
tions in the bile of many species (82). If cholestanol (the
␤-oxidation of the side chain to remove three carbon atoms and
convert the terminal carbon to a carboxyl group; and 3) epimeriza- saturated 5␣-derivative of cholesterol) is administered to
tion of the 3␤-hydroxy group to a 3␣-hydroxy group. For a discus- rabbits, it is converted to 5␣ bile acids (107). Allocholic
sion of the enzymes involved, see (155). acid, the biological precursor of allodeoxycholic acid, has

1558 Journal of Lipid Research Volume 55, 2014


junction, the pattern of hydroxylation, on the steroid nu-
cleus and side chain or both, defines each individual bile
alcohol or bile acid.

Hydroxylation sites
The steroid nucleus. C24 PRIMARY BILE ACIDS. Bile acid no-
menclature, 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 cor-
rect 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, giv-
ing rise to the polysyllabic name of chenodeoxycholic
Fig. 4. Depictions of the two epimers of decalin, a saturated C10 (acid). Other trihydroxy bile acids were named for their
hydrocarbon that is used to illustrate the two A/B ring juncture animal source: muricholic acid (from rodents) and hyo-
epimers of bile acids. When the two bridgehead hydrogen atoms cholic acid (from pigs).
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.
We have proposed (82) that the root C24 bile acid should
When bile acid structure is shown in frontal view (Fig. 2), the only be CDCA, as every bile acid must have a hydroxyl group at
indication of the stereochemistry of the A/B ring juncture is the C-3 (from the C-3 hydroxyl group of cholesterol) and a
orientation of the hydrogen atom, i.e., whether ␣ or ␤. However, in hydroxyl group at C-7, as 7␣-hydroxylation of cholesterol
bile acids, the substituent at the upper bridgehead carbon atom is is the rate limiting step in the major pathway of bile acid
the C-19 methyl group. Often bile acid structure is shown without
biosynthesis. Thus, all natural primary C24 trihydroxy bile
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 acids can be considered as derivatives of CDCA. The only
5␤ (A/B cis) bile acids are much more common in nature than 5␣ exception to this useful convention occurs in those few
(A/B trans) bile acids. A/B trans bile acids are termed “allo” bile mammals (nutria, beavers, bears, and some cavimorphs)
acids. where UDCA, the 7␤-hydroxy epimer of CDCA, is a pri-
mary bile acid. The route of formation of this 7␤-hydroxy
been prepared synthetically and some its biological prop- epimer of CDCA is unknown. One can speculate on the
erties explored (108). Allo bile acids have also been identi- existence of a cholesterol 7␤-hydroxylase or a pathway that
fied in fecal bile acids (109), presumably generated by results in the epimerization of the C-7 hydroxy group.
bacterial enzymes acting on the 3-oxo-⌬ intermediate
4,6
The most frequent site (in terms of the number of ver-
that is formed during the process of C-7 dehydroxylation tebrate species) of additional hydroxylation of CDCA is at
(110). Enantiomeric bile acids, in which the steric configu- C-12 in cholic acid. The second most common site of hy-
ration of all ring junctions is reversed, have recently been droxylation is probably at C-16, because so many avian spe-
synthesized (111). cies have the 3␣,7␣,16␣-trihydroxy bile acid as a major
Bile alcohols (C27) with a 5␣ configuration are not un- biliary bile acid (82). Another common site of hydroxyl-
common in nature. The simplest biotransformation of ation is at C-6, occurring in hyocholic acid (pigs) and the
cholesterol (⌬ ) into a bile alcohol results in a 5␣ nucleus,
5
three muricholic acids (rats and mice). Other “third site”
and many early evolving vertebrates form A/B trans bile (nuclear) hydroxylations occurring in biliary bile acids of
alcohols (25, 26, 82). the healthy animal are at C-1 (␣ or ␤), C-2 (␤), C-4 (␤), C-5
The preceding discussion thus indicates that there are (␤), C-9 (␤), C-11 (␣), C-15 (␣), and C-19 (␤). Table 1
only four carbon skeletons in most natural bile acids and summarizes the species in which these rare biliary bile ac-
alcohols, the C27 cholestane skeleton (with either A/B cis ids occur, and the appropriate citation.
or A/B trans structure) and the C24 cholane skeleton (with C24 bile acids with four hydroxyl groups on the steroid
either A/B cis or A/B trans A/B structure). The side chain nucleus occur rarely. Some snakes have 3␣,7␣,12␣,16␣-
of each of the four skeletons may end in either a primary tetrahydroxy bile acids (82). Tetrahydroxy bile acids of
alcohol group or a carboxyl group. As noted above, C24 unknown structure are found in trace proportions in the
bile alcohols are very rare in nature. Thus, to simplify bile bile of many vertebrate species. After experimental liga-
acid and alcohol nomenclature, it is useful to divide all tion of the common bile duct, tetrahydroxy and even pen-
“cholanoids” and “cholestanoids” into only three great tahydroxy C24 bile acids occur in the plasma and urine of
classes, C27 bile alcohols, C27 bile acids, and C24 bile acids. mice (115). Such bile acids also occur in mice when the
In each of these classes, there are 5␣ and 5␤ epimers. canalicular bile salt export pump (BSEP) has been deleted
Within these three great classes, and for a given A/B ring genetically (116).

History of bile acid research 1559


TABLE 1. Common and uncommon sites of additional nuclear hydroxylation (in addition to the default
structure of the 3␣,7␣-dihydroxy compound) in primary C24 bile acids, C27 bile acids, and C27 bile alcohols

Uncommona

Class Common Site Natural occurrence References

C24 bile acids 6␣,6␤,12␣,16␣ 1␣ Australian opossum 474


1␤ Pigeons, neonates 475, 476
2␤ Neonates 477
4␤ Chinese pheasant 473
9␣ Bear 478
11␣ Sunfish 472
15␣ Swan, goose 478, 479
b
19 Neonates 480
C27 bile acids 12␣ 1␣ Tinamou 125
16␣ Ancient birds 473
C27 bile alcohols 12␣ 2␣ Arapaima 126
6␣ and 6␤ Elephants, hyrax, manatee 127
11␣ Sunfish 473
a
Hydroxylation occurs at C-5 with 24-nor (C23) UDCA in hamsters (481).
The bile acid described is a tetrahydroxy bile acid (3␣,7␣,12␣,19-tetrahydroxy).
b

An ad hoc committee of bile acid workers met in 16␣-trihydroxy) has not been identified, but bacterial
Freiburg in 1980 under the auspices of the Falk Founda- 16-dehydroxylation has been reported for some steroids
tion. The group proposed working rules for bile acid no- (118), and is likely to occur in some birds. Such 16␣-
menclature and bile acid abbreviations that were published dehydroxylation would result in the formation of CDCA,
and noted by the IUPAC nomenclature committee. At the that in turn would likely undergo 7␣-dehydroxylation to
suggestion of Gerhart Kurz of the University of Freiburg, it form lithocholic acid.
was agreed that there would be no more trivial names, ex- During transit through the large intestine, any hydroxyl
cept for conversational purposes (117). group can be oxidized and then reduced. If the reduction
C24 SECONDARY BILE ACIDS. As noted before, bile acids are is to the other possible epimer, a new epimer will be
modified by bacterial enzymes in the distal intestine. Mod- formed. 3␤-Hydroxy bile acids (“iso” bile acids) are major
ification of the hydroxyl groups forms new bile acids that fecal bile acids in man (119). 7␤-Hydroxy bile acids and
are termed “secondary”. This distinction has limitations, even 12␤-hydroxy bile acids are also formed, but to a far
because bile acids such as UDCA, or certain of the hy- lesser extent. Elimination of the 3-hydroxy group with for-
droxy-oxo bile acids, can be both primary and secondary. mation of a ⌬2 or ⌬3 bile acid may also occur (120).
A consideration of secondary bile acids is important be- Secondary bile acids may undergo additional hydroxyl-
cause they can be major biliary bile acids. After their for- ation during hepatocyte transport. Such rehydroxylation
mation in the distal intestine, they are absorbed in part. at C-7 generates the original primary bile acid that had
They return to the liver in portal venous blood, and tra- undergone bacterial 7-dehydroxylation in the intestine.
verse the hepatocyte where they may or may not undergo This rehydroxylation does not occur in man and the cow,
further metabolism before joining the pool of primary bile occurs to a limited extent in the rat and hamster, and is
acids (formed de novo from cholesterol) in the enterohe- complete in the prairie dog (121). Alternatively, second-
patic circulation. ary bile acids may undergo hydroxylation at a new site,
The major bacterial biotransformations of conjugated generating a novel “tertiary” bile acid. One example is
bile acids are deconjugation, 7-dehydroxylation, oxido- 16␣-hydroxylation of DCA to form “pythocholic” acid
reduction, epimerization, and side chain desaturation [de- (3␣,12␣,16␣-trihydroxy) that occurs in the python (75). In
tailed in (110)]. Deconjugation, i.e., hydrolysis of the amide the wombat, lithocholic acid undergoes hydroxylation at
bond between the terminal carboxylic acid group of the C-15 (122). Iso (3␤-hydroxy) bile acids are epimerized to
bile acid and the amino group of taurine or glycine, is medi- 3␣-hydroxy bile acids during transport through the hepa-
ated by many bacterial species and does not require anaero- tocyte (123, 124). Biotransformation of the 3␤-epimers of
bic conditions. Unconjugated bile acids formed by bacterial the muricholic and hyocholic acid families has not been re-
deconjugation are still considered primary bile acids. ported, but is likely to occur. In man, the 7␤-hydroxy group
In the large intestine, anaerobic modifications of of UDCA does not undergo epimerization during transport
the hydroxyl groups occur. The most striking change is through the hepatocyte, and the small proportion of UDCA
7-dehydroxylation that occurs via a 3-oxo-⌬4,6 nucleotide in the biliary bile acids is likely to be formed from CDCA in
intermediate (110). The 7-dehydroxylation product of the distal intestine by bacteria. Bacterial modification of
cholic acid is DCA; that of CDCA or UDCA is lithocholic bile acids and subsequent hepatic biotransformation has
acid. The 7-deoxy derivative of hyocholic acid and ␻- been termed by us, “damage and repair”.
muricholic acid is termed hyodeoxycholic acid and that The percent of secondary bile acids in biliary bile acids
of ␣- and ␤-muricholic acid is termed murideoxycholic is usually less than 50%, but there are exceptions. The
acid. To date, the C-7 deoxy derivative of “avicholic” (3␣,7␣, most remarkable example is in the rabbit, in which DCA

1560 Journal of Lipid Research Volume 55, 2014


constitutes 80% of the circulating bile acids (82). In some Haemulcholic acid was also identified in freshwater fish by
snakes, the 3␣,12␣,16␣-trihdroxy bile acid, which can be Haslewood during his Zaire River expedition (75).
formed by 16␣-hydroxylation of DCA, exceeds 50% (82). Side chain desaturation forming ⌬22 bile acids is a major
C27 BILE ACIDS. Nuclear hydroxylation in C27 bile acids pathway in rats (136, 137) and cavimorphs (82). Such bile
occurs at C-3 and C-7 (the default structure), and also at acids could arise from further ␤-oxidation of the side
C-12 in most species. In the Andean condor, hydroxyl- chain in the liver and/or by bacterial action in the distal
ation at C-16 occurs (82). We have identified a 1␤,3␣,7␣- intestine.
trihydroxy C27 bile acid in the bile of the tinamou, an ancient C27 BILE ACIDS. Elucidation of the structure of the C27
flightless bird (125). A tetrahydroxy bile acid with hydroxyl bile acids with hydroxyl group substituents on the C8 side
groups at C-3, C-7, and C-12 plus a 2␤-hydroxy group has chain was not simple, as side products such as lactones
been isolated from the bile of the large freshwater fish may be generated during alkaline hydrolysis. Unfortu-
Arapaima gigas (126). It seems likely that there are addi- nately, an enzyme preparation that hydrolyzes the amide
tional sites of nuclear hydroxylation in C27 bile acids yet to bond of C27 taurine conjugated bile acids is not available at
be discovered. present. (In contrast, cholylglycyl hydrolase, which rapidly
C27 bile acids undergo 7-dehydroxylation in the alligator cleaves the amide bond in C24 conjugated bile acids, is
(97), but the extent of 7-dehydroxylation of C27 bile acids available commercially.)
in other species has received little attention. So far, all reported C27 bile acids have only a single hy-
C27 BILE ALCOHOLS. In C27 bile alcohols, default nuclear droxyl group in the side chain. It occurs at C-22 in turtles,
hydroxylation occurs at C-3 and C-7; additional hydroxyl- at C-24 in lizards, and at C-26 in bullfrogs (25, 26); each of
ation occurs at C-12, and rarely at C-6 or C-16 (25, 26, these bile acids has four potential epimers. The four cho-
127). The C-3 hydroxyl group has a ␤-configuration in lat- lestanoic acid epimers with a hydroxyl group at C-24 have
each been synthesized by Une et al. (138). The most com-
imerol and myxinol, meaning that it remains unchanged
mon diastereoismer is varanic acid, which has the 24R,25R
from that of cholesterol. Hydroxylation at C-16 occurs in
configuration. In collaboration with the Iida and Une
myxinol. The bile of Arapaima gigas contains a bile alcohol
groups, we have found that the 24R,25S epimer is present
with a ␤-hydroxy group at C-2 in addition to hydroxyl
in the bile of several varanids (139).
groups at C-3, C-7, and C-12, so that C27 bile alcohols with
C27 BILE ALCOHOLS. As noted earlier, Hammarsten deter-
four hydroxyl groups on the nucleus do occur (126).
mined that he had isolated a C27 bile alcohol from the bile
No C27 bile alcohols are recognized as “secondary”, i.e.,
of the Greenland shark (100). The challenge of determin-
with hydroxyl groups modified by bacterial enzymes. How-
ing the structure of this compound would occupy chemists
ever, the presence of 7-deoxyscymnol in the megamouth
for many decades. Windaus, Bergmann, and König (140)
shark, as reported by Ishida et al. (128), suggests that bac- initially took up the problem of determining the structure
terial dehydroxylation at C-7 may also occur for C27 bile of Hammarsten’s compound (named scymnol) and pro-
alcohols. posed that it contained one primary and three secondary
hydroxyl groups with an epoxy ring. The three secondary
The side chain. C24 BILE ACIDS. With only four available
hydroxyl groups were found to be located in the sterol
carbons in the side chain, C24 bile acids are likely to un-
nucleus (at positions 3␣, 7␣, and 12␣), as scymnol could
dergo side chain hydroxylation only at C-23 or C-22, each
be converted into cholic acid by partial oxidation of the
of which has potential R and S epimers. The most com- side chain (141, 142). Because the side chain of choles-
mon C-23 hydroxy bile acid is termed “phocaecholic acid” terol has a C-27 methyl group, different paired combina-
and has the nucleus of CDCA. [The name “phoca” is given tions of carbons 23, 24, and 25 were proposed to contain
to the marine mammal family from which Hammarsten the epoxy group, with the primary alcohol group being on
(129) isolated the bile acid.] The structure of phocaecho- C-27. Bridgwater and Haslewood showed (using Ham-
lic acid was later established as the 23-hydroxy derivative of marsten’s original sample) that the epoxy group was an
CDCA by Windaus and van Schoor (130). The R and S artifact of alkaline hydrolysis, and that the side chain of
epimers have been synthesized (131, 132) and the R form scymnol actually contained two secondary hydroxyl groups
identified as the naturally occurring epimer (131). In ad- and a primary hydroxyl group bonded to sulfate (143,
dition to marine mammals, phocaecholic acid also occurs 144). In 1961, Cross used 1H-NMR to elucidate the struc-
in wading birds and some song birds (82). The 12-hydroxy ture of both scymnol and anhydroscymnol, the epoxide
derivative of phocaecholic acid also occurs in marine formed during alkaline hydrolysis (145). He found that
mammals and is therefore a natural tetrahydroxy bile acid. the side chain hydroxyl group of scymnol was unexpect-
A 3␣,7␣-dihydroxy bile acid with a hydroxy group at edly on C-26 (the only carbon with a single proton), con-
C-22 was shown to occur in the fish Parapristipoma trilinea- sistent with a side chain structure having hydroxyl groups
tum by the Hoshita laboratory (133) and given the awk- at C-24 and C-26, and a primary hydroxyl group at C-27.
ward name of haemulcholic acid. The compound was The structure of scymnol was ultimately established as
synthesized by both the Hoshita laboratory (134) and the 3␣,7␣,12␣,24,26,27-hexahydroxy-5␤-cholestane by com-
Pellicciari laboratory (135), and the S epimer was shown to parison with a synthetic standard (143). C-24 is chiral, and
occur in nature. Some of its physicochemical and biologi- the configuration of the 24-hydroxyl group was deter-
cal properties were characterized by Roda et al. (135). mined to be 24R in the crystallographic study of Ishida

History of bile acid research 1561


et al. (146). This group has also found that whereas most That bile acids are present in bile in “conjugated” form
shark species have the (24R,25R)-epimer of scymnol, a was recognized in the 19th century. At that time, neither
few species also form the (24R,25S)-epimer (147). Other the structure of the bile acid nor the site of linkage was
named C27 bile alcohols are tabulated in our previous re- known. This resulted in bile acid conjugates having a mis-
view (82). Additional information is available in the co- leading name, in that “taurocholate” implies that the cho-
gent review of Une and Hoshita (26). lic acid moiety is capable of ionization. We and others
Bile alcohols, just as bile acids, have been named for the spe- have suggested that the name “cholyltaurine” (analogous
cies in which they occur, and the name gives no clue what- to glycylglycine) would be more appropriate (117), but
soever to the structure of the bile alcohol. It is convenient few investigators have followed this proposal. The ex-
to think of the trihydroxy bile alcohol with ␣-hydroxyl tremely low proportion of unconjugated bile acids in biliary
groups at C-3 and C-7, and a primary alcohol function at bile acids has led some investigators to conclude that the
C-27, as the “root” C27 bile alcohol, and to look on other conjugation process is highly efficient. However, unconju-
bile alcohols as derivatives of that simplest bile alcohol. At gated bile acids, especially dihydroxy bile acids, if secreted
present, this root bile alcohol has no trivial name, but has into bile are rapidly absorbed by the biliary ductules (163,
been synthesized by the Mosbach group (148). 164), resulting in their removal from ductal bile. Thus, the
state of conjugation in canalicular bile can be considerably
Conjugation less than that of ductal bile.
N-acyl amidation with taurine or glycine. In virtually all
species, bile acids, whether C24 or C27, are found in bile in Other types of bile acid conjugation. When the term “con-
N-acyl amidated form, i.e., conjugated in amide linkage jugated” was applied to bile acids, it denoted N-acyl amida-
with taurine (or a taurine derivative) or far less commonly tion with taurine or glycine. When bile acid sulfation and
with glycine. Taurine conjugation is the rule in fish, am- glucuronidation were discovered to be important path-
phibians, reptiles, and most avian species. It also occurs to ways under some circumstances, it was necessary to distin-
a variable extent in mammals (82). In some angelfish spe- guish different modes of “conjugation” for bile acids. In
cies, N-methyltaurine conjugates are present (149), the N- principle, there are six modes of bile acid conjugation: N-
methyltaurine thought to be of dietary origin. Similarly, in acyl amidation (using glycine, taurine, or possibly other
some marine fish, bile acids are conjugated with cysteino- amino acids), sulfation, glucuronidation, conjugation with
lic acid, a hydroxy-ethyl derivative of taurine that is of di- glutathione, and conjugation with N-acetylglucosamine or
etary origin (150). Glycine conjugation occurs in a few glucose; and bile acids may have more than one mode of
conjugation. In vertebrates, the only bile acid sulfates that
avian species, some bovids, cavimorphs, and primates, in-
occur in appreciable proportions in biliary bile acids are
cluding humans (82). Sarcosine (N-methylglycine) conju-
the 3-sulfate of lithocholylglycine and lithocholyltaurine
gates have been synthesized and shown to be more
amidates that occur in man (165) and probably great apes
resistant to bacterial deconjugation during enterohepatic
(166). In patients with severe cholestasis, bile acid sulfa-
cycling (151–154). Some workers have considered bile
tion (mostly at C-3) followed by urinary excretion becomes
acid amidation to be the final step in bile acid biosynthe-
a major pathway (167, 168). In the bile duct-ligated ham-
sis (155). It seems more logical to us to define the bile
ster, esterification with sulfate occurs at C-7 (169).
acid biosynthesis pathway as ending with a mature bile
In the Mayo Clinic studies of Thistle and Schoenfield, in
acid or bile alcohol before it undergoes a phase II bio- which bile acids were administered to gallstone patients
transformation step such as N-acylamidation, sulfation, or (35), one group received hyodeoxycholic acid (3␣,6␣-
glucuronidation. dihydroxy). Because hyodeoxycholic acid should be ab-
The process of conjugation occurs both in hepatocytes sorbed efficiently from the small intestine by passive
and in cholangiocytes (156). However, conjugation in the mechanisms, its absence in biliary bile acids was com-
hepatocyte greatly exceeds that occurring in cholangio- pletely mystifying. In 1978, B. Almè and J. Sjövall reported
cytes. The conjugation process begins with the formation a patient with malabsorption who excreted 6-hydroxy bile
of a bile acid adenylate (157) followed by formation of the acids in urine that were present as O-linked (ethereal)
CoA thioester. A bile acid aminotransferase then transfers glucuronides (170). Five years later, Sacquet et al., work-
the CoA bile acid to the amino group of glycine or taurine; ing in the laboratory of Reca Infante in Paris, reported
the human enzyme has been cloned and shown to mediate that when hyodeoxycholic acid was administered to hu-
conjugation with both glycine and taurine (158). Conjuga- man subjects, it was largely excreted in urine as its glucuro-
tion of newly synthesized bile acids occurs in peroxisomes nide (171), explaining its absence in biliary bile acids. This
(159), and recent work suggests that “reconjugation” of group did not know the site of glucuronidation but pro-
unconjugated bile acids returning from the intestine also posed that it was at C-3.
occurs in these organelles (160). Other than their pKa Four years later, Anna Radominska-Pyrek and colleagues
value, which in turn is responsible for the pH-solubility showed that human microsomes rapidly glucuronidate
profile (see later), there is little difference in the biologi- hyodeoxycholic acid at C-6 (172). Thus the curious fate of
cal properties of glycine- and taurine-conjugated bile ac- hyodeoxycholic acid in humans was finally settled. It pre-
ids, except that taurine conjugates are deconjugated by sumably results from defective N-acylamidation, and hyo-
bacteria more slowly than glycine conjugates (161, 162). deoxycholic acid is consequently metabolized in the same

1562 Journal of Lipid Research Volume 55, 2014


manner as a C24, nor (C23) dihydroxy bile acid (see below), acid extruders in the small intestine epithelium are not
i.e., by glucuronidation in the endoplasmic reticulum. To upregulated (182). In patients with biliary obstruction,
date, this pathway has not been identified in any other bile acid elimination is by urinary excretion, albeit with
animal. When hyodeoxycholic acid is given to rats, it is greatly increased levels in plasma and liver. In addition to
probably conjugated with taurine and not glucuroni- cholesterol excretion in bile (as such and after conversion
dated (173). to bile acids), there is a small flux of cholesterol directly
When C23 dihydroxy-bile acids, are administered to from the small intestine (183).
animals including man, little amidation occurs, as both For a bile acid to serve as the chemical form of choles-
the CoA ligase and the glycine/taurine amino acid terol elimination, there are only a few molecular require-
transferase act to only a very limited extent on bile acids ments. The molecule must be too polar to be absorbed
with a shortened (C4) side chain (174). Such bile acids passively and too large to pass through the paracellular
undergo glucuronidation both at C-3 and C-23 (163, 164). junctions between enterocytes. It also must be resistant to
In man, when norUDCA is given, it forms the C-23 ester pancreatic enzymes.
glucuronide which is excreted in both bile and urine The bile acid molecule is too large to pass via the tight
(175). In rats, N-acyl amidation is also much less effi- junctions of the intestinal epithelium. Amidation with gly-
cient for ␣-hydroxy bile acids (131, 132) and hydrophilic cine or taurine converts unconjugated bile acids from
epimers of cholic acid, for example 3␣,7␤,12␤-trihydroxy- weak acids (pKa of 5) to a pKa of 4 for glycine amidates and
cholanoate (176). a pKa of less than 2 for taurine conjugates (184). Thus, bile
In patients ingesting UDCA, UDCA also undergoes con- acids are present in bile and intestinal content as organic
jugation with N-acetylglucosamine at C-7, in addition to anions, and therefore will not cross membranes unless a
amidation with glycine or taurine at C-24 (177). In Nie- transporter is present. Bile acid glycine and taurine ami-
mann-Pick disease type C, a novel ⌬5 3␤,7␤-dihydroxy cho- dates are resistant to pancreatic carboxypeptidases, whereas
lestanoic acid is formed (178); this compound may be bile acids amidated with amino acids other than glycine
considered an acidic 7␤-hydroxy derivative of cholesterol. or taurine (with the exception of aspartic acid and cysteic
This compound undergoes sulfation at C-3, conjugation acid), are hydrolyzed rapidly by pancreatic carboxypepti-
with N-acetylglucosamine at C-7 (178), and amidation with dases (185).
glycine or taurine at C-24. In both circumstances, the mul-
In vertebrates with a cecum, deconjugation and dehy-
tiply-conjugated metabolites are eliminated in urine rather
droxylation convert bile acids to membrane permeable
than bile, probably because they are not substrates for the
molecules and they are in part absorbed, as signaled by the
canalicular transporters. For UDCA, conjugation with N-
presence of unconjugated bile acids in the systemic circu-
acetylglucosamine is a very minor pathway, less than 0.3%
lation. Isao Makino, who had worked in the Sjövall labora-
of the oral dose.
tory in Stockholm and returned to Japan, was among the
Glutathione conjugates of bile acids (presumably as
first to report the presence in healthy individuals of un-
thioesters) have been identified in rat and human infant
conjugated bile acids in plasma using GC (186). Some de-
bile by the group of Shigeo Ikegawa, but their concentra-
cades later, this early finding was confirmed by Kenneth
tion is several orders of magnitude less than that of the
Setchell et al., using GC-MS (187). Nonetheless, absorp-
major biliary bile acids (179).
To date all C27 bile acids have been found to be conju- tion from the cecum is not complete, as unconjugated bile
gated exclusively with taurine. Bile alcohols are esterified acids have poor solubility at the acidic pH (pH 5.5) of the
with sulfate, and thus have a pKa similar to that of taurine cecum and are bound to bacteria and unabsorbed dietary
conjugates. components.
Unconjugated (and conjugated) bile acids that are not
Structure-function relationships absorbed are finally eliminated from the body by the pro-
In the steady state adult animal, cholesterol input, from cess of defecation. As there is negligible excretion of bile
de novo synthesis and dietary cholesterol, must be bal- acids in urine, breath, and skin, and as the bile acid nu-
anced by fecal excretion as cholesterol and its bacterial cleus is considered to be indestructible by bacterial enzymes,
metabolites (neutral steroids) and bile acids (acidic ste- fecal excretion of bile acids is equal to their synthesis from
roids). There is thus a mole fraction consisting of total bile cholesterol. However, it must be stressed that there is little
acids (numerator) divided by total cholesterol secretion information on the fecal bile acid composition across the
(acidic plus neutral steroids) (denominator). In man, based vertebrate spectrum (188). There are some species that by
on balance studies, this figure is 0.46; in the mouse it is our analyses have very low fecal bile acids, suggesting that
0.45, whereas in the hamster it is 0.57, according to John the bile acid molecule can in fact be degraded by bacte-
Dietschy and Steve Turley (180). The very low cholesterol ria. The degradation of bile acids by individual bacterial
proportions in the biliary lipids of many vertebrate species strains, with bile acids as the major carbon source, has
(181), suggest that in most vertebrates this number should been characterized in great detail by S. Hayakawa (189).
be well above 0.5. The route of bile acid excretion is solely The biological relevance of these studies is unclear. To
via bile, as the amount of bile acids in urine is negligible. date, there are no studies in which bile acids tagged with
14
In patients with complete biliary obstruction, fecal bile C in the steroid nucleus are administered and 14CO2 in
acid output is extremely low, indicating that possible bile breath is collected.

History of bile acid research 1563


There is very little information on the fate of bile alco- bile acids by Jan Sjövall to separate individual unconju-
hols in the distal intestine. In the Asiatic carp we found gated bile acids (191). Sjövall and Haslewood then collab-
unchanged 5␣-cyprinol sulfate in the distal intestine (190). orated to apply the method to Haslewood’s vast collection
Even if there were hydrolysis of the ester sulfate, the liber- of bile samples from different animals (192).
ated bile alcohol is likely to have negligible passive mem- Gas-liquid (partition) chromatography was developed
brane permeability because of the number of hydroxyl some 10 years later by A. J. P. Martin working with A. T.
groups. James. The earliest application to bile acids came in the
For solubilizing lipids in the form of mixed micelles, 1960s (193, 194), and Eneroth and Sjövall tabulated reten-
bile acids must be amphipathic, i.e., they must have a polar tion times for different stationery phases in 1971 (195). A
side and a nonpolar side. So far as is known, all major nat- decade later, capillary GC was described, greatly increas-
ural biliary bile acids and bile alcohols are amphipathic, ing resolving power. GC using flame ionization detectors
although the hydrophobic side (␤ face) is somewhat reduced affords both separation and quantification. However, only
in UDCA. Figure 5 shows a space-filling model of a bile when a peak is further analyzed by MS can one be certain
acid molecule, emphasizing its amphipathic nature. To that the peak is truly a bile acid.
the best of our knowledge, no one has synthetized a mol- TLC became widely used when a commercial device for
ecule unrelated to the bile acid molecule which has the coating the adsorbent (usually silicic acid or alumina) on
same physicochemical properties of bile acids as regards glass plates became available, and excellent adsorbents
solubilizing membrane lipids. 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
MEASUREMENT bile acids, molecules separate according to the number
and orientation of hydroxyl groups, but a clear separation
Isolation and chromatographic separation of DCA from CDCA with the commonly employed solvent
Elucidation of the metabolism of bile acids in health systems does not occur. Moreover, quantification of indi-
and disease requires accurate, reliable, and sensitive meth- vidual spots was difficult. Nonetheless, TLC is a highly use-
ods for measurement, as well as techniques for extracting ful quasi-quantitative method for assessing the purity of
bile acids (or alcohols) from complex biological matrices. bile acids and monitoring chemical reactions.
The complexity of bile acid structure and the enormous The senior author was privileged to have access to the
range in polarity of bile acids and their conjugates have first TLC apparatus to enter Sweden, purchased in 1960 by
always proved a challenge to the analytical chemist. A co- Borgström for the separation of lipids. Borgström had
gent review by W. Griffiths and J. Sjövall on current analyti- been impressed with the remarkable separations of neu-
cal approaches was published in 2010 (9). These two tral lipids that were obtained using this technique by
chemists have contributed greatly to the advances in bile Malins and Mangold (197), then working at the Hormel
acid analysis. Institute in Minnesota. With TLC using silicic acid as the
The modern era of bile acid analysis began with the de- stationery phase and phosphomolybdic acid (in ethanol)
velopment of improved chromatographic methods for as the detection reagent, most bile acids (as well as unsatu-
separation. Paper chromatography based on liquid-liquid rated lipids) appear as deep blue spots on a brilliant yellow
partition was developed in the 1940s by the great English background (the colors of the Swedish flag). A technique
chemists A. J. P. Martin and R. L. Synge, who shared the was described for coating silicic acid on microscope slides
Nobel Prize in 1952. In the 1950s, it was first applied to (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.
Reverse-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 re-
view in 1971 (195). Some of their values have been shown
as actual simulated chromatograms in a chapter by the se-
nior author (202).
In our laboratory, we have found TLC to be of great
value for quantifying hepatic biotransformation of radio-
Fig. 5. Silhouette of a C24 conjugated bile acid molecule empha- active bile acids. Small (1 mm) bands of adsorbent can be
sizing its possession of a hydrophobic side (␤ face) and a hydro-
directed into scintillation counter vials, using the auto-
philic side (␣ face). Common sites of hydroxylation in addition to
the default structure (3␣, 7␣) are at C-6, C-12, and C-16; all of these matic device described by Snyder and Kimble (203), or
are on the hydrophilic side of the molecule. Modified from Roda manually, by transferring the adsorbent to a vial using a
et al. (360). brush.

1564 Journal of Lipid Research Volume 55, 2014


HPLC was developed in the 1970s and was soon applied Enzymatic and competitive binding assays
to bile acids. Today, the technique, when used for bile acid Development of an enzymatic technique for determin-
analysis, is usually executed in the reverse-phase mode, ing total 3␣-hydroxy bile acids based on oxidation of the
i.e., with C18 or C8 alkyl-bonded silicic acid as the lipophilic 3␣-hydroxy group by a 3␣-hydroxy steroid dehydrogenase
solid phase. Numerous laboratories have reported the was a major advance in bile acid analysis (207), especially
power of this analytical method for separating and mea- for workers in the field of bile acid physiology. This tech-
suring conjugated bile acids in bile and intestinal content nique was based on the pioneering work of Talalay (208),
(9). Conjugated, i.e., N-acyl amidated, bile acids have an who developed enzymatic analysis of 3␣-hydroxy hor-
amide bond that has absorbance at 205 nm, permitting monal steroids using a steroid dehydrogenase isolated
their measurement in the effluent. Unconjugated bile acids, from Pseudomonas testosteroni. Details of the method were
nonamidated bile acid sulfates, and bile alcohol sulfates clarified by Turley and Dietschy (209). The sensitivity of
are not detected at 205 nm. HPLC with UV detection is too the method was increased by measuring the reduced NAD
insensitive for analysis of serum bile acids unless large vol- by changes in fluorescence or by coupling the oxidation to
umes of serum are used (204). One solution to the nonde- reduction of a tetrazolium dye (210). The enzyme tech-
tection of unconjugated bile acids and bile alcohol sulfates
nique using 3␣-hydroxysteroid dehydrogenase does not
is to use a light scattering detector that responds approxi-
measure 3␤-hydroxy bile acids. Thus, when used for deter-
mately equally to all bile acids (205). A second approach,
mination of fecal bile acids, it will underestimate total bile
useful for unconjugated bile acids, is to couple the bile
acids (119). An enzyme recycling system has recently been
acid to a chromophore such as the phenylacyl group
marketed by Diazyme™ and is quite sensitive.
(206).
In our laboratory, based on collaboration with Marlene
In our laboratory, we developed an HPLC method for
DeLuca, Aldo Roda developed an enzymatic biolumines-
separating the 12 conjugated bile acids predominant in
cence assay for primary bile acids. The method was based
human bile (165), and we have found this method to be
on coimmobilizing on beads a 7␣-hydroxysteroid dehydro-
essential in our analysis of vertebrate bile samples (82).
genase, an oxido-reductase, and luciferase (211). The
Nonetheless, when used alone for bile analysis, there are
method was validated by showing excellent agreement
many unknown peaks that may or may not be bile acids.
Therefore peak identification requires confirmation by MS. with GC-MS measurements (212). However, this method,
despite its simplicity and sensitivity, has not been widely
MS adopted. Limitations in the enzymatic method are sum-
marized in the review by Griffiths and Sjövall (9).
MS had been developed in the 1930s and was used to
All of the chromatographic techniques discussed above
analyze mixtures of small molecules. MS of large organic
are labor intensive and time consuming. For measuring
molecules emerging from the gas chromatograph was de-
veloped by R. Ryhage in Sweden, and the first commercial diurnal variations in plasma levels of total bile acids, two
instrument for this purpose was marketed by LKB in 1965. approaches were taken to develop a rapid simple proce-
William Elliott wrote in 1988: “Two years later [1965], I dure for measuring total bile acids. The first was the en-
returned to the Karolinska to devote the summer to learn- zyme method that measures total 3␣-hydroxy bile acids, as
ing about the GC-MS which Ragner Ryhage had built, and just discussed. The second approach was the development
was then produced by LKB. Our LKB 9000 is still operat- of competitive binding techniques, such as radioimmuno-
ing, beginning its 23rd year.” Since then, there have been assay. What is measured depends on the affinity of the an-
continuous improvements in techniques for sample vola- tibody, and thus one determines “immunoreactive” bile
tilization and instrument sensitivity, culminating in the acids. At the Mayo Clinic, Wilfred Simmonds et al. (213)
MALDI analysis of proteins. In our opinion, anyone who developed a radioimmunoassay specific for cholyl conju-
wishes to do a complete bile acid analysis must have gates, which was then used by LaRusso et al. (214) to de-
HPLC/MS/MS available. It is also desirable to have GC- fine the time course of the postprandial elevation of such
MS availability when needed, as capillary GC has greater cholyl conjugates in health and in patients with bile acid
resolving power than HPLC. malabsorption. Later, Schalm et al. (215) developed a ra-
The wedding of HPLC and MS/MS is thus an essential dioimmunoassay for chenodeoxycholyl conjugates, and
step in the analysis of unconjugated and conjugated bile used it to show that the postprandial peak of CDC conju-
acids. Currently, internal standards containing three or gates occurs earlier than that of cholyl conjugates (216), as
more deuterium atoms are not available for many bile shown in Fig. 6. Other radioimmunoassays have been de-
acids and are sorely needed. For a total analysis, class sepa- veloped, as summarized in the review by Griffiths and
ration by ion exchange chromatography developed by Sjövall (9).
Setchell and Sjövall should be performed before HPLC/ All of the above methods for measuring bile acids iso-
MS/MS (9). For state of the art analysis, either capillary late the bile acids from the body tissue or fluid, and then
GC or capillary HPLC will be required to separate com- quantify the bile acids. We have long needed a bile acid-
plex mixtures of bile acids. For unknown bile acids, sensitive molecule whose signals would provide informa-
structure assignment requires NMR for elucidation of tion on the concentration of bile acids within the living
structure, and ultimately synthesis for confirmation of as- cell. This need has now been supplied by the development
signed structure. of an indicator molecule consisting of a protein possessing

History of bile acid research 1565


Fig. 6. Time course in healthy subjects of levels in
peripheral venous plasma of immunoreactive conju-
gates of cholic acid (cholyl conjugates) and those of
CDCA (here termed “chenyl” conjugates) in re-
sponse 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 conju-
gates are also higher than those of cholyl conjugates
despite similar proportions in bile because of lower
fractional extraction by the liver. From (216).

a bile acid binding site (based on the binding site of FXR) Urinary bile acids represent filtered bile acids plus any bile
and two reporter molecules whose fluorescence is deter- acids secreted by the tubules minus those absorbed by the
mined by the extent to which bile acids occupy the bind- tubules. Ductal bile which is usually analyzed differs from
ing site, the greater the binding, the less the fluorescence. canalicular bile by that fraction of conjugated and uncon-
The technique, as described by van der Velden et al. (217), jugated bile acids that are absorbed by the biliary ductules.
uses the well-known Forster resonance energy transfer Gallbladder bile is ductal bile minus bile acids absorbed
(FRET) principle, and has already been used to character- from the gallbladder. Fecal bile acids represent bile acids
ize bile acid transport into isolated hepatocytes. entering the colon with or without modification by bacte-
rial enzymes minus those that were absorbed.
Diagnostic utility of plasma bile acid levels The power of current analytical techniques may result in
Despite the simplicity of competitive binding techniques an enormous variety of bile acids being detected. When such
for measuring conjugated bile acids, they have not been results are tabulated, we suggest organizing bile acids by pri-
widely adapted by the clinical chemists. The measurement mary bile acids, and then listing secondary bile acids derived
of plasma bile acids is not part of the usual panel of liver from the primary bile acid underneath their precursor. Bile
tests (aminotransferases, alkaline phosphatase, total bili- acids that are present in trace proportions should be lumped
rubin). The major reason is that bile acid levels are prob- together to simplify the presentation. The choice of the ana-
ably inferior in sensitivity and specificity in detecting liver lytical method should be determined by the biological ques-
injury when compared with the standard liver panel. This tion that is being asked. For nomenclature in reporting bile
conclusion is based on clinical studies (218, 219). In addi- acid analyses, the recommendations of an ad hoc nomencla-
tion, a recent animal study (220) in which liver injury was ture committee (117) should be followed.
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 measure- BILE ACID METABOLISM: TRANSPORT AND THE
ments, there is the belief that the current liver tests are ENTEROHEPATIC CIRCULATION
adequate. Moreover, the instruments that automate deter-
mination of a serum “chemical panel” do not have a bile Bile acid transport
acid “channel”. For purely cholestatic diseases, such as pri- Overview. Glycine and taurine conjugated bile acids
mary cholestasis of pregnancy, the level of serum bile acids are fully ionized at the pH conditions prevailing in plasma,
has value for predicting prognosis (221). bile, and small intestinal content. Therefore, these mole-
cules will not cross a cell membrane and enter a cell unless
Determinants of bile acid profiles in body fluids a transporter is present. What is responsible for the en-
In assessing the significance of bile acid concentrations terohepatic circulation is the vectorial transport of conju-
that in turn define the bile acid profile, it is important to gated bile acids through the ileal enterocyte and the
realize that plasma, hepatic, biliary, urinary, and fecal bile hepatocyte. The enterohepatic circulation of bile acids
acids all have a different composition in health. Plasma results from the action of two chemical pumps and three
bile acids, in health, represent the balance between instan- fluid flows. The chemical pumps are the vectorial trans-
taneous intestinal input and net hepatic uptake, with first port systems of the ileal enterocyte and the hepatocyte,
pass clearance ranging from 40 to 90%, depending on the each of which has basolateral and apical transporters. The
bile acid. Hepatic bile acids (as distinguished from hepato- fluid flows are the aboral movement of intestinal content
cyte bile acids) represent the balance between input of due to intestinal propulsive activity, portal venous blood
unconjugated and conjugated bile acids, subsequent bio- flow that transports bile acids from the intestine to the
transformation of these molecules, as well as bile acids that liver, and biliary flow that returns the bile acids to the in-
have been secreted by transporters into the canaliculus. testine. The molecules that constitute the circulating pool

1566 Journal of Lipid Research Volume 55, 2014


of bile acids must be substrates for the vectorial transport Reabsorption of bile acids from the small intestine gives
systems of both the ileal enterocyte and the hepatocyte in a cycling “pool” for each bile acid. The size of the bile acid
order to circulate enterohepatically. pool and its turnover rate differ for each bile acid, al-
The initial input into the hepatic limb of the enterohe- though the turnover rate of CDCA and DCA are about the
patic circulation consists of primary bile acids formed and same. Most of each individual bile acid pool is stored in
conjugated in the pericentral hepatocytes. The newly synthe- the gallbladder during overnight fasting, and biliary bile
sized conjugated bile acids are pumped across the canalicular acid composition is determined by the relative propor-
membrane and flow in canalicular bile from the pericentral tions of each type of bile acid, this in turn being deter-
region to the periportal region, their direction of flow being mined by the sizes of the individual bile acid pools.
opposite from that of sinusoidal blood. In the periportal re-
gion, newly synthesized bile acids are joined by bile acids re- Transport by the ileal enterocyte: the apical transporter. The
turning from the intestine and pumped into bile. first person to observe preferential absorption of conju-
The return pathway of the enterohepatic circulation gated bile acids by the ileum was Hermann Tappeiner,
features three inputs into the portal venous blood from who presented his findings to the Vienna Academy of Sci-
bile acids that have been absorbed by enterocytes. The ences in 1887 (223). Tappeiner studied the absorption of
dominant input into portal venous blood consists of “un- taurocholate from different regions of the small intestine
damaged” conjugated bile acids that have been absorbed using anesthetized dogs. He found that taurocholate was
by the ileal transport system. A second input is that of un- absorbed preferentially in the ileum. Rather than conclud-
conjugated primary bile acids formed by bacterial decon- ing that the ileum had special properties, Tappeiner was
jugation in the distal small intestine and large intestine. A puzzled by the failure of the proximal intestine to absorb
taurocholate. Verzar, working in Hungary, authored one
third input is that of “secondary” (damaged) bile acids
of the first monographs on intestinal absorption (224). He
that are formed by bacterial modification of the hydroxyl
was skeptical of the results of Tappeiner and directed his
groups of primary bile acids. Only a fraction of the second-
colleague Fröhlicher to repeat Tappeiner’s experiments.
ary bile acids that are formed are absorbed, and those that
Fröhlicher obtained identical results (225), again showing
are not absorbed are eventually eliminated by defecation.
that ileal absorption of conjugated bile acids was greater
As discussed previously, those secondary bile acids that
than that of jejunal absorption. However, in Verzar’s book
are absorbed from the distal intestine may undergo up-
on intestinal absorption, the concept of specific transport-
take by the hepatocyte, or in some cases urinary excretion
ers with differing substrate affinities is not considered. At
(or both). After hepatocyte uptake, “damaged” bile acids
that time, the mechanism of absorption of small molecules
undergo “repair” in the hepatocyte by “reconjugation” as
was considered to be passive, analogous to diffusion.
well as reepimerization of hydroxy groups (from ␤ to ␣)
Baker and Searle, working at the University of Iowa, re-
and reduction of oxo groups to hydroxy groups. They are discovered the work of Tappeiner and Fröhlicher, and
then secreted into bile to join the recycling primary bile confirmed the selective ileal absorption of taurocholate in
acids. In man, the flux of bile acids returning from the in- the rat ileum in 1960 (226).
testine is at least 20-fold greater than the input of newly A decade before, T. H. Wilson, an American working in
synthesized bile acids. More details of bacterial damage the laboratory of G. Wiseman at the University of Shef-
and hepatocyte repair are given in the introduction. field, developed the everted gut sack technique and used
In man, the major secondary bile acids in colonic con- this method to show that glucose was actively transported
tent are lithocholic acid, formed by 7-dehydroxylation of (227). (It is rumored that this technique was the result of
CDCA, and DCA, formed similarly from cholic acid. In a drunken party in Paris some years before, where physi-
man, lithocholic acid is not only N-acylamidated, but also ologists were bemoaning the difficulty of studying intesti-
sulfated in part. Such sulfo-lithocholyl amidates are se- nal absorption in the intact animal. Someone, never
creted into bile, but are poorly absorbed from the small identified, said the only solution was to turn the animal
intestine (222), so they do not have an enterohepatic cir- inside out. And someone else is supposed to have said,
culation. Those unsulfated amidates secreted in bile re- “Maybe that’s a good idea!”) In 1960, Robert Crane, who
turn to the liver, undergo sulfation in part, are resecreted had studied glucose absorption since his postdoctoral
into bile, and eliminated via the fecal route. The end re- studies with the Cori’s in St. Louis, published his idea that
sult is that lithocholic acid has a very rapid turnover in glucose absorption was sodium dependent, and that the
man, and sulfated and unsulfated lithocholyl amidates downhill transport of sodium served to energize the uphill
constitute less than 5% of biliary bile acids (165). In ro- transport of glucose (228). (The late Robert Crane was a
dents, lithocholic acid is not sulfated but undergoes hy- tall handsome charmer who always had an eye for attrac-
droxylation at C-6 or C-7 (15). tive women. At a party, he tried to persuade a lovely young
As noted previously, DCA may pass through the hepato- scientist to go home with him; when she refused, he started
cyte without further metabolism, or may be rehydroxyl- attacking her data.)
ated to varying degrees in a species-dependent manner. In 1961, Leon Lack and Ike Weiner, young faculty mem-
The “repaired” and reconjugated bile acids are secreted bers at Johns Hopkins, were discussing how to teach medi-
from the hepatocyte and join the cycling conjugated bile cal students about the enterohepatic circulation. They
acids. realized that nothing was known about the mechanism of

History of bile acid research 1567


bile acid absorption. They used the everted sac technique decided to attempt to clone the ileal bile acid transporter.
to show that the last fourth of the small intestine of rats Dawson was well-educated for the task, having worked with
and guinea pigs actively transports conjugated bile acids Joseph Goldstein, Michael Brown, and David Russell at
against a concentration gradient (229). Their data are il- University of Texas Southwestern in Dallas. Dawson, now
lustrated in Fig. 7. As noted earlier, the senior author, relocated to Wake Forest University, reported the success-
working with his mentor Bengt Borgström and Göran ful cloning of the ileal apical sodium-dependent bile acid
Lundh (a surgeon and brother-in-law of Borgström), per- transporter [protein, apical sodium-codependent bile acid
fused the small intestine of healthy volunteers and ob- transporter (ASBT); gene, SLC10A2], using a somewhat
tained strong evidence for ileal absorption of conjugated different expression technique (236). This achievement,
bile acids in man (20). Peter Holt, working in the labora- more than a century after Tappeiner’s original study, per-
tory of Kurt Isselbacher at Harvard, showed that conju- mitted the testing of individuals for defects in the gene
gated bile acid absorption in the everted sac preparation encoding ASBT, as well as providing a molecular target
was sodium-dependent (230). Thus, it seemed that a Na+- for pharmaceutical companies who might wish to develop
dependent transporter for conjugated bile acids was pres- an inhibitor.
ent in ileal enterocytes. (The term “enterocyte” was coined In Dawson’s original paper, he found that ASBT was
by Christopher Booth, a prominent English gastroenter- also present in the kidney, in agreement with earlier stud-
ologist and bibliophile, who discovered that vitamin B12, ies by F. Wilson et al. (237). Later, the laboratories of G.
like conjugated bile acids, was also absorbed predomi- Alpini in Texas (238) and N. LaRusso (239) at the Mayo
nantly in the ileum.) In 1978, H. Lücke, G. Stange, R. Clinic showed that ASBT was also present in cholangio-
Kinne, and H. Murer, working at the Max-Planck Institute cytes. ASBT is highly specific for bile acids, although the
in Frankfurt, prepared vesicles from ileal enterocyte brush integrity of the 7␣-hydroxyl group seems more important
borders, and used this preparation to show that taurocho- than that of the 3␣-hydroxyl group, as some substituents
late uptake was carrier mediated and Na+ dependent can be added to the 3␣- position without impairing trans-
(231). Attempts were made to isolate the apical conju- port (240). As yet, the transporter has not been crystal-
gated bile acid transporter by G. Kurz, working in Freiburg, lized, although the structure of a bacterial homolog has
using the photoaffinity technique with 3H-tagged azido recently been published (241). That paper proposes that
bile acids (232). These studies were continued by his stu- two sodium ions act allosterically to open a binding pocket.
dent, Werner Kramer at the Hoechst Company in Frank- An overview of the SLC10 transporter family is available
furt (233), but they failed to clearly identify the ileal apical (242).
bile acid transporter.
During this time, ideas of cell polarity were developing Transport by the hepatocyte: bile acid uptake. Bile acid up-
(234). Thus, it seemed that at a minimum, four transporters take by the liver was known to be highly efficient. For ex-
would be required for the enterohepatic circulation of bile ample, in an isolated perfused dog preparation studied at
acids. These would be an apical and basolateral transporter the Mayo Clinic by N. Hoffman et al., uptake of taurocho-
in the ileal enterocyte, and an apical (canalicular) and baso- late was 100% and that of the taurine conjugate of CDCA
lateral (sinusoidal) transporter in the hepatocyte. was 70% (243). In a study published that same year from
The great breakthrough, in our judgment, came in 1987 the Erlinger laboratory in Paris, first pass extraction of tau-
with the use of expression cloning by Matthias Hediger rocholate in the dog was found to be 40–80% and satura-
working in the laboratory of Ernie Wright at University of ble, consistent with uptake being carrier mediated (244).
California, Los Angeles. Using the technique of injecting Conjugated bile acid uptake was saturable, and using ei-
mRNA into frog eggs (obtained from Xenopus laevis), they ther hepatocytes (245) or the isolated perfused rat liver
were able to report the cloning of the sodium-coupled (245) was sodium-dependent (246). Thus it seemed highly
transporter for glucose (235). Seven years later, Paul Dawson likely that one or more sodium-coupled transporters were
present in the sinusoidal membrane and mediated the ef-
ficient uptake of bile acids.
A sodium-dependent basolateral hepatocyte protein,
named Na+ taurocholate cotransporting polypeptide
(NTCP) (sodium taurocholate porter), was cloned in
1991, again using expression cloning, by Bruno Hagen-
buch, Bruno Stieger, and colleagues working in the labo-
ratory of Peter Meier in Zurich (247). When the Na+
coupled ileal transporter ASBT was cloned 4 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
Fig. 7. Schematic depiction of the work of L. Lack and I. Weiner were related, and both used the sodium gradient to power
on conjugated bile acid (here, taurocholate) absorption by everted
sacs of rat intestine, showing preferential and active absorption by the uptake. However, NTCP has a much broader specific-
the distal ileum. At time zero, bile acid concentrations are identical ity than ASBT (249, 250). NTCP can be considered an an-
on both sides of the intestine. Modified from (229). ion transporter that transports not only bile acids but also

1568 Journal of Lipid Research Volume 55, 2014


other organic anions into the hepatocyte from plasma, bile acids. They suggested that FATP5 was involved in
whereas ASBT selectively conserves solely conjugated bile both the uptake and CoA formation of unconjugated bile
acids, thereby generating their enterohepatic circulation. acids returning from the intestine. It is attractive to pos-
NTCP is not the only bile acid transporter in the baso- tulate that this transporter mediates not only adenylate
lateral (sinusoidal) membranes of the hepatocyte. A family formation but also subsequent CoA thioester formation
of Na+-independent transporters, organic anion trans- of the unconjugated bile acids that enter the hepatocyte.
porting polypeptide (OATP), was identified, and several Steinberg et al. (256) characterized a homolog of a per-
of these multi-specific transporters have the ability to oxisomal very long chain CoA synthetase. This homo-
transport bile acids, as tabulated in cogent reviews by log was present in the smooth endoplasmic reticulum.
Hagenbuch and Stieger (249) and Klaassen and Alaskans They found that it had CoA synthetase activity for cholic
(250). In man, these include OATP1A2, OATP1B1, acid, and postulated a role for this enzyme in cholic acid
OATP1B3, and OATP1C1. The genes encoding OATP reconjugation.
transporters are denoted by the term solute carrier or- In the isolated perfused liver, rapid uptake of DCA oc-
ganic anion (SLCO). curs, presumably followed immediately by CoA formation.
To what extent OATP transporters contribute to the Formation of the adenylate and subsequent CoA thioester
highly efficient uptake of conjugated bile acids from sinu- serves to prevent back diffusion across the sinusoidal mem-
soidal plasma is not known, but it is believed that the ma- brane and thereby traps the bile acid CoA in the hepato-
jority of bile acid uptake is mediated by hepatocyte NTCP. cyte. In contrast, 24-norDCA, which does not form a CoA
The efficient uptake of dihydroxy conjugated bile acids is thioester, regurgitates back from the hepatocyte into the
truly remarkable, as these are 99% bound to albumin perfusate (257).
(251), and presumably uptake is from the extremely low The basolateral transporter, multidrug resistant protein
concentration (nanomolar) of unbound species. In our (MRP)4, has been shown to mediate the coefflux of conju-
view, it is reasonable to think that more than one trans- gated bile acids and reduced glutathione, in work from
porter may be involved in the sinusoidal uptake of conju- the laboratory of Dieter Keppler and colleagues (258). For
gated bile acids. Presumably, the transporters involved in this work, we provided tritium-labeled conjugated bile ac-
bile acid uptake have a zonal distribution and are enriched ids of high specific activity (259). Keppler (260) has sug-
in periportal hepatocytes. gested that conjugated bile acids percolate in and out of
During enterohepatic cycling, there is continuous the sinusoidal membrane, and that such efflux from the
bacterial deconjugation in the small intestine followed by hepatocyte into the space of Disse may serve to prevent
hepatic reconjugation, presumably occurring in the peri- toxic concentrations of bile acids in the hepatocyte.
portal hepatocytes. This was first recognized in rat stud-
ies by Norman, working in Lund, Sweden in 1955 (252), Canalicular secretion of bile acids. The next challenge was
and much later confirmed for man in our own studies at to identify the canalicular transporter or transporters.
the Mayo Clinic (253). The rate of “reconjugation” in the This had to be a powerful transporter, because the intra-
periportal cells is several fold greater than the rate of con- cellular concentration of bile acids was thought to be in
jugation of newly synthesized bile acids in the pericentral the low micromolar range, whereas the concentration of
cells. monomeric bile acids in ductal bile, based on the dialysis
The uptake of unconjugated bile acids is by both passive studies of Duane (261), was about 1,000 ␮M. There was
and carrier-mediated mechanisms, and depends on the thus an uphill transport of three orders of magnitude.
number and orientation of hydroxyl substituents. Litho- To study this transport system, one needed vesicles con-
cholic acid, CDCA, DCA, UDCA, and presumably 3,6-dihy- sisting predominantly of canalicular membranes. The first
droxy bile acids are highly membrane permeable (221, preparation was achieved by Inoue and Kinne, then work-
245), and in our judgment do not require a transporter ing in the laboratory of Irwin Arias in New York (262). The
for rapid uptake into the hepatocyte. Cholic acid, in con- canalicular membranes were isolated by nitrogen cavita-
trast, passes across cell membranes slowly and requires a tion and calcium precipitation. At about the same time,
transporter for rapid cellular entry. A systematic study of Peter Meier, working in the laboratory of James Boyer at
the passive membrane permeability of the three muricho- Yale, developed a method for preparing canalicular vesi-
lic acid epimers is lacking. cles using zonal flotation and high speed centrifugation
At least two transporters appear to be involved in the through discontinuous sucrose gradients (263). These
uptake of unconjugated trihydroxy bile acids. The labora- vesicles were used by Meier and his colleagues to show that
tory of Curt Klaassen in Kansas City showed that OATP1B2 taurocholate uptake by canalicular vesicles was carrier me-
(a sodium-independent transporter) appears to be in- diated and probably electrogenic (264). A similar study
volved in the uptake of unconjugated bile acids that re- was published by Inoue et al. (265).
turn from the intestine (254). Knockout of the transporter In 1986, Eamon O’Maille, who had studied bile acid se-
in mice caused a marked elevation in the plasma level of cretion in the intact dog, was working in our laboratory as
unconjugated bile acids. Hubbard et al. (255) showed that a guest investigator. He carefully measured the Tmax of tau-
fatty acid transport protein 5 (FATP5) knockout mice rocholate transport in the rat and concluded it was too
have a major fraction of unconjugated bile acids in bile, and great to be explained by membrane potential alone (266).
such unconjugated bile acids were enriched in secondary Y. Adachi returned to Japan from the Arias lab, and in

History of bile acid research 1569


1991 reported that taurocholate was actively transported The basolateral transporter of the ileal enterocyte. There was
into canalicular vesicles if an ATP generating system was one last carrier to be identified, the basolateral transporter
present (267). That same year, the Arias lab [Nishida of the ileal enterocyte. Also, if conjugated bile acids were
et al. (268)] also reported that rat canalicular membrane absorbed by cholangiocytes using ASBT and returned via
vesicles contain an ATP-dependent bile acid transport the periductular capillary plexus to the hepatocyte, there
system. had to be a basolateral transporter in cholangiocytes. The
A year or so later, the Zurich group began to work on hepatocyte did not require a basolateral efflux transporter,
the ATP-dependent process of conjugated bile acid extru- as it already had one in MRP4, and most bile acid efflux
sion by the canaliculus. They studied a variety of cell sys- from the hepatocyte was across the canalicular membrane
tems to find one which did not display ATP-stimulated bile into bile mediated by BSEP.
In 2001, Wei Wang, working with James Boyer and
acid transport and finally ended up with Sf9 (insect) cells.
Nazzareno Ballatori at the Mount Desert Island laboratory,
They noted a paper from the laboratory of Victor Ling in +
identified two genes encoding a Na -independent bile
Vancouver, Canada that showed a partial sequence of a
alcohol sulfate transporter in the skate (280). The genes
transport protein named “sister of P-glycoprotein,” later
were isolated using the Xenopus laevis technique of expres-
named multidrug resistance (MDR)1 (269). Using this
sion cloning, and they mediated transport only when they
sequence, an antibody to the putative canalicular trans-
were expressed simultaneously. They were named organic
porter was generated. The Zurich group then used phage solute transporters A and B (OST␣/OST␤). Subsequently,
libraries, their antibody, and Sf9 cells to monitor expres- Ballatori collaborated with the laboratory of Paul Dawson
sion. They were ultimately successful in cloning the BSEP and identified this dimeric organic solute transporter as the
(protein, BSEP; gene, Abcb11) (270). Our laboratory pro- long sought ileal enterocyte basolateral transporter (281).
vided 3H-labeled bile acids with high specific activity (259) The two proteins were present predominantly in ileum and
that were used to define its substrate affinity. kidney, and immunohistochemistry showed that the di-
It was not long until children with cholestatic liver dis- meric transporter was present on the basolateral membrane
ease were found to have mutations in Abcb11, the gene en- of the ileal enterocyte and renal proximal tubular cell.
coding BSEP (271). This disease had already been named When OST␣/OST␤ was expressed in polarized monolayers
progressive familial intrahepatic cholestasis type 2 (PFIC2), at the basolateral domain and ASBT was expressed at the
and as the defect is canalicular, markers of ductular injury, apical domain, bile acid transport from the apical side to
such as ␥-glutamyl-transferase, are not elevated in patients the basolateral side was observed (281). Further study of
with this genetic defect. The name PFIC2 is widely used, OST␣/OST␤ showed that it was a facilitated diffusion car-
but it seems preferable to call the disease BSEP deficiency. rier and not entirely specific for bile acids, in contrast to
There are other ABC-type canalicular transporters such as ASBT, which is highly specific for bile acids. In cholestasis,
MRP2, MDR1, and BCRP, but these are not believed to be hepatocyte OST␣/OST␤ is upregulated on the basolateral
very important in the canalicular transport of unsulfated (sinusoidal) membranes, and mediates bile acid efflux
bile acids in man, based on the severe cholestasis that oc- from the hepatocyte into sinusoidal plasma (282).
curs in patients with BSEP deficiency as well as evidence To date, no diseases caused by defective OST␣/OST␤
from in vitro studies of MRP2 (272). have been identified. A mouse knockout has been created,
The laboratory of Victor Ling, in Vancouver, reported and it differs from the ASBT knockout in that with the
the creation of an Abcb11 knockout mouse, which ap- OST␣/OST␤ knockout, there is no marked increase in
peared to have only mild cholestasis (273). The mildness bile acid biosynthesis despite bile acid malabsorption
of the mouse phenotype contrasted strikingly with the se- (283). In contrast, in the Slc10A2 (ASBT) knockout, bile
vere cholestatic phenotype that occurs in infants with a acid synthesis increases nearly 20-fold (284). This appar-
ently anomalous absence of a compensatory increase in
genetic defect. This striking difference between BSEP de-
bile acid synthesis is presumed to be the result of bile acids
ficiency in humans and that in mice appears to have at
accumulating in the ileal enterocyte and upregulating the
least three explanations. First, in mice, a major bile acid is
nuclear receptor FXR, which in turn stimulates the release
␤-muricholic acid, which is much less cytotoxic than DCA
of FGF15 into portal venous blood (285). Hepatocyte up-
and CDCA, these being major bile acids in man. Second,
take of FGF15 acts to downregulate bile acid synthesis, as
in mice, MDR1 is upregulated and is likely to be an effec-
noted in the Introduction. If there is no absorption of bile
tive conjugated bile acid extruder (274). Third, BSEP-defi- acids from the ileum, yet synthesis does not increase, the
cient mice form tetra-hydroxy and penta-hydroxy bile net result should be a bile acid deficiency in the proximal
acids (275, 276), some or all of which are likely to be trans- small intestine, as secretion will fall and become equal to
ported by MRP2 (277). Humans with cholestatic liver dis- synthesis. This in turn should cause malabsorption of satu-
ease do not form tetra- or penta-hydroxy bile acids to any rated fatty acids and fat soluble vitamins.
extent. In mice, the defect in bile acid export from the he-
patocyte is easily shown by feeding such mice cholic acid Imaging of bile acid transport. An 11C-labeled analog of
(278), which in this condition is highly toxic. Impaired taurocholate (cholyltaurine) has been synthesized by the
␤-fatty acid oxidation is also present in in such cholic acid- positron emission tomography scan center of Aarhus Uni-
11
fed animals (279). versity Hospital (286). The molecule, cholyl-N- CH3-glycine

1570 Journal of Lipid Research Volume 55, 2014


(cholylsarcosine) permits the kinetics of conjugated bile circulation of bile acids with values for man was presented
acid hepatic uptake and excretion into the biliary tract to by Sune Bergström in 1959 (8) and is shown in Fig. 8.
be measured and visualized in real time. Because of the Whether ancient vertebrates, e.g., the coelacanth and the
extremely short half-life of 11C (20 min), this compound hagfish, have an enterohepatic circulation with their C27
can be used only in positron emission tomography centers bile alcohol sulfates is not known.
that have a reactor (needed for the production of 11C)
close to the patient. The clinical utility of this molecule in Determinants, properties, and measurement of the enterohepatic
assessing cholestatic disease is under active investigation. circulation. As noted previously, the enterohepatic circu-
While this review was being written, a book entitled Hep- lation is the resultant of vectorial transport by the ileal en-
atobiliary Transport in Health and Disease was published terocyte and the hepatocyte, as well the flow in the portal
(287). It should cover the topics discussed in our review in venous system, the small intestine, and the biliary tract.
much more detail. The only molecules that have an enterohepatic circulation
are those that are transported by the membrane transport-
The enterohepatic circulation in health ers of the ileal enterocyte and the hepatocyte. Because
History. The enterohepatic cycling of bile was clearly only bile acids are transported by specific transporters,
described by Borelli in 1681 (288). He wrote: “Bile is col- only bile acids have an enterohepatic circulation. The pos-
lected in the portal vein [from the intestine] and sepa- sible enterohepatic circulation of drugs is beyond the scope
rated from the blood again in the liver. It is then sent into of this review.
the bile vessels. It leaves these and repeats its circuit The vast majority of the circulating bile acids are in the
though the intestine and the mesenteric veins again and biliary tract and intestine, because plasma and hepatocyte
again.” Liebig, a prominent German chemist who worked levels are very low. Plasma levels are low (<10 ␮M) because
in the mid-19th century, was fully aware of the enterohe- of efficient hepatic extraction as well as dilution. Urinary
patic circulation of some or all of the constituents of bile levels are also low (<10 ␮M) because only a fraction of
(289). Lehmann, in his text book of physiological chemis- plasma bile acids enter the glomerular filtrate, as bile acids
try published in 1853, wrote: “… the greater part of the are bound to plasma proteins, mostly to albumin (293).
bile is again resorbed as it passes through the intestine. Moreover, much of the fraction of bile acids that enters
The chief biliary constituents which are resorbed are the the glomerular filtrate is reabsorbed in the proximal tu-
soluble salts and cholic acid.” (290). Moritz Schiff, work- bules via ASBT and OST␣/␤. We found that cholyl conju-
ing in Florence, Italy, prepared anesthetized dogs with an gates were 70% bound, and dihydroxy conjugates were
“amphibolic” catheter. When bile was diverted to the out- >95% bound in human serum (251), so the glomerular
side, 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 anesthe-
tized 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 entero-
hepatic 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 contrac-
tion into the small intestine during digestion, is absorbed
from the distal intestine, returns to the liver, and may ei-
ther be stored in the gallbladder or resecreted into the
intestine. When a meal is ingested, the hormone cholecys-
tokinin is released from the small intestine. Cholecystoki-
nin induces gallbladder contraction as well as relaxation Fig. 8. Depiction of cholesterol and bile acid metabolism as well
of the valve (sphincter of Oddi) at the end of the common as the enterohepatic circulation by Bergström, Danielsson, and
bile duct where it empties into the small intestine. Bile Samuelsson in 1959 (8). In the accompanying text, they note that
then enters the duodenum. Some of the bile acids are ab- the mass of cholesterol entering the small intestine in bile is greater
sorbed in the jejunum, but most are transported by intesti- than that of dietary cholesterol. The figure shows neither the spill-
nal peristalsis to the distal ileum where they are efficiently over of absorbed bile acids into the systemic circulation nor se-
lective absorption of conjugated bile acids by the terminal ileum.
absorbed. The bile acid molecules pass through ileal en- Values for biliary bile acid secretion and fecal bile acid excre-
terocytes and enter portal venous blood to return to the tion are about 2-fold higher than those obtained in more recent
liver. One of the early illustrations of the enterohepatic studies.

History of bile acid research 1571


filtrate should contain mostly conjugates of cholic acid in positions (299). N. LaRusso, as his first research project,
humans. The end result is that the concentration of bile examined the stability of this label, and found it was good,
acids in urine in healthy humans is extremely low, and uri- but not perfect (300). For labeling CDCA and lithocholic
acid, we synthesized their ⌬ derivatives, and reduced
11
nary bile acids constitute a negligible fraction of bile acid
3
excretion. them with H. Again, this label was good, but not perfect
The mass of cycling bile acids in humans was first mea- (301). Finally, in San Diego, in work led by our chemist,
sured by Sven Lindstedt, a member of the Bergström Claudio Schteingart, we prepared bile acids with a double
group, then in Lund (14). Lindstedt prepared cholic acid bond between C-22 and C-23, and then reductively triti-
tagged with tritium and followed the decay of bile acid- ated these molecules (259). The 22-23 3H label was shown
specific activity (in bile) with time in human subjects. The to be completely stable during enterohepatic cycling in a
specific activity of the isolated cholic acid fell expo- collaborative study performed with W. Duane at the Univer-
nentially with time, indicating that loss of radioactivity sity of Minnesota (302). Isotope dilution studies have not
followed first order kinetics. The slope of the natural loga- been performed with ␤-muricholic acid because of the non-
availability of labeled material. However, the ⌬ compound,
22
rithm of the bile acid-specific activity decay curve can be
extrapolated to zero time to permit calculation of the pool the precursor for reductive tritiation, has been synthesized
size. The slope of the specific activity decay curve times the by the Iida group (137).
pool size gives the synthesis rate. The technique of isotope
dilution described by Lindstedt has been used to measure The enterohepatic circulation in humans. Because of bac-
the pool size and synthesis rate of the two primary bile ac- terial deconjugation in the intestine followed by absorp-
ids, cholic acid and CDCA, repeatedly (294). Such studies tion from the intestine and reconjugation in the liver, the
have shown that the rate of cholic acid synthesis is about turnover rate of the steroid moiety of C24 bile acids is con-
twice that of CDCA. siderably slower than that of its glycine moiety. As noted
The technique can also be performed with stable iso- previously, this deconjugation-reconjugation pathway was
topes, measuring the atoms percent excess in plasma bile identified in Sweden by A. Norman in rats (252), by Yamsaki
acids by MS, an approach developed by Franz Stellaard in Japan (303), and also somewhat later by clinical studies
and his colleagues in Groningen (295). When radioactive in the laboratory of the senior author at the Mayo Clinic
bile acids are used in the usual amounts, the specific activ- (253). Deconjugation-reconjugation also occurs with tau-
ity of plasma bile acids is too low to measure accurately. rine-conjugated bile acids, but the difference in turnover
The isotope dilution technique has also been used for rates between the steroid and amino acid moiety is less,
DCA (296), but input now represents absorption of newly because taurine conjugates are deconjugated more slowly
formed DCA rather than synthesis. The input of DCA is than glycine conjugates (162). These studies, performed
some fraction of cholic acid synthesis, and could therefore at the Mayo Clinic in the laboratory of the senior author,
vary from 0 to nearly 1.0. This fraction, DCA input/cholic have never been repeated, so confirmation of their find-
acid synthesis, has been termed the fdehydrox and ranges in ings is desirable.
healthy adults from 0.2 to 0.5 (297). No information is available on the turnover rates of ei-
In the past, there has been concern because values for ther the steroid moiety or the taurine moiety of C27 bile
bile acid synthesis obtained by isotope dilution were often acids. For most C27 bile alcohols, the process of deconjuga-
somewhat higher than those obtained by analysis of fecal tion-reconjugation is unlikely, as the liberated bile alco-
bile acids. The belief, at present, is that the discrepancy hol, in contrast to that of unconjugated bile acids, is too
between these two methods, which should give identical polar to be passively absorbed (190).
results, is attributable to the difficulty of analyzing all fecal Early workers measured the bile acid pool by isotope dilu-
bile acids. Thus some fecal bile acids are not measured, tion and guessed at the number of times that the pool cy-
and a higher result for bile acid synthesis is obtained by cled, the product being bile acid secretion. In our view, this
the isotope dilution technique. Also, if a considerable vol- assumption is not valid, and is analogous to trying to predict
ume of bile is removed during the daily duodenal bile sam- cardiac output from the measurement of blood volume. A
pling used in the isotope dilution technique; this will flow can only be estimated by indicator dilution techniques.
increase bile acid synthesis. Workers at the Mayo Clinic used such an indicator dilution
No isotope dilution studies have been performed with technique to measure bile acid secretion and found that it
C27 bile acids or bile alcohols. averaged about 12 g/day, rising during meal digestion and
falling during the interprandial interval, as well as during
Synthesis of labeled bile acids for metabolic studies. In the overnight fasting (304, 305). Bile acid secretion during di-
laboratory of the senior author at the Mayo Clinic, better gestion was about 0.3 ␮mol/kg/min, a value confirmed in a
radioactive tags for bile acids were sought in order to char- separate study of gallbladder emptying by G. Van Berge He-
acterize bile acid metabolism. The Bergström group was negouwen and colleagues, also at the Mayo Clinic (306).
the first to synthesize 24-14C-tagged bile acids (298), and Bile acid secretion was also quantified in another intestinal
this label was stable during enterohepatic cycling. In order perfusion study by T. Northfield and the senior author, and
3
to prepare H-tagged bile acids that would also be stable similar results were obtained (307).
during enterohepatic cycling, we used a method devel- Portal venous blood delivers bile acids to the hepatic si-
oped by Peter Klein to exchange 3H into the C-2 and C-4 nusoids, and bile acids are efficiently extracted, first pass

1572 Journal of Lipid Research Volume 55, 2014


fractional extraction being 50–90%, depending on the underwent cholehepatic shunting, and induced a bicar-
bile acid. C. Einarsson, B. Angelin, and their colleagues bonate-rich hypercholeresis. This report from the Erlinger
performed experiments in which they measured bile acid laboratory gave rise to the erroneous belief that UDCA
concentrations in portal venous blood and hepatic venous had unique choleretic properties. In cholestatic disease,
blood in patients at surgery, and thus could directly mea- most patients appear to conjugate their ingested UDCA
sure hepatic extraction (308). M. Korman and his col- completely based on analyses of biliary bile acids (316),
leagues found that hepatic extraction of glycocholate was but if conjugation were incomplete and unconjugated
identical during both the fasting and postprandial state UDCA were secreted into bile, it would most likely be ab-
(309). This constancy of efficient hepatic extraction means sorbed in the biliary ductules, thereby undergoing chole-
that hepatic extraction is bloodflow limited, and this view hepatic shunting. In further work from our laboratory, we
was confirmed in a careful study of hepatic bile acid ex- showed that sulindac, but not other NSAIDs, also under-
traction in the dog from the laboratory of R. Hanson went cholehepatic shunting (317).
(310). First pass clearance for CDCA in healthy volunteers
was determined by van Berge Henegouwen and colleagues Determinants of plasma bile acids. The plasma level of bile
(311). They compared the area under the curve (AUC) acids is determined by the instantaneous rates of intestinal
after intravenous administration with the AUC after oral absorption and hepatic uptake. Dogs with a chronic biliary
administration. A value of 62% was obtained. Unconju- fistula have no detectable bile acids in their systemic ve-
gated UDCA has a first pass extraction of about 50% (312). nous blood, thus confirming that intestinal absorption is
In general, dihydroxy conjugates, i.e., the conjugates of the source of plasma bile acids, at least in health (318).
DCA and CDCA, have a lower first pass extraction than The profile of plasma bile acids is not identical to the pro-
cholyl conjugates, perhaps because the dihydroxy conju- file of biliary bile acids. Plasma bile acids are enriched
gates are bound much more tightly by albumin. For a in dihydroxy conjugates because of their lower hepatic
given steroid moiety, conjugated bile acids are extracted extraction.
more efficiently than the unconjugated bile acid.
Regulation of the enterohepatic circulation. The enterohe-
Absorption of bile acids in the biliary ductules, cholehepatic patic circulation of bile acids must balance two conflicting
shunting. The discovery that cholangiocytes contain physiological functions. On the one hand, bile acids must
ASBT (238, 239) meant that a fraction of conjugated bile serve as the end products of cholesterol metabolism and
acids secreted into canalicular bile is absorbed during pas- excretion is necessary for cholesterol balance. For this
sage through the biliary ductules. The biliary ductules are function, enterohepatic cycling is unimportant. On the
nurtured by a periductular capillary plexus that empties other hand, the ileal conservation of bile acids is needed
into the portal region of the hepatic sinusoids. If conjuga- to provide ample bile acids for efficient lipid digestion,
tion is not complete, unconjugated bile acids will enter and bile acid excretion is unimportant. Given these con-
into canalicular bile. In addition, we found in our labora- flicting aims, it should be highly likely that the enterohe-
tory that 24-nor C23 dihydroxy bile acids (norCDCA, nor- patic circulation of bile acids would be tightly regulated.
DCA, and norUDCA), which are inefficiently conjugated, The idea of regulation of the enterohepatic circulation
are absorbed passively from the biliary ductules (163, 164). is not new. Whipple, in his 1922 review (91), noted that
The absorbed molecules return to the hepatocyte and are there is little increase in total bile acids in dogs ingesting
once again resecreted into canalicular bile, thus undergo- bile acids chronically, and suggested that absorption must
ing a cholehepatic circulation. The absorption of the un- be regulated. Sten Eriksson, working in Lund, observed
conjugated bile acid removes a proton from the canalicular that bile acid synthesis increased with time in the biliary
bile, generating a bicarbonate anion, the result being no fistula rat (17). Thus the working hypothesis was that bile
change in bile osmolarity. Lamri et al. (313) provided acid synthesis is determined by the hepatocyte concen-
additional evidence for the validity of cholehepatic shunt- tration of bile acids. When this falls, bile acid synthesis
ing using immunohistochemical techniques. The existence increases. When bile acids are fed, bile acid synthesis de-
of a “cholehepatic shunt” for unconjugated mono- and creases. The senior investigator characterized bile acid
dihydroxy bile acids has now been accepted by most metabolism in patients with ileal resection. A marked in-
hepatologists. crease, up to 10-fold, was present in such patients, again
When the unconjugated bile acid returns to the sinu- attributed to bile acid malabsorption lowering the concen-
soid, it is taken up by the hepatocyte and resecreted into tration of bile acids in the hepatocyte (319). A similar in-
bile, thereby inducing additional canalicular bile flow, as crease in bile acid synthesis was induced by cholestyramine
evidenced by increased mannitol clearance. Mannitol administration (320). Studies in the rhesus monkey, with
clearance is considered a measure of canalicular secretion an exteriorized enterohepatic circulation of bile acids,
(314). The increase in bile flow enriched in bicarbonate is also provided convincing evidence that bile acid synthesis
termed “hypercholeresis”. Hypercholeresis was originally was regulated in a negative feedback manner (321). In
described by the laboratory of Serge Erlinger (315). In these studies performed by R. H. Dowling, D. Small, and E.
their experiments in biliary fistula rats, UDCA was infused Mack (the surgeon who prepared the monkeys), the re-
at a rate exceeding the hepatocyte’s capacity to conjugate turn of bile acids to the liver could be systematically de-
it. As a result, unconjugated UDCA was secreted into bile, creased, and an increase in synthesis quantified.

History of bile acid research 1573


However, two observations did not seem to support this present. (To date, such a defect has not been identified.)
useful hypothesis. The first was that bile acid synthesis in- Bacterial overgrowth can be present, leading to deconju-
creased, at least acutely, when the bile duct was ligated gation, and sometimes, to dehydroxylation. Unconjugated
(115). The second, made independently by the group of bile acids are rapidly absorbed, leading to an intralumi-
Reno Vlahcevic in Richmond, VA (322) and the group of nal bile acid deficiency, which in turn can cause lipid
Chiijiwa in Japan (323), was that upregulated synthesis in malabsorption.
the biliary fistula rat could be returned to normal levels by Return of bile acids from the small intestine can be
intestinal, but not by intravenous infusion of bile acids. caused by defects in ileal transport, either at the apical or
Both types of infusion should lead to an increase in the basolateral domains of the ileal enterocyte, or by absence
intracellular concentration of bile acids. The Richmond of the ileum. In addition, if rapid intestinal transport pro-
group proposed that a blood-borne signal from the intes- pels bile acids past the terminal ileum at a rate exceeding
tine to the liver might downregulate bile acid synthesis. its absorptive capacity, spillover into the colon will occur,
The breakthrough came with the identification of FGF15 and such may induce diarrhea. Portal venous bloodflow
(in the rat) and FGF19 (in humans) as a protein released by may be obstructed or portal-caval shunting may occur. Fi-
the ileal enterocyte in response to FXR induction by bile nally hepatocyte uptake, transport through the hepatocyte,
acids. This observation by Inagaki and Kliewer and col- or canalicular secretion may be impaired. It is beyond the
leagues at University of Texas Southwestern (58) was a ma- scope of this historical review to deal with each of these
jor advance in our understanding of the regulation of the perturbations on bile acid metabolism.
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; and conservation, PHYSICOCHEMICAL ASPECTS OF BILE ACIDS
i.e., ileal transport is regulated at the level of the ileal en-
terocyte. Both synthesis as well as ileal transport are down- Overview
regulated by FGF19 (53). FGF19 enters the hepatocyte via The physical chemistry of biliary lipids and digestive lip-
a receptor [fibroblast growth factor receptor 4 (FGFR4)] ids are similar, in that both involve solubilization of polar
and directly or indirectly downregulates bile acid synthe- lipids in the form of mixed micelles. Mixed micelle forma-
sis. Synthesis of FGF19 in the ileal enterocyte is under the tion, whether during biliary lipid secretion or triglyceride
control of FXR, the nuclear receptor activated by bile ac- digestion, has received a great deal of attention from phy-
ids. Nonetheless, in some species, it remains possible that sicians working together with physical chemists in an at-
bile acid synthesis is regulated by the bile acid concentra- tempt to unravel the complexities of these processes.
tion in the hepatocyte rather than by FGF19 (324). In 1983, the Kroc Foundation sponsored a small confer-
The regulation of ileal conservation is poorly under- ence (9 physicians, 15 chemists) entitled, “The Physical
stood. J. Lillienau, working with the senior author, found Chemistry of Bile in Health and Disease”. Many of the top-
that bile salt feeding in the guinea pig downregulated ileal ics discussed below are treated in more detail in the pub-
transport, whereas administration of a bile acid seques- lished proceedings of that meeting (328). Sadly, it was to
trant upregulated ileal transport (325). Upregulation of be the last Kroc Foundation conference. Ray Kroc, patron
ileal bile acid transport was associated with recruitment of of the foundation (and founder of McDonald’s, a global
more proximal enterocytes to transport bile acids. None- fast food chain) died shortly thereafter, and his widow,
theless, with complete cholestasis or with parenteral ali- Joan Kroc, had other priorities for her generous philan-
mentation, bile acid transport by the ileal enterocyte thropy. The foundation was dissolved.
decreases (326, 327). Thus, there must be multiple mecha- In 1966, the senior author collaborated with Donald
nisms of control. It is likely that the downregulation of the Small in the writing of a brief review on the physicochemi-
ileal bile acid transporter (ASBT) with bile acid feeding cal properties of bile acids (329). In 1985, Martin Carey
involves FXR agonism and FGF19 release. authored a systematic review of the physicochemical prop-
erties of C24 bile acids (330). His review included tabula-
Perturbations of the enterohepatic circulation in disease tion of physical constants, crystal structure as determined
Perturbations of the enterohepatic circulation result by X-ray diffraction, surface chemistry of bile acids, pH
from defects in bile acid transport or flow, assuming that solubility profiles, ionization behavior, and solubilization
bile acid synthesis and conjugation are normal. Bile flow properties of bile acid solutions. Together, these two re-
begins at the canaliculus and there can be ductular ob- views summarized existing knowledge of the physicochem-
struction, as occurs in cholestatic liver disease such as pri- ical properties of bile acids, as envisioned by medically
mary biliary cirrhosis. Physical obstruction can occur with trained physician-scientists. In the discussion below, only
choledocholithiasis or cholangiocarcinoma. Bile flow can selected aspects of the physicochemical properties are
be diverted to the outside, as in patients with partial or considered.
complete biliary fistula. Bile acids can regurgitate into the
stomach and from there into the esophagus. In the small Bile acids in aqueous solution
intestine, conjugated bile acids might leak across the Solubility of the protonated acid. The first detailed studies
epithelium if increased paracellular permeability were on the solubility of protonated unconjugated bile acids

1574 Journal of Lipid Research Volume 55, 2014


were performed by A. Roda and A. Fini at the University of The physicochemical significance of conjugation is that
Bologna in Italy (331). They reported solubility values at it increases solubility markedly at intestinal pH. Dihydroxy
pH 3 for 10 common bile acids. Values varied enormously. bile acids, which constitute 60% of human bile acids, are
That of lithocholic acid was 0.05 ␮M whereas that of the insoluble at the pH conditions prevailing in the small in-
7␤ epimer of cholic acid (so called “ursocholic” acid) was testine during digestion (pH 6–7). In vitro, the solubility
1,670 ␮M. Another observation of clinical relevance was of the bile acid ion increases exponentially with increasing
that the two bile acids used for gallstone dissolution had pH. When the concentration of bile acid reaches the criti-
quite different aqueous solubilities: that of CDCA was cal micellization concentration (CMC), solubility increases
27 ␮M, whereas that of UDCA was 9 ␮M. The greater markedly, in that a crystalline excess becomes transformed
aqueous solubility of CDCA contributes to its excellent into micelles. There is thus a “critical micellization pH”
gastrointestinal absorption (210); UDCA, with a lower mo- (CMpH) which defines an approximate concentration
nomeric solubility, is absorbed less efficiently and its crys- above which the bile acid is highly soluble and below which
tal dissolution is rate limiting in its absorption (332). the bile acid is insoluble (339). Lillienau, Schteingart, and
The solubility of several protonated glycine-conjugated Hofmann (338), working in our laboratory, found the
bile acids has also been determined by A. Roda and his CMpH of glycocholate to be 4.8. That of CDCA was found
colleagues. They are quite low (A. Roda, personal observa- by van Berge Henegouwen to be about 6.8 (340) and that
tion). Taurine-conjugated bile acids, in contrast, even in of UDCA has been calculated to be 7.9 (339). The con-
the acid form, are water soluble. verse of CMpH has been termed “the pH of precipitation”
As yet, we have no solubility values for any of the uncon- (333) and denotes the pH at which bile acids begin to pre-
jugated C27 bile acids. The solubility of 5␣-cyprinol was cipitate from a true solution of the bile acid, the value be-
found to be 360 ␮M, thus being similar to that of cholic ing dependent on the bile acid concentration. The matter
acid (190). No other solubility data are available for C27 is discussed extensively in the review of Donald Small
bile alcohols. which gives values for the pH of precipitation of the CDC,
DCA, and cholic acid (333).
Ionization. One of the purposes of the Kroc Founda- K. J. Mysels derived the following equation: CMpH =
tion meeting was to reach a consensus on the pKa values of log[CMC] + pKa + BApS, where BApS is the negative loga-
bile acids, as titration studies performed by D. Small (333) rithm of the aqueous solubility of the protonated mono-
and H. Igimi and M. C. Carey (334) gave conflicting re- mer. The high CMpH for UDCA is thus explained by its
sults, suggesting that the pKa of an unconjugated bile acid was low aqueous solubility and a relatively high CMC.
influenced by the number of hydroxyl groups. B. Josephson, Despite their excellent pH-solubility profile, glycine-
working in Stockholm in the 1930s as a predoctoral stu- conjugated bile acids will precipitate in vivo if the pH is
dent, determined that taurine-conjugated bile acids were sufficiently acidic. This occurs in patients with gastrin-se-
strong acids, that glycine conjugates had a pKa of 4–5, and creting tumors in whom markedly increased gastric acid
that unconjugated bile acids had a pKa of about 6 (335). A secretion overwhelms pancreatic bicarbonate secretion
similar conclusion for unconjugated bile acids was reached and the small intestinal content becomes acidic (341). In
by the laboratory of Per Ekwall working in Turku, Finland the porcupine, glycine-conjugated bile acids reflux from
(336). the duodenum into the stomach and precipitate, forming
At the Kroc meeting, Aldo Roda presented work per- gastric bezoars (342).
formed by him and A. Fini which gave accurate pKa values
for unconjugated bile acids (331). Roda used a technique Solubility of calcium (Ca2+) salts of bile acids. Conjugation
previously used to determine the pKa of long chain fatty also greatly increases the resistance to precipitation by di-
2+
acids. One measures the apparent pKa in mixtures of in- valent cations such as Ca . Studies from our laboratory
creasing proportions of water in methanol and extrapo- (343) in collaboration with K. Mysels (a legendary physical
lates to 100% water. The values reported were about 5.1 chemist) showed that the calcium salts of taurine conju-
for the common natural unconjugated bile acids with no gates of common natural bile acids are water soluble. Gly-
substituents on the side chain. Later, Roda and Fini ob- cine conjugation has a very small effect on the ion product
2+ 2 2+
tained values of about 4 for glycine conjugates (337). The of Ca ion × [BA-] , and the solubility products of Ca
pKa of glycine conjugates is lower than that of unconju- salts of glycine-conjugated bile acids are quite low. How-
gated bile acids, because the electron withdrawal effect of ever, the supersaturated solutions of the calcium salts of
the amide bond enhances the loss of a proton from the glycine-conjugated bile acids are metastable, resisting for-
carboxyl group (338). The pKa of cholylsarcosine was de- mation of the insoluble Ca2+ salt for many weeks. The solu-
2+
termined by our laboratory to be 3.7 (338), about 0.3 pKa bilities of Ca salts of unconjugated bile acids, like those
units lower than that of cholylglycine. of glycine-conjugated bile acids, are also quite low (344).
For unconjugated bile acids, the pKa can be reduced by However, metastability of supersaturated solutions is not
electronegative substituents on the side chain. Roda et al. observed with unconjugated bile acids.
(135) found the pKa of 23R-hydroxy-CDCA to be 4.8, In the enterohepatic axis, the most dangerous site for
about 0.2 units lower than that of CDCA. It should be pos- possible insoluble calcium salt formation is the gallblad-
sible to lower the pKa of unconjugated bile acids considerably der, where ionized Ca2+ concentrations rise during the
by adding one or two fluorine atoms to the C-23 carbon. concentration of bile by the gallbladder epithelium (345).

History of bile acid research 1575


Stone formation remains rare, as the concentration of ion- the CMT of sodium taurodeoxycholate:sodium palmitate
ized Ca2+ is kept rather low in the biliary tract and small mixtures with a progressive increase in the mole fraction
2+
intestine. The low concentration of ionized Ca is ex- of sodium taurodeoxycholate. One can then estimate the
+
plained by the high Na concentration, which decreases CMT value of pure sodium taurodeoxycholate by extrapo-
the activity coefficient of calcium (339). lation. The extrapolated values were well below 0°C (350).
Formation of Ca2+ bile salt gallstones occurs when “cal- Thus, because of their solubility at 0°C, the common natu-
cium-sensitive” bile acids are present in appreciable pro- ral polyhydroxy C24 bile acids may be considered excellent
portions in biliary bile acids. In general, this has occurred “cold water detergents”.
because of the feeding of unusual bile acids or their pre- The influence of the A/B ring juncture on the CMT of
cursors (346). Examples are the feeding of cholestanol C24 bile acids has not been defined. There is also no in-
(the saturated derivative of cholesterol with an A/B trans formation on the CMT values of C27 bile acids, as well as
structure) to the rabbit. The saturated sterol is converted those of bile alcohol sulfates, with the single exception of
to allocholic acid, which in turn is converted to allodeoxy- 5␣-cyprinol sulfate, whose CMT was reported to be <0°C
cholic acid by intestinal bacteria. The allodeoxycholic acid (190).
is conjugated with glycine and the insoluble glycine conju-
gate (glycoallodeoxycholate) precipitates from solution Self-association in solution: micelle formation. In 1936, G.
forming concretions in the gallbladder (107). Other instances S. Hartley, working in London, wrote a small lucid mono-
of the formation of insoluble Ca2+ salts are exemplified by graph entitled Aqueous Solutions of Paraffin-Chain Salts: A
the feeding of lithocholic acid to the taurine-deficient rat Study in Micelle Formation (351). This work summarized the
(347, 348), as well as the feeding of murideoxycholic acid evidence, mostly based on conductivity measurements, for
to the prairie dog (349). micelle formation by long chain fatty acid anions as well as
The biological significance of conjugation is that it en- alkyl sulfates, and proposed a structure for the spherical
hances ionization at the pH present in jejunal content micelle. A. S. C. Lawrence, a colleague of Hartley’s once
during digestion, preventing passive absorption across the said to the senior author: “Hartley wrote down what every-
apical membrane of the enterocyte. The bile acid mole- one else was thinking!” Hartley notes in the final chapter:
cule, whether unconjugated or conjugated, is too large to “The bile salts are obviously amphipathic.” At the end of
traverse the paracellular junctions of the healthy intes- the Second World War, Hartley returned to academic life
tine. This “nonabsorbability” of conjugated bile acids per- and decided to take up the problem of micelle formation
mits high concentrations of conjugated bile acids to be in bile acid solutions. However, his postdoctoral student
maintained in the biliary tract and proximal small intes- developed mental problems, and the work stopped. Hart-
tine where the pH ranges from pH 6 to pH 7, and allows ley himself left the academic world and became a scientist
them to function as biological solubilizers. Conjugation is
at Fisons, where he invented novel sprayers for crops, as
also likely to prevent bile acids from partitioning into the
well as developing new pesticides.
endoplasmic reticulum during their transit through the
The concept of micelle formation had been first ad-
hepatocyte.
vanced by James McBain in 1913, who presented this idea
Temperature effects on solubility. Temperature effects are at a meeting of the Royal Society. The chairman listened
not very important in the biology of bile acids. Solutions of carefully, and began the discussion with, “Nonsense,
the common primary bile acids are stable at 0°C. Friedrich McBain.” McBain, who spent the latter part of his scientific
Krafft, working in Heidelberg, Germany, observed that career at Stanford, spent much of his subsequent years
turbid dispersions of soaps became a clear solution over a characterizing soap solutions, and undertook limited stud-
very narrow temperature range, and his observation has ies with bile acid solutions. McBain also noted in 1941 that
been immortalized in the term “Krafft point”, which de- detergent solutions can solubilize insoluble dyes. His
notes this temperature of a phase transition. One defini- widow and E. Hutchinson published a book entitled Solu-
tion of the Krafft point is the temperature at which the bilization and Related Phenomena in 1955, which summarized
concentration of the bile acid monomer reaches the CMC. many of the observations on this topic in the McBain labo-
With a further increase in temperature, the excess solid ratory (352). J. Bashour and L. Bauman, working in New
phase disappears and the suspension is transformed into a York, developed a method for synthesizing conjugated
clear micellar solution. A thorough discussion of the Krafft bile acids, and in 1937 used their synthetically prepared
point can be found in the monograph of Laughlin (350). conjugates to show that bile acid solutions solubilize cho-
The term “critical micellization temperature” (CMT) lesterol in a concentration-dependent manner (353).
was proposed, but has not been widely adapted. Sodium In 1942, O. Mellander and E. Stenhagen, working in
cholanoate (no hydroxyl groups) and most mono-hydroxyl Sweden, performed conductivity studies with sodium tau-
bile acids (including lithocholic acid) have high CMT val- rocholate and found evidence of aggregate formation
ues, well above body temperature. (354). Per Ekwall, working in Turku, Finland, summarized
In contrast, primary dihydroxy and trihydroxy bile acid the evidence for micelle formation in sodium cholate solu-
conjugates have very low CMT values. An attempt was tions in 1951 (355). Ekwall also characterized surface
made by the senior author to define the CMT values of properties of bile acids and made many contributions to
sodium taurodeoxycholate. This was done by measuring the physical chemistry of bile acid solutions (356).

1576 Journal of Lipid Research Volume 55, 2014


As noted above, a program on the biology and chemis- samples. He obtained values for the CMC as well as for the
try of bile acids was initiated by Sune Bergström in Lund, intermicellar concentration of bile acids, which were quite
Sweden after the war. Arne Norman synthesized the gly- similar to the CMC in bile acid-monoglyceride systems
cine and taurine conjugates of most of the major bile ac- (261).
ids. He then collaborated with Per Ekwall to study the
solubilization of 20-methylcholanthrene and thus define Solubilization of polar lipids by bile acid micelles
the CMC for dilute solutions of pure conjugated bile acids Physical chemistry of biliary lipids. Bile acids are present
(357). Shortly thereafter, and independently, the senior at micellar concentrations only in bile and small intestinal
author, working in Lund, Sweden, synthesized and mea- content. In humans, the concentrations in plasma (2–10
sured the CMC of the major bile acids present in human ␮M) and in colonic content (400 ␮M) are well below the
bile using trans-azobenzene as the solubilizate (358). CMC. In health, bile acids are always associated with a po-
In contrast to typical ionic surfactants, the aggregation lar lipid. In bile, this is predominantly phosphatidylcho-
of bile acid anions is gradual, and there is no well-defined line (PC). In small intestinal content, during digestion,
CMC. Thus, the CMC for bile acid solutions is often taken the polar lipids are monoacyl glycerol and fatty acids
as the midpoint of a small concentration range over which (partly ionized). The only circumstance in which bile acid
micelles form. K. J. Mysels suggested the term “uncritical micelles are present without an accompanying polar lipid
micellization concentration.” P. Mukerjee has pointed out is in the Mdr2 knockout mouse and in patients with a de-
that bile acids are rigid molecules in contrast to typical fective Mdr3 protein. Such mice (and patients) lack this
ionic detergents which have a flexible alkyl chain. There- ATP-activated PC extruder in the canaliculus of the hepa-
fore, bile acid molecules, just as dye molecules, stack tocyte and are believed to have essentially no phospho-
rather than form spherical micelles. The aggregates (from lipid in ductular bile. The micellar bile acids, if sufficiently
tetramers up to 20-mers, depending on bile acid structure hydrophobic, attack the membranes of the cholangiocytes,
and concentration) may be termed multimers rather than leading to ductal inflammation in mice (362) and to a
micelles. The size of the aggregates was estimated by light variety of biliary problems in patients (363). Nonetheless,
scattering, especially by D. Small (329, 333) and J. Krahtohvil the bile of many vertebrate species has an extremely low
(359). However, as noted below, aggregates of bile acid ratio of phospholipids to bile acids (181) without any sign
molecules alone, i.e., without an accompanying lipid, do of biliary ductule injury. In such species, it may be that
not occur in nature, except when phospholipid extrusion phospholipid, after secretion into canalicular bile, is ab-
into bile is defective. sorbed in the biliary ductules with the result that it is not
The most recent measurement of CMC values for a present in gallbladder bile in appreciable proportions, or
large number of unconjugated and conjugated bile acids that the ductules are highly resistant to attack by bile acids
was made by Aldo Roda in our laboratory, in collaboration or both.
with K. J. Mysels (360). This study was successful because The recognition that lecithin was present in bile oc-
of two major advances. The first was the synthesis of a large curred early in the 20th century. G. Whipple in his 1922
number of bile acid epimers by Takashi Iida and Frederick review (91) tabulated the reported analyses of biliary leci-
Chang (361), and in a few cases, by our laboratory. The thin. In 1952, Polonovski and Bourillon, working in Paris,
second was the availability of a maximum bubble pressure confirmed that the dominant phospholipid in bile was
device designed, constructed, and characterized by K. J. lecithin, later to be named PC (364). G. Phillips separated
Mysels. A key aspect of this technique for measuring sur- biliary phospholipids chromatographically and confirmed
face tension in relation to concentration is that a new bub- that the dominant phospholipid in bile was PC (365). An-
ble surface is constantly being formed whose composition other article in this series will give the history of phospho-
consists of the dominant constituent of the solution. In lipids in more detail.
contrast, with surface tension methods using a tensiome- Franz Ingelfinger, one of the dominant figures in Amer-
ter, a more surface-active impurity may concentrate at the ican Gastroenterology in the mid 1900s, was unhappy with
surface of the solution to be analyzed and lead to errone- the level of ignorance of the pathogenesis of the common
ous values for the CMC. Roda validated his bubble pres- disease of gallstones. He arranged for Donald Small, one
sure values by showing that identical values were obtained of his brightest fellows, to spend two years in Paris working
with the technique of dye solubilization using Orange OT under the direction of a scholarly colloid chemist, Digren
(1-O-tolyl-azo-2-naphthol) as the water-insoluble dye. Val- Dervichian. During those two years, the behavior of model
ues are reported in the absence and presence of 0.15 M systems simulating bile was described clearly using triangu-
Na+ concentration. lar coordinates. The ternary systems, water-lecithin-bile
The senior author, when working in Sweden in the acid, water-cholesterol-bile acid, and water-cholesterol-
1960s observed that the aggregation of monoglycerides lecithin, were carefully defined using sodium cholate, egg
and bile acids to form mixed micelles was cooperative, so lecithin, and cholesterol. The behavior of the three lipids
that solubilization occurred at a concentration well below and water could then be described by a tetrahedron. A
the CMC of bile acids in the absence of such amphiphilic slice across the tetrahedron at 10% total lipid concentra-
lipids (358). W. Duane used dialysis to measure the inter- tion gave a triangle with three coordinates, cholesterol,
micellar concentration of bile acids (monomers plus sim- lecithin (PC), and bile acid. A line defined the limits of the
ple micelles) in both model bile samples and human bile micellar zone. Accordingly, points lying above the line

History of bile acid research 1577


would be supersaturated in cholesterol, either in liquid with a drum shaped perimeter was coated on its surface
crystalline or crystalline form. This classic work is sum- with bile acid molecules (369). Later, dimers of bile acids
marized in (333) and illustrated in Fig. 9. The stage was interdigitating between the PC bilayer were added (330).
set for applying this pure exercise in model systems to The model was widely accepted.
the pathogenesis of cholesterol gallstone formation (see At the Kroc meeting in 1984, G. Lindblom of the Uni-
later). versity of Lund proposed that bile acids could also interact
That phospholipids play a key role in the solubilization with phospholipid bilayers by lying flat in the bilayer with
of cholesterol in bile was perhaps first recognized in the the hydrophobic face of the bile acid molecule pushing
mid 1950s by B. Isaksson, working in Sweden (366). Work the charged heads of PC apart (370). W. Nichols and J.
on the solubilization of lecithin by bile acid solutions was Ozarowski proposed that bile acids lie flat on the surface
carefully studied by S. Yoshimuta (367), a member of the of worm-shaped micelles (371), in contrast to the mixed
Department of Surgery at Kyushu University in Japan whose disc model. Rex Hjelm, a biophysicist working at the Los
director, E. Miyake, had a longstanding interest in gallstone Alamos National Laboratory, examined bile acid-PC-water
formation. This work was published in the 1960s, about the systems using small angle neutron scattering. Hjelm con-
same time that D. Small was studying model systems simulat- cluded that as the PC/bile salt ratio increased, the change
ing bile in Paris. Another physician with interest in this in scattering indicated that PC-bile acid micelles increase
problem was Kerrison Juniper who attributed cholesterol in size by the elongation of constant diameter rods (372),
solubilization in bile to a macromolecular complex (368). providing additional support for the “radial shell model”
A review by D. Small and the senior author, published in structure advanced by Nichols and Ozarowski. Hjelm also
1967, advanced the idea that the physical chemistry of mi- concluded that the disk-shaped micelle originally pro-
celle formation in bile and micelle formation in small in-
posed by Small et al. (369) could not be correct (372).
testinal content during triglyceride digestion were similar
Nonetheless, the conclusions of Hjelm are not widely
physicochemical processes (329).
known in the medical community, because much of his
Thus by 1967, both bile and small intestinal content
work was published in chemical journals that are not in-
were considered micellar solutions, the only difference be-
cluded in the PubMed database. A depiction of the origi-
ing the nature of the accompanying lipids. Simple bile
nal model for the mixed micelle proposed by Small and
acid micelles are poor solubilizers for cholesterol or non-
Carey (330, 369) is compared with that of the model in-
polar dietary lipids. The mixed micelle provides a hydro-
ferred from the small angle neuron studies of Hjelm and
carbon interior in which poorly soluble lipids can dissolve.
In bile, this insoluble lipid is cholesterol; in small intesti- colleagues in Fig. 10.
nal content, insoluble lipids of physiological importance In 2002, S. Marrink and A. Mark, working in the Nether-
are the fat soluble vitamins. The remaining vexing prob- lands, used the technique of molecular dynamic simula-
lem was the molecular arrangement of the mixed micelles tions, in which molecules aggregate in water to give the
present in bile and small intestinal content. lowest free energy (373). They concluded that: “… phos-
pholipids are packed radially with their headgroups at the
Molecular arrangement of mixed micelles. To solve the surface and the hydrophobic tales [sic] point … toward
problem of the structural arrangement of the mixed mi- the micellar center. The bile salts act as wedges between
celles in bile, D. Small traveled to England to the labora- the phospholipid headgroups.” They concluded that the
tory of Dennis Chapman (F.R.S.), a physical chemist with structure of the micelles generated in their system re-
a deep interest in membrane structure. They examined semble the radial shell model proposed by Nichols and
solutions by NMR and found that the angular carbons of Hjelm. Additional work by Long, Kaler, and Lee (374) us-
the bile salt molecule were in a hydrophobic environment. ing small angle X-ray scattering and quasi-elastic light scat-
On the basis of the NMR spectra, they proposed the tering were also in agreement with the model proposed by
“mixed disc” model in which a bilayer of PC molecules Nichols.

Fig. 9. Depiction of the use of triangular coordi-


nates 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
indicating a constant water composition (10% sol-
ids, 90% water) in the tetrahedron consisting of the
four biliary components (water, lecithin, choles-
terol, 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 composi-
tion is above the upper limit of the micellar zone,
the sample is supersaturated in cholesterol and at
risk for cholesterol gallstone formation (376). If bili-
ary lipids are known, the percent saturation can also
be obtained from tables published by Carey and col-
league (393, 394).

1578 Journal of Lipid Research Volume 55, 2014


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) pub-
lished in 1957 to describe the proportions of fats, carbohy-
drates, 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 choles-
terol gallstones is discussed later.

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
Fig. 10. Molecular arrangement of the drum-shaped mixed mi-
bile acid. At low ratios, cholesterol was crystalline. At
celle micelle originally proposed by Small, Penkett, and Chapman higher ratios, the excess was liquid crystalline, i.e., in la-
(369) and modified by Carey (330), as compared with the radial mellae of PC and cholesterol. Norman Mazer and Martin
shell model proposed by Nichols and Ozarowski (371). The radial Carey used quasi-elastic light scattering to characterize
shell model was confirmed by Hjelm and colleagues using small model systems simulating bile. At higher PC to bile acid
angle neutron scattering (372, 403, 404) and also by molecular dy-
ratios, they found evidence for “microprecipitates,” al-
namic studies of Marrink and Mark (373).
though they also suggested that these microprecipitates
were vesicles enriched in cholesterol compared with
Another approach to study the interaction of bile acids mixed micelles (379, 380). About the same time, Giora
and phospholipids was taken by Heuman, Bajaj, and Lin Somjen and Tuvia Gilat, working in Israel, used gel perme-
(375). They prepared single layer vesicles and character-
ation chromatography to separate mixed micelles from
ized the binding of bile acids to them. They also deter-
vesicles (381). They equilibrated their columns with
mined at what phospholipid/bile acid ratios the vesicles
10 mM cholate prior to chromatography and thus the ve-
were transformed into micelles.
sicular phase remained unchanged during the chromato-
For teaching purposes, it would be nice to have a video
graphic separation. They also noted that if the columns
with realistic molecular models showing the transfor-
were preequilibrated with buffer alone, a bile sample
mation of lipid bilayers into mixed micelles by bile acid
would become entirely vesicular, as the bile salt molecules
anions.
moved into the aqueous spaces in the gel permeation col-
Physical chemistry of bile and cholesterol umn, thereby increasing the phospholipid/bile acid ratio.
gallstone disease Thomas Holzbach, working in Cleveland, also found vesi-
Supersaturated bile in bile from gallstone patients. D. Small cles in human bile samples and showed that nucleation of
returned from France to Boston in 1965, and began to ap- cholesterol crystals from the cholesterol-rich vesicles oc-
ply his studies on model systems simulating bile to human curred rapidly (382). William Higuchi, working in Salt
biliary lipid compositions. Bile from radiolucent gallstone Lake City, showed, using dialysis techniques, that model
patients and healthy control subjects was collected. The bile systems contained four constituents: monomers, sim-
proportions of bile acid, PC, and cholesterol were plotted ple bile acid micelles, mixed micelles (containing bile ac-
using triangular coordinates in a study in which W. Admi- ids, PC, and cholesterol), and vesicles (also containing the
rand did much of the leg work. A line denoting equilib- three biliary lipids). The vesicles were relatively enriched
rium cholesterol saturation had been obtained by D. Small in cholesterol (383).
based on his studies in Paris: samples above the line were Because of the complexity of the physicochemical as-
supersaturated with cholesterol, whereas those below the pects of bile and because of the widespread interest in gall-
line were unsaturated in cholesterol. When the data were stone pathogenesis and treatment, in 1989, the National
plotted, bile from patients with gallstones was supersatu- Institutes of Health (NIH) sponsored a workshop entitled
rated with or without cholesterol microcrystals, whereas “Biliary Cholesterol Transport and Precipitation.” The
bile from most nongallstone patients was unsaturated proceedings (including much animated discussion) were
(376). [The paper also reintroduced triangular (or ter- published as a supplement to Hepatology the following year
nary) coordinates to the medical audience. Triangular co- (with financial support from the Ciba-Geigy Corporation)
ordinates can be used when there are three variables (384). The meeting included a wide range of scientists
adding up to a constant sum, i.e., there are two degrees of from pure membrane biophysicists to gastroenterologists
freedom, permitting the constructions of graphs in two di- and surgeons, and the discussion often indicated prob-
mensions. They were originally proposed in 1873 by the lems in communication. A summary of the meeting was
great physical chemist, J. Willard Gibbs of Yale University, authored by Steven Strasberg, a scholarly surgeon, and his

History of bile acid research 1579


associate Robert Harvey. Their summary (385) presented positional specificity of pancreatic lipase (395) and then
a reasonably coherent view of bile supersaturation. Cho- showed that triglyceride was absorbed from the intestine
lesterol and phospholipids are secreted in vesicular form. in the chemical form of monoacylglycerol and fatty acid
Bile acids, initially present as simple bile acid micelles, (396).] The solubility of fatty acids in bile acid micelles
solubilize phospholipids and cholesterol. However, the was pH dependent, as protonated fatty acid has a rather
transfer process, presumably occurring by collision, solubi- low solubility in bile acid micelles, whereas fully ionized
lizes phospholipids preferentially, leaving behind vesicles fatty acid (soap) is infinitely soluble (350). Simple titra-
containing an excess of cholesterol (from which choles- tion experiments (397) indicated the pKa of fatty acids
terol crystallizes). Thus, there is a progressive change in in a taurine-conjugated bile acid micelle was about 6.9,
the physical state of bile as it moves from the canaliculus indicating that fatty acids at proximal small intestinal
into the gallbladder. pH (where most fat absorption occurs) are about half
At this meeting, J. Donovan, then working in the Carey ionized. Monoolein, whether the 1- or 2-isomer, was
laboratory, tabulated the existing clinical data (386). The highly soluble, with the ratio of solubilized monoolein
proportion of cholesterol in vesicles was higher in hepatic to micellar bile acid approaching 1.8 (358). In contrast,
bile than in gallbladder bile in all studies, indicating that di- and triacylglycerides had little solubility in bile acid
the transformation of vesicles to mixed micelles continued micelles.
during gallbladder storage of bile. C. Schriever and D. In 1988, M. Svärd, working in the group of Björn Lind-
Juengst showed, using bile samples from gallstone patients, man in Lund, investigated the taurocholate-monoolein-wa-
that the ratio of cholesterol to phospholipid in vesicles pu- ter system by a great variety of techniques, and constructed
rified by gel filtration was 2.0 (387), a ratio that favors for- the complete phase diagram (398). In her paper, she also
mation of cholesterol crystals. noted minor, but consistent, differences between this sys-
Since this meeting, especially because of the declining tem and the bile acid-PC-water system elucidated by the pio-
interest in medical dissolution of gallstones, work on the neering work of D. Small two decades before. What both
physical chemistry of bile has waned. W. Higuchi contin- diagrams share in common is a large micellar zone, as
ued his work on model systems, emphasizing the “activity” shown for the taurocholate-monolein-water system illus-
of monomeric cholesterol (388). Cholesterol saturation trated in Fig. 11. The paper of Svärd et al. (398) also pro-
continued to be calculated from the original triangular poses that bile acid molecules lie flat on the surface of the
figure of D. Small, even though the limit to the micellar bilayers.
zone was moved downward, based on in vitro studies of
model systems from the laboratories of Henrik Dam in Presence of a micellar phase in vivo during triglyceride diges-
Copenhagen (389) and R. T. Holzbach in Cleveland (390). tion. The in vitro findings indicated that fatty acids and
[Henrik Dam, a Dane, had a long interest in cholesterol. monoglycerides were highly soluble in bile acid micelles,
He received the Nobel Prize in 1943 for his discovery of but that di- and triglycerides were not. These observations
vitamin K. In the 1960s, he turned his enormous energy to led to the prediction that during fat digestion, glycerides
the study of cholesterol gallstones and did both animal and fatty acids would be in two phases. There would be an
(391) and clinical studies (392) of note. Sadly most of his
publications are to be found in an obscure journal (Zeitschrift
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 choles-
terol, as well as varying total lipid concentration (393).
The line of saturation is also influenced by bile acid type,
being lower when bile acids are enriched in conjugates of
UDCA. M. Carey has also published critical tables for this
special circumstance (394).

Physical chemistry of fat digestion


Studies on model systems simulating triglyceride digestion. The
physical chemistry of model systems simulating fat diges-
tion 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 glycer-
Fig. 11. Phase diagram of the water-monoolein (monooleylgl-
ides (monoglycerides) were not. Fortunately, they were
ycerol)-taurocholate system showing the large micellar area (cross-
generously supplied by Fred Mattson, a chemist working at hatched), thus depicting the excellent solubilizing potency of bile
the Miami Valley laboratories of Proctor and Gamble acids for polar lipids. Liquid crystal states are indicated by vertical
in Ohio. [Mattson, a gifted biochemist, discovered the and horizontal cross-hatching. Modified from Svärd et al. (398).

1580 Journal of Lipid Research Volume 55, 2014


oil phase containing di- and triacylglycerides and a micel- fatty acids (409). It is well-established that bile acids are
lar phase consisting of bile acid micelles containing partly required for the absorption of fat soluble vitamins.
ionized fatty acid and monoglyceride. The oil phase might The lack of requirement of bile acids for efficient ab-
also contain protonated fatty acids if these were generated sorption of fatty acids with appreciable aqueous solubility
by pancreatic lipolysis at a rate exceeding that which could provides the rationale for using medium chain triglycer-
be solubilized in mixed micelles. Later, healthy volunteers ides for nutritional support in patients with a deficiency of
were intubated, fed a mixed meal, and intestinal content bile acids in the small intestine. The liberated medium
was collected. It was then ultracentrifuged at body tem- chain fatty acids are quite water soluble and are rapidly
perature, and both the oil phase and micellar phase ana- absorbed in the complete absence of bile acids.
lyzed. 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 DISSOLUTION OF CHOLESTEROL GALLSTONES
the M. Carey laboratory. They studied both model systems BY ORAL BILE ACIDS AND TOPICAL SOLVENTS
(400), as well as small intestinal content obtained by intu-
bation in healthy volunteers who had ingested a triglycer- Gallstone dissolution by oral bile acids
ide-rich meal (401). They confirmed the older studies of As noted in the introduction, the famous triangle of
the senior author, but noted in addition, that vesicles were D. Small (and W. Admirand) indicated how biliary lipid
present. They proposed that vesicles form at the surface of composition could be plotted. [The term “biliary lipids” to
the oil droplets, and that these are fairly rapidly converted include bile acids, PC (lecithin), and cholesterol was prob-
to mixed micelles when they encounter unsaturated bile ably introduced by D. Small in the 1960s. It did not in-
acid micelles. In 1959, A. S. C. Lawrence, a colloid chemist clude bilirubin diglucuronide (which gives bile its color),
at Sheffield University, proposed that detergents adsorb to as this pigment, derived from heme, is a very minor con-
“dirt” and form liquid crystalline myelin figures (multilay- stituent of bile. Biliary lipids differ from plasma lipids in
ered vesicles). His analysis (402) is probably an apt de- that biliary lipids include bile acids, and do not include
scription of the formation of vesicles composed of fatty glycerides and fatty acids.]
acid and monoacylglycerol at the surface of the triglycer- In the 1950s, Charles Johnston, a surgeon at Wayne
ide droplet. State University, was interested in determining what he
A closing chapter on this topic was authored by R. Hjelm, termed the “cholesterol holding capacity” of bile, that is,
in which model mixtures simulating fat digestion were how much additional cholesterol could be dissolved when
prepared in our laboratory and examined by small angle added to bile samples. He observed that T-tube bile from
neutron scattering. Hjelm concluded that mixed micelles gallstone patients was “saturated”, whereas bile samples
containing a bile acid and either monoglyceride (monoac- obtained from healthy subjects undergoing surgery was
ylglycerol) or monoglycerides plus fatty acids had essen- unsaturated. With a young Japanese surgeon, Fumio Na-
tially the same structure as the mixed micelles containing kayama, he fed cholic acid to gallstone patients, but found
bile acids and PC that he had studied previously (403, that it did not increase the cholesterol holding capacity. In
404). In both, the bile acid molecules lie flat on the sur- the discussion of his paper, he noted that he hoped to try
face of the spherical or worm shaped micelles. CDCA, but of course, at that time, there was no CDCA to
be had (410). [Fumio Nakayama continued to study gall-
Necessity of bile acids for efficient absorption of triglycerides. Nu- stone pathogenesis using chemical techniques and had a
merous studies dating back to the 19th century had shown long and distinguished career as a Professor of Surgery in
that triglyceride absorption efficiency was impaired in ani- Kyushu, Japan. After he retired he summarized his life-
mals with a biliary fistula and that feeding of bile acids long interest in gallstone formation and treatment in a
would correct fat malabsorption (405). Nonetheless, there monograph (411).]
were efforts to show that bile acids were unnecessary for After J. Thistle and L. Schoenfield made the seminal dis-
fatty acid absorption (406). The view of the senior author covery that CDCA, but not cholic acid, changed bile from
is that bile acids are essential for the efficient absorption supersaturated to unsaturated, it was logical to perform a
of those fatty acids that have very low aqueous solubili- clinical trial of CDCA for gallstone dissolution. The effort
ties, i.e., saturated fatty acids of C16 length or longer (407). was spearheaded by J. Thistle, as L. Schoenfield took a new
Fatty acids with appreciable aqueous solubility (unsatu- position in Los Angeles in 1970. In 1971, the first observa-
rated fatty acids and saturated acids having a chain length tions were made that gallstones were decreasing in size,
<C16) do not require bile acids for efficient absorption. probably the first witnesses being J. Thistle (and his radio-
This view that bile acids facilitate the absorption of satu- logical colleague) and the senior author. In 1972, success-
rated fatty acids (⭓C16 in length) is based on two observa- ful gallstone dissolution was reported in May at the annual
tions. First, fecal fatty acid analyses indicate that exogenous meeting of the American Gastroenterological Associa-
bile acids cause a decrease in the proportion of long chain tion and later that year in Europe at the 2nd Interna-
saturated fatty acids in short bowel syndrome patients tional Bile Acid Meeting sponsored by the Falk Foundation
(408). Second, administration of cholestyramine to rats of Freiburg, Germany. The early Mayo Clinic experi-
causes selective malabsorption of long chain saturated ence with gallstone dissolution was published in the New

History of bile acid research 1581


England Journal of Medicine in 1972 and became a “Cita- Europe which showed that CDCA had unequivocal effi-
tion Classic” (37). Later that year, gallstone dissolution cacy and minimal toxicity (424, 425).
by CDCA was reported by the group of R. H. Dowling, An analysis of the kinetics of gallstone dissolution by
working at Guy’s Hospital in London (38). In 1973, Dr. William Higuchi indicated that the reduction in diameter
Falk Pharma decided to market CDCA for gallstone induced by the presence of unsaturated bile occurred in-
dissolution. dependently of size (426). Thus, the bigger the stone, the
In February 1972, a small meeting was held at the NIH longer the time needed for complete dissolution. This
to discuss a national multicenter placebo-controlled trial prediction was later confirmed by Senior et al. (427) in a
of CDCA. It was felt that the absence of patent protection thoughtful analysis of X-ray changes of gallstone size dur-
would preclude development by pharmaceutical compa- ing medical dissolution.
nies. A “Request for Proposals” was issued in July 1972. L. Just as the NCGS was beginning, word came from Japan
Schoenfield submitted a proposal which included commit- (a careful study led by Isao Makino and his colleagues)
tee structure for the proposed National Cooperative Gall- that ingestion of UDCA, the 7␤ epimer of CDCA, would
stone Study (NCGS). On the committees were most every also induce gallstone dissolution (40). Earlier, there were
American scientist or physician interested in gallstone several anecdotal cases in which subjects had taken large
disease! doses of UDCA and induced their own gallstone dissolu-
The study was delayed, because of the Food and Drug tion, and this was done without knowledge of the Ameri-
Administration (FDA) concern that CDCA might cause can studies with CDCA. Thus, medical dissolution of
liver damage. The FDA demanded that liver biopsies be gallstones must be considered an American and Japanese
performed on the first 100 patients, including those on discovery.
placebo. Despite the ethical problems inherent in subject- In the United States, the NCGS drafted a protocol that
ing placebo group patients to liver biopsy, the study was would permit a study similar to the original NCGS, but
performed without any adverse events. would compare UDCA with CDCA; however, the study was
The concern of the FDA was based in part on two re- not funded by the NIH. CDCA was approved for market-
ports from the Huntington Laboratories in England of se- ing in the United States by the FDA in 1983, and UDCA
vere toxicity induced by CDCA feeding in the rhesus was approved a few years later. UDCA, like CDCA, was also
monkey (412). As a result of these reports, European stud- toxic in the rhesus monkey (428) and rabbit (153, 429)
ies were stopped in 1973. Report of the primate toxicity because of lithocholic acid accumulation in the circulat-
provided a justification for liver biopsies in the patients ing bile acids.
when therapy was stopped (413). An international registry Despite the safety and efficacy of CDCA and UDCA,
of biopsies was set up, and a careful review of the biopsy there were some physicians who were unimpressed by
histology showed no important histological changes. Tri- medical therapy as compared with surgical therapy.
als in Europe recommenced. There were several problems with medical therapy. First,
The answer to this remarkable species difference in it took a long time, months to years, to obtain complete
CDCA toxicity came from work by the senior investigator dissolution, especially for larger stones. Second, studies
and his colleagues, who showed that in humans, lithocho- by Wolpers and Hofmann (430) suggested that with the
lic acid, the major bacterial metabolite of CDCA, is sul- first attack of biliary colic, the resulting inflammation
fated and rapidly excreted from the body (414, 415); would cause a surface layer of noncholesterol constitu-
whereas in the rhesus monkey, lithocholic acid is ex- ents on the gallstone, slowing or precluding dissolution.
tremely poorly sulfated, accumulates in the circulating Thus, there was no window of opportunity: when stones
bile acids, and induces liver damage (416, 417). Indepen- became symptomatic, they had already become resistant
dent studies by the laboratory of E. Mosbach reported that to medical dissolution. Moreover, it took many months
CDCA was also toxic in the rabbit, again because of litho- of therapy before one could be sure that the stone was
cholic acid accumulation (418). Similar findings were ob- resistant. Finally, after stones dissolved, there was recur-
served when rhesus monkeys (419) and baboons (420) rence (431). It was gallstone recurrence that led surgeons
were given CDCA. a half century before to switch from cholecystotomy (re-
The NCGS began in 1976 and its results were published moval of the stone but leaving the gallbladder in place)
in 1981 (421). The study was a double blind study with to cholecystectomy (432). A thoughtful review by Gracie
three groups: placebo, 350 mg CDCA/day, and 750 mg and Ransohoff concluded that it was better to “wait and
CDCA/day. Dissolution occurred in the placebo group cut” than to “screen and treat,” because of the benign
(11%), was greater (24%) in the low dose group, and natural history of most gallstones (433). An editorial by
highest (41%) in the high dose group. During the study, it K. Isselbacher was entitled “Disillusion with Dissolution”
was recognized that the high dose was probably still too (434).
low a dosage (422), but the study could not be changed. In
the high dose group, 8% of patients had to discontinue Acceleration of dissolution by extracorporeal shockwave
the study because of an increase in serum aminotransfer- lithotripsy
ase levels. Toxicity was shown to be attributable to CDCA Attempts were made to speed up dissolution. One ap-
and not to lithocholic acid (423). While the NCGS was proach was to fragment the stones by using extracor-
proceeding, controlled studies were being performed in poreal shockwave lithotripsy, a technique that had been

1582 Journal of Lipid Research Volume 55, 2014


developed for the fragmentation of kidney stones. For was a report from France of MTBE escaping into the blood
gallstones, the stone fragments would be smaller than the stream and causing severe renal damage (444). In the
original stone(s), have cholesterol surfaces exposed by the United States, two deaths occurred unrelated to usage of
shattering of the stones, and thus, medical dissolution the solvent, but nonetheless in association with solvent us-
would occur more rapidly. However, there was a major dif- age. A company was formed to bring the pump and solvent
ference between kidney stones and gallbladder stones. into clinical practice. However, venture capital was never
When kidney stones were fractured by this technique, the obtained, and the approach slowly passed into oblivion. A
fragments passed spontaneously down the urinary tract review of topical dissolution by our group was published
and were expelled during urination. With gallbladder stones, (445).
the fragments sunk to the bottom of the gallbladder, so
that dissolution by desaturating bile acids was needed. An- Concurrent advances in gallstone management
other limitation of extracorpeal shock wave lithotripsy was Two other developments were occurring during this
that only certain types of stones (one or two in number) time that doomed widespread usage of medical dissolu-
could be treated. Thorough clinical studies of the efficacy tion. The first was the development of laparoscopic chole-
and safety of this technique for gallstone dissolution were cystectomy, a major surgical advance. Laparoscopic
conducted by the Munich group led by Gustav Paumgartner surgery was an import from France, used first in gyneco-
(435). Leslie Schoenfield led one of several efforts in the logical procedures. Using this technique, a gallbladder
United States (436). could be removed quickly, without a linear scar, and the
patient could leave the hospital in a few days. The gallbladder
Topical dissolution with organic solvents with its gallstones had been removed, and the patient was
Another approach to nonsurgical treatment of choles- cured. In experienced hands, the technique was as safe as
terol gallstones was contact (topical) dissolution with or- the traditional open cholecystectomy which had been per-
ganic solvents. The Mayo Clinic group, led by J. Thistle, formed widely for the past half century. The composition
introduced the idea of placing a percutaneous transhe- of the stone(s) was unimportant, whereas with oral bile
patic catheter into the gallbladder and then lavaging the acid therapy only gallstones that had cholesterol as the
stone with methyl tert-butyl ether (MTBE), a solvent that major constituent could be successfully treated.
had become available as a gasoline additive (437) and was The second advance was ultrasound imaging of the gall-
a superb cholesterol solvent. When the solvent was bladder and its contents. All types of stones were detected,
pumped into and then aspirated from the gallbladder, and there was no radiation exposure. In contrast, oral cho-
stones would dissolve in a matter of a few days. A spark-free lecystography required the ingestion of iodine-rich con-
pump was designed and patented by the Mayo Clinic. Eu- trast agents and subsequent X-ray to detect the presence
ropean trials commenced in a few centers (438). of gallstones.
In our laboratory at University of California, San Diego, Thus in twenty years, medical dissolution had been dis-
we had not planned to become involved in this approach. covered, prospered, and then slowly disappeared. In the
A patient with symptomatic gallstones who was a poor op- long history of biliary disease, medical dissolution was a
erative risk presented with symptomatic gallstones, and we “bubble”. Once again symptomatic cholelithiasis became a
reluctantly carried out successful dissolution by manual surgical disease. Nonetheless, medical dissolution was
instillation and aspiration of MTBE. A “smart” pump was “proof of principle”, and medical therapy with UDCA can
designed by S. Zakko so that solvent could be pumped into be used to prevent cholesterol gallstone formation. Gall-
and out of the gallbladder at a high rate. The pump was stone prophylaxis might be useful in some clinical situa-
programmed to continuously sense intraluminal pressure tions, for example, in patients with recurrent gallstone
and aspirate gallbladder contents whenever the gallblad- pancreatitis who did not wish to have surgery.
der started to contract, thus avoiding spillage of the sol- The availability of UDCA led to the accidental discov-
vent into the common bile duct (439). A new solvent, ethyl ery of its utility in the treatment of primary biliary cir-
propionate, was tested and found to dissolve gallstones rhosis and cholestasis of pregnancy, as noted in the
nearly as rapidly as MTBE. Ethyl propionate did not in- introduction.
duce nausea in contrast to MTBE because of rapid re-
moval from plasma (440). Patients were treated successfully
(441). EPILOGUE: OMISSIONS AND APOLOGIES
However, there were multiple problems with contact
dissolution. The solvents used were commercial samples Space limitations means that historical aspects of many
and they were not manufactured according to FDA stan- facets of biology and chemistry have been omitted. We
dards. MTBE was flammable and considered toxic (442). have not discussed the history of inborn errors of bile acid
Preclinical studies had not been done. The procedure re- synthesis and conjugation. Our understanding of these
quired some hours for complete dissolution, and as de- genetic defects is quite recent, and an excellent review
scribed, it required professional personnel to perform the (446) by Clayton is available. In the United States, Ken
infusion and aspiration. In studies performed in pigs, the Setchell, a talented mass spectroscopist, has described
gallbladder epithelium was observed to be destroyed by several new inborn errors of bile acid biosynthesis, as well
either solvent, although it recovered rapidly (443). There as a defect in bile acid conjugation. Identification of the

History of bile acid research 1583


exact enzyme defect requires MS to detect the presence There is still much is to be learned about the effect of in-
of bile acid precursors and gene sequencing to identify dividual bile acids on the microbiome (465).
the defect (156). The rewards of discovery are great for The last two generations have had powerful tools to elu-
the patient, as bile acid replacement may be life-saving. cidate bile acid metabolism. One of these was the discov-
Some of the early history is given in J. Sjövall’s retrospec- ery of 14C, a discovery that almost occurred by chance
tive review (10). (466). The availability of 14C in chemical form permitted
14
A second area not discussed is the utility of bile acids C-labeled bile acids to be prepared and their metabolism
and bile alcohols to disclose evolutionary relationships, an to be defined. A second essential tool has been chroma-
area pioneered by G. A. D. Haslewood (21). Often bile tography, adsorption, reverse-phase (HPLC), and gas/liq-
acids agree with the perceived evolutionary relationships, uid. A final tool is MS, which when coupled with GC or
but sometimes they do not. It would be of interest to ex- HPLC provides both qualitative and quantitative informa-
plore these nonconcordancies in detail. tion on new molecules (9). For the assignment of struc-
A third area not discussed is the role of bile acids in co- ture to newly discovered bile acids, proton and 13C NMR
lonic secretion (447, 448) and absorption (449), as well as measurements are mandatory. Of course, in today’s re-
recent work suggesting that bile acid activation of TGR5 is search, generation of specific knockouts, characterized by
essential for normal defecation, at least in the mouse (70). multisystem phenotyping, combined with PCR, have enor-
Details of the biochemical mechanism(s) for bile acid-in- mous power in unraveling the recently discovered signal-
duced intestinal secretion are being elucidated by several ing properties of bile acids.
laboratories, among these being that of Stephen Keely in At present (2014), at least two types of drugs based on
Dublin (448), and that of Mrinalini Rao in Chicago (450). natural bile acids are being tested for their utility in the
Bile acid malabsorption causes diarrhea, and this can be treatment of liver disease. The first, obeticholic acid, the
treated by sequestrant administration (451, 452). A new 6␣-ethyl derivative of CDCA, is a potent FXR agonist. Its
syndrome of defective FGF19 release causing an inappro- efficacy in the treatment of primary biliary cirrhosis (467)
priate increase in synthesis of bile acids has been identi- and nonalcoholic steatohepatitis (468) is being tested in
fied by Julian Walters et al. (453), and is considered to be clinical trials. norUDCA, the C23 homolog of UDCA, is also
present in many patients with irritable bowel syndrome of in clinical trials for the treatment of primary sclerosing
the diarrhea type (454), as well as in patients with so-called cholangitis. In the bile duct ligated mouse, its feeding
primary bile acid malabsorption. Plasma levels of “C4” causes less damage than UDCA feeding (469).
(7␣-hydroxy-cholest-4-ene-3-one), an intermediate in C24 Inhibitors of ASBT, the sodium dependent bile
bile acid biosynthesis, have been shown to correlate highly acid transporter present in the apical domain of the ileal
with the rate of bile acid synthesis (455, 456). The neces- enterocyte, are once again being developed. They were
sity of measuring C4 (as a marker of increased bile acid originally aimed at providing adjuvant therapy for hyper-
synthesis) and FGF15/19 levels (to detect impaired cholesterolemic patients who responded inadequately
FGF15/19 secretion from the ileal enterocyte) in every pa- to statins (470). Now the target is cholestatic liver disease
tient with unexplained diarrhea is slowly being recognized or its symptom of pruritus, as well as type 2 diabetes
by gastroenterologists (457). The group of Michael Camil- (471) and constipation (472). Colesevelam is an improved
leri at the Mayo Clinic is exploring the relationship of fe- bile acid sequestrant that should have a similar pharma-
cal bile acid profile to constipation as well as diarrheal codynamic activity as that of the ASBT inhibitors; how-
conditions (458). Our laboratory identified some children ever, it is only approved for type II diabetes, and its use to
with functional constipation whose fecal bile acids were treat bile acid diarrhea is off label. Bile acid derivatives
predominantly the 3-sulfate of CDCA (119). The absence that activate solely TGR5, or both TGR5 and FXR, have
of C-12 hydroxylation in these children could indicate a been synthesized (55) and may eventually enter clinical
deficiency in cyp8B1, the enzyme that hydroxylates C4 at trials.
the C-12 position (459). In our opinion, the remarkable progress in our under-
A fourth area not discussed is the cellular toxicity of bile standing of bile acid biology and chemistry is a tribute to
acids. Mechanisms of toxicity are multiple, involving acti- the hundreds of scientists who have worked with such dili-
vation of the death receptor (460), as well as induction of gence and talent throughout the world. We have men-
reactive oxygen species (461). UDCA is cytoprotective and tioned only a few in our brief history, and we have surely
mechanisms responsible for this effect are being studied omitted some key advances. There are many workers
(462). A new aspect of bile acid toxicity is the proinflam- whose contributions have not been recognized because of
matory action of retained bile acids (463). space limitations. As we noted in the introduction, this is a
One last area not discussed is the possible influence of personal history, and other writers are likely to stress dif-
bile acids on the microbiome. The presence of secondary ferent aspects of the historical record. We have probably
bile acids in bile has long been a sign that metabolic activ- overemphasized the contributions of our own laboratory
ity of the microbiome influences bile acid metabolism. In- because we know them best. For those readers who would
deed, characterization of bile acid metabolism in the like more historical detail on some of the topics discussed
germ-free animal (464) by Gustafsson and his colleagues above, we refer them to a series of beautifully written his-
in Lund aided in the initial distinction of primary and sec- torical reviews by Adrian Reuben published in the journal,
ondary bile acids by the laboratory of Sune Bergström (8). Hepatology, between 2002 and 2006 (c.f. 482).

1584 Journal of Lipid Research Volume 55, 2014


Let us hope that future research in the chemistry and 26. Une, M., and T. Hoshita. 1994. Natural occurrence and chemical
synthesis of bile alcohols, higher bile acids, and short side chain
biology of bile acids will be as exciting as that of the last 80
bile acids. Hiroshima J. Med. Sci. 43: 37–67.
years! 27. Yamasaki, K. 1972. 40-year studies on bile acids [article in
German]. Yonago Acta Med. 15: 171–187.
28. Carey, J. B., Jr. 1956. Chenodeoxycholic acid in human blood
The authors are grateful to many of our colleagues who helped serum. Science. 123: 892.
us in this review. In particular, we thank Takashi Iida, Bruno 29. Carey, J. B., Jr., I. D. Wilson, F. G. Zaki, and R. F. Hanson. 1966.
Stieger, and David Mangelsdorf, as well as the anonymous The metabolism of bile acids with special reference to liver injury.
Medicine. 45: 461–470.
reviewers. 30. Ahrens, E. H., Jr., and L. C. Craig. 1952. The extraction and sepa-
ration of bile acids. J. Biol. Chem. 195: 763–778.
REFERENCES 31. Grundy, S. M., E. H. Ahrens, Jr., and T. A. Miettinen. 1965.
Quantitative isolation and gas-liquid chromatographic analysis of
1. Thannhauser, S. J., and M. Lenke. 1928. Ueber die Herkunft der total fecal bile acids. J. Lipid Res. 6: 397–410.
Gallensäuren, Cholesterin-Gallensäurenbilanzen beim Hund mit 32. McGuire, D. K., and S. M. Grundy. 2011. Interview with Dr. Scott
totaler Gallenfistel. Arch. Exp. Pathol. Pharmakol. 1130: 292–307. Grundy during AHA EPI/PAM meeting in March 2011. J. Clin.
2. Neubauer, E. 1924. Über die Cholagoge Wirkung der Lipidol. 5: 371–372.
Dehydrocholsäure beim Menschen. Klin. Wochenschr. 3: 883. 33. Rajaratnam, R. A., H. Gylling, and T. A. Miettinen. 2001.
3. Shimizu, T. 1935. Über die Chemie and Physiologie der Cholesterol absorption, synthesis, and fecal output in post-
Gallensäuren. Verlag von M. Muramoto, Japan. menopausal women with and without coronary artery disease.
4. Sarett, L. H. 1946. Partial synthesis of pregnene 4 triol 17(␤),20(␤), Arterioscler. Thromb. Vasc. Biol. 21: 1650–1655.
21 dione 3,11 and pregnene 4 diol 17(␤), 21 trione 3,11,20 mono- 34. Grundy, S. M., A. F. Hofmann, J. Davignon, and E. H. Ahrens, Jr.
acetate. J. Biol. Chem. 162: 601–631. 1966. Human cholesterol synthesis is regulated by bile acids. J.
5. Hillier, S. G. 2007. Diamonds are forever: the cortisone legacy. J. Clin. Invest. 45: 1018–1019.
Endocrinol. 195: 1–6. 35. Thistle, J. L., and L. J. Schoenfield. 1971. Induced altera-
6. Hench, P. S. 1964. The reversibility of certain rheumatic and non- tions in composition of bile of persons having cholelithiasis.
rheumatic conditions by the use of cortisone or of the pituitary Gastroenterology. 61: 488–496.
adrenocorticotropic hormone. In Nobel Lectures. Physiology or 36. Danzinger, R. G., A. F. Hofmann, L. J. Schoenfield, and J. L.
Medicine 1942–1962. Elsevier Publishing Company, Amsterdam. Thistle. 1972. Dissolution of cholesterol gallstones by chenode-
312–341. oxycholic acid. N. Engl. J. Med. 286: 1–8.
7. Fieser, L. F., and M. Fieser. 1959. Adrenocortical hormones. In 37. Bell, G. D., B. Whitney, and R. H. Dowling. 1972. Gallstone disso-
Steroids. Reinhold Publishing Corporation, New York. 600–726. lution in man using chenodeoxycholic acid. Lancet. 2: 1213–1216.
8. Bergström, S., H. Danielsson, and B. Samuelsson. 1960. Formation 38. Hench, P. S. 1938. Effect of jaundice on rheumatoid arthritis.
and metabolism of bile acids. In Lipide Metabolism. K. Bloch, edi- BMJ. 2: 394–398.
tor. John Wiley & Sons, New York, NY. 291–336. 39. Sugata, F., and M. Shimizu. 1974. Retrospective studies on gall-
9. Griffiths, W. J., and J. Sjövall. 2010. Bile acids: analysis in biologi- stone disappearance [article in Japanese]. Nihon Shokakibyo Gakkai
cal fluids and tissues. J. Lipid Res. 51: 23–41. Zasshi. 71: 75–80.
10. Sjövall, J. 2004. Fifty years with bile acids and steroids in health 40. Nakagawa, S., I. Makino, T. Ishazaki, and I. Dohi. 1977. Dissolution
and disease. Lipids. 39: 703–722. of cholesterol gallstones by ursodeoxycholic acid. Lancet. 2:
11. Norman, A. 1955. Preparation of conjugated bile acids using 367–369.
mixed carboxylic acid anhydrides. Ark. Kemi. 8: 331–342. 41. Makino, I., and H. Tanaka. 1998. From a choleretic to an immu-
12. Norman, A., and J. Sjövall. 1958. On the transformation and en- nomodulator: historical review of ursodeoxycholic acid as a medi-
terohepatic circulation of cholic acid in the rat. J. Biol. Chem. 233: cament. J. Gastroenterol. Hepatol. 13: 659–664.
872–885. 42. Leuschner, U., and W. Kurtz. 1987. Treatment of primary bili-
13. Norman, A., and J. Sjövall. 1960. Formation of lithocholic acid from ary cirrhosis and cholestatic disorders with ursodeoxycholic acid.
chenodeoxycholic acid in the rat. Acta Chem. Scand. 14: 1815–1818. Lancet. 2: 508.
14. Lindstedt, S. 1957. The turnover of cholic acid in man. Acta 43. Poupon, R., Y. Chretien, R. E. Poupon, F. Ballett, Y. Calmus, and
Physiol. Scand. 40: 1–9. F. Darnis. 1987. Is ursodeoxycholic acid an effective treatment for
15. Danielsson, H., and J. Sjövall. 1975. Bile acid metabolism. Annu. primary biliary cirrhosis? Lancet. 1: 834–836.
Rev. Biochem. 44: 233–253. 44. Lindor, K. D., E. R. Dickson, W. P. Baldus, R. A. Jorgensen, J.
16. Samuelsson, B. 1959. On the metabolism of ursodeoxycholic acid Ludwig, P. A. Murtaugh, J. M. Harrison, R. H. Wiesner, M. L.
in the rat. Acta Chem. Scand. 13: 970–975. Anderson, S. M. Lange, et al. 1994. Ursodeoxycholic acid in
17. Eriksson, S. 1957. Biliary excretion of bile acids and cholesterol in the treatment of primary biliary cirrhosis. Gastroenterology. 106:
bile fistula rats. Proc. Soc. Exp. Biol. Med. 94: 578–582. 1284–1290.
18. Borgström, B., A. Dahlqvist, G. Lundh, and J. Sjövall. 1957. Studies 45. Palma, J., H. Reyes, J. Ribalta, I. Hernandez, L. Sandoval, R.
of intestinal digestion and absorption in the human. J. Clin. Invest. Almuna, J. Liepins, F. Lira, M. Sadano, O. Silva, et al. 1997.
36: 1521–1536. Ursodeoxycholic acid in the treatment of cholestasis of preg-
19. Lack, L., and I. M. Weiner. 1961. In vitro absorption of bile salts by nancy: a randomized, double-blind study controlled with placebo.
small intestine of rats and guinea pigs. Am. J. Physiol. 200: 313–317. J. Hepatol. 27: 1022–1028.
20. Borgström, B., G. Lundh, and A. F. Hofmann. 1963. The site of 46. Nonappa, and U. Maitra. 2008. Unlocking the potential of bile
absorption of conjugated bile salts in man. Gastroenterology. 45: acids in synthesis, supramolecular/materials chemistry and nano-
229–238. science. Org. Biomol. Chem. 6: 657–669.
21. Haslewood, G. A. D. 1967. Bile salt evolution. J. Lipid Res. 8: 47. Allen, R. D., I. Y. Wan, G. M. Wallraff, R. A. DiPietro, D. C. Hofer,
535–550. and R. R. Kunz. 1995. Resolution and etch resistance of a family
22. Hsia, S. L., W. H. Elliott, J. T. Matschiner, E. A. Doisy, Jr., S. A. of 193nm positive resists. J. Photopolym. Sci. Technol. 8: 623–626.
Thayer, and E. A. Doisy. 1958. Bile acids. XI. Structures of the 48. Steinberg, D. 2013. In celebration of the 100th anniversary of the
isomeric 3 alpha,6.7-trihydroxycholanic acids. J. Biol. Chem. 233: lipid hypothesis of atherosclerosis. J. Lipid Res. 54: 2946–2949.
1337–1339. 49. Makishima, M., A. Y. Okamoto, J. J. Repa, H. Tu, R. M. Learned,
23. Shaw, R., and W. H. Elliott. 1978. Bile acids LVII. Analysis of bile A. Luk, M. V. Hull, K. D. Lustig, D. J. Mangelsdorf, and B. Shan.
acids by high pressure liquid chromatography and mass spectrom- 1999. Identification of a nuclear receptor for bile acids. Science.
etry. Lipids. 13: 971–975. 284: 1362–1365.
24. Haslewood, G. A. D. 1958. Prof. T. Shimizu (obituary). Nature. 50. Wang, H., J. Chen, K. Hollister, L. C. Sowers, and B. M. Forman.
181: 880. 1999. Endogenous bile acids are ligands for the nuclear receptor
25. Hoshita, T. 1985. Bile alcohols and primitive bile acids. In Sterols FXR/BAR. Mol. Cell. 3: 543–553.
and Bile Acids. H. Danielsson and J. Sjövall, editors. Elsevier 51. Parks, D. J., S. G. Blanchard, R. K. Bledsoe, G. Chandra, T. G.
Science Publishers, Amsterdam. 279–302. Consler, S. A. Kliewer, J. B. Stimmel, T. M. Willson, A. M. Zavacki,

History of bile acid research 1585


D. D. Moore, et al. 1999. Bile acids: natural ligands for an orphan is required for normal defecation in mice. Gastroenterology. 144:
nuclear receptor. Science. 284: 1365–1368. 145–154.
52. Matsubara, T., F. Lei, and F. J. Gonzalez. 2013. FXR signaling in 71. Wang, Y-D., W-D. Chen, D. Yu, B. M. Forman, and W. Huang.
the enterohepatic system. Mol. Cell. Endocrinol. 368: 17–29. 2011. The G-protein-coupled bile acid receptor. Gpbar1 (TGR5),
53. Gonzalez, F. J. 2012. Nuclear receptor control of enterohepatic negatively regulates hepatic inflammatory response through an-
circulation. Compr. Physiol. 2: 2811–2828. tagonizing nuclear factor Kappa light-chain enhancer of activated
54. Porez, G., J. Prawitt, B. Gross, and B. Staels. 2012. Bile acid recep- B cells (NF-␬B) in mice. Hepatology. 54: 1421–1432.
tors as targets for the treatment of dyslipidemia and cardiovascu- 72. Sobotka, H. 1938. The Chemistry of the Steroids. Williams and
lar disease. J. Lipid Res. 53: 1723–1737. Wilkins, Baltimore, MD.
55. Inagaki, T., M. Choi, A. Moschetta, L. Peng, C. L. Cummins, J. 73. Sobotka, H. 1937. Physiological Chemistry of the Bile. Williams
G. McDonald, G. Luo, S. A. Jones, B. Goodwin, J. A. Richardson, and Wilkins, Baltimore, MD.
et al. 2005. Fibroblast growth factor 15 functions as an enterohepatic 74. Haslewood, G. A. D. 1967. Bile Salts. Methuen, London.
signal to regulate bile acid homeostasis. Cell Metab. 2: 217–225. 75. Haslewood, G. A. D. 1978. The Biological Importance of Bile Salts.
56. Jung, D., T. Inagaki, R. D. Gerard, P. A. Dawson, S. A. Kliewer, D. North-Holland Publishing Co., Amsterdam, The Netherlands.
J. Mangelsdorf, and A. Moschetta. 2007. FXR agonists and FGF15 76. 1962. Elsevier’s Encyclopedia of Organic Chemistry (suppl) F.
reduce fecal bile acid excretion in a mouse model of bile acid Radt, editor. Elsevier, Amsterdam, The Netherlands. 22915–36725.
malabsorption. J. Lipid Res. 48: 2693–2700. 77a. Nair, P. P., and D. Kritchevsky, editors. 1971. The Bile Acids. Vol.
57. Uriarte, I., M. G. Fernandez-Barrena, M. J. Monte, M. U. Latasa, I. Chemistry. Plenum Press, New York.
H. C. Y. Chiang, S. Carotti, U. Vespasiani-Gentilucci, S. Morini, 77b. Nair, P. P., and D. Kritchevsky, editors. 1973. The Bile Acids: Vol.
E. Vicente, A. R. Concepcion, et al. 2013. Identification of fibro- 2 Physiology and Metabolism. Plenum Press, New York.
blast growth factor 15. A novel mediator of liver regeneration and 77c. Nair, P. P., and D. Kritchevsky, editors. 1976. The Bile Acids: Vol.
its application in the prevention of post-resection liver failure in 3 Pathophysiology. Plenum Press, New York.
mice. Gut. 62: 899–910. 77d. Setchell, K. D. R., D. Kritchevsky, and P. P. Nair, editors. The Bile
58. Mudaliar, S., R. R. Henry, A. J. Sanyal, L. Marrow, H. U. Marschall, Acids. Vol. 4. Chemistry, Physiology, and Metabolism. Plenum
M. Kipnes, L. Adorini, C. I. Sciacca, P. Clopton, E. Castelloe, Press, New York.
et al. 2013. Efficacy and safety of the farnesoid X receptor agonist 78. Danielsson, H., and J. Sjövall, editors. 1985. Sterols and Bile Acids.
obeticholic acid in patients with type 2 diabetes and nonalcoholic In New Comprehensive Biochemistry, Vol. 12. A. Neuberger and
fatty liver disease. Gastroenterology. 145: 574–582. L. L. M. VanDeenen, editors. Elsevier, Amsterdam.
59. Gadaleta, R. M., K. J. van Erpecum, B. Oldenburg, E. C. 79. Roda, A., E. Roda, and A. F. Hofmann, editors. 1999. Acidi biliari
Willemsen, W. Renooij, S. Murzilli, L. W. Klomp, P. D. Siersema, 2000. Aggiornamento per il future. Masson, Milano, Italy.
M. E. Schipper, S. Danese, et al. 2011. Farnesoid X receptor acti- 80. Hofmann, A. F. 1985. Bile acid research: some major themes during
vation inhibits inflammation and preserves the intestinal barrier the past 60 years. In Trends in Hepatology. L. Bianchi, W. Gerok, and
in inflammatory bowel disease. Gut. 60: 463–472. H. Popper, editors. MTP Press Limited, Lancaster, England. 3–28.
81. Dowling, R. H. 1985. Bile acids in therapy – pre-1924, circa 1924,
60. Sato, H., A. Macchiarulo, C. Thomas, A. Gioiello, M. Une, A. F.
and in 1984. In Trends in Hepatology. L. Bianchi, W. Gerok, and H.
Hofmann, R. Saladin, K. Schoonjans, R. Pellicciari, and J. Auwerx.
Popper, editors. MTP Press Limited, Lancaster, England. 29–40.
2008. Novel potent and selective bile acid derivatives as TGR5
82. Hofmann, A. F., L. R. Hagey, and M. D. Krasowski. 2010. Bile salts
agonists: biological screening, structure-activity relationships, and
of vertebrates: structural variation and possible evolutionary sig-
molecular modeling studies. J. Med. Chem. 51: 1831–1841.
nificance. J. Lipid Res. 51: 226–246.
61. Pellicciari, R., S. Fiorucci, E. Camaioni, C. Clerici, G. Costantino,
83. Wieland, H. 1928. The chemistry of the bile acids. In Nobel
P. R. Maloney, A. Morelli, D. J. Parks, and T. M. Willson. 2002. 6␣-
Lectures, Chemistry 1922–1941. Elsevier Publishing Company,
ethyl chenodeoxycholic acid (6-ECDCA), a potent and selective
Amsterdam, The Netherlands. 94–102.
FXR agonist endowed with anticholestatic activity. J. Med. Chem. 84. Windaus, A. 1928. Constitution of sterols and their connec-
45: 3569–3572. tion with other substances occurring nature. In Nobel Lectures,
62. Adorini, L., M. Pruzanski, and D. Shapiro. 2012. Farnesoid X re- Chemistry 1922–1941. Elsevier Publishing Company, Amsterdam,
ceptor targeting to treat nonalcoholic steatohepatitis. Drug Discov. The Netherlands. 105–121.
Today. 17: 988–997. 85. Bernal, J. D. 1932. Crystal structures of vitamin D and related com-
63. Maruyama, T., Y. Miyamoto, T. Nakamura, Y. Tamai, H. Okada, pounds. Nature. 129: 277–278.
E. Sugiyama, T. Nakamura, H. Itadani, and K. Tanaka. 2002. 86. Rosenheim, O., and H. King. 1934. The chemistry of the sterols,
Identification of membrane-type receptor for bile acids (M-BAR). bile acids, and other cyclic constituents of natural fats and oils.
Biochem. Biophys. Res. Commun. 298: 714–719. Annu. Rev. Biochem. 3: 87–110.
64. Kawamata, Y., R. Fujii, M. Hosova, M. Harada, H. Yoshida, M. 87. Lindley, P. F., and M. C. Carey. 1987. Molecular packing of bile ac-
Miwa, S. Fukusumi, Y. Habata, T. Itoh, Y. Shintani, et al. 2003. A ids: structure of ursodeoxycholic acid. J. Crystallogr. Spectrosc. Res.
G protein-coupled receptor responsive to bile acids. J. Biol. Chem. 17: 231–249.
278: 9435–9440. 88. Kametani, T., K. Suzuki, and H. Nemoto. 1981. First total synthesis
65. Poole, D. P., C. Godfrey, F. Cattanruzza, G. S. Cottrell, J. G. of (+)-chenodeoxycholic acid. J. Am. Chem. Soc. 103: 2891–2892.
Kirkland, J. C. Pelayo, N. W. Bunnett, and C. U. Corvera. 2010. 89. Swann, B., and F. Aprahamian, editors. 1999. J. D. Bernal: A Life
Expression and function of the bile acid receptor GpBAR1 in Science and Politics. Verso, London, UK.
(TGR5) in the murine enteric nervous system. Neurogastroenterol. 90. Brown, A. 2005. J. D. Bernal. The Sage of Science. Oxford
Motil. 22: 814–825. University Press, Oxford, UK .
66. Alemi, F., E. Kwon, D. P. Poole, T. Lieu, V. Lyo, F. Cattaruzzza, F. 91. Whipple, G. H. 1922. The origin and significance of the constitu-
Cevikbas, M. Steinhoff, R. Nassini, S. Materazzi, et al. 2013. The ents of the bile. Physiol. Rev. 2: 440–459.
TGR5 receptor mediates bile acid-induced itch and analgesia. J. 92. Ruzicka, L. 1973. In the borderland between bioorganic chemis-
Clin. Invest. 123: 1513–1530. try and biochemistry. Annu. Rev. Biochem. 42: 1–20.
67. Keitel, V., and D. Häussinger. 2013. TGR5 in cholangiocytes. Curr. 93. Berthold, H. K. 1998. Rudolf Schoenheimer (1898–1941): Leben
Opin. Gastroenterol. 29: 299–304. und Werk. Falk Foundation, e.V., Freiburg, Germany.
68. Thomas, C., A. Gioiello, L. Noriega, A. Strehle, J. Oury, G. Rizzo, 94. Bloch, K., B. N. Berg, and D. Rittenberg. 1943. The biological con-
A. Macchiarulo, H. Yamamoto, C. Mataki, M. Pruzanski, et al. version of cholesterol to cholic acid. J. Biol. Chem. 149: 511–517.
2009. TGR5-mediated bile acid sensing controls glucose homeo- 95. Shimizu, T., and T. Oda. 1934. Untersuchung der Krotengalle. II.
stasis. Cell Metab. 10: 167–177. Trioxy-bufo-sterocholensäure C28H46O5 aus Wintergalle. Z. Phys.
69. Keitel, V., R. Reinehr, P. Gatsios, C. Rupprecht, B. Görg, O. Chem. 227: 74–83.
Selbach, D. Häussinger, and R. Kubitz. 2007. The G-protein cou- 96. Suganami, T., and K. Yamasaki. 1942. Uber die
pled bile salt receptor TGR5 is expressed in liver sinusoidal endo- Alligatorschildkrotengalle. II. Trioxy-stero- and trioxy-isostero-
thelial cells. Hepatology. 45: 695–704. cholansäure. J. Biochem. 35: 155–172.
70. Alemi, F., D. P. Poole, J. Chiu, K. Schoonjans, F. Cattaruzza, J. R. 97. Tint, G. S., B. Dayal, A. K. Batta, S. Shefer, T. Joanen, L. McNease,
Grider, N. W. Bunnett, and C. U. Corvera. 2013. The receptor and G. Salen. 1980. Biliary bile acids, bile alcohols, and sterols of
TGR5 mediates the prokinetic actions of intestinal bile acids and Alligator mississippiensis. J. Lipid Res. 21: 110–117.

1586 Journal of Lipid Research Volume 55, 2014


98. Bridgwater, R. J., and G. A. D. Haslewood. 1952. Comparative 120. Kelsey, M. I., J. E. Molina, S. K. Huang, and K. K. Hwang. 1980.
studies of bile salts. 6. Partial synthesis of coprostanic acid, a stem The identification of microbial metabolites of sulfolithocholic
acid of primitive bile salts. Biochem. J. 52: 588–590. acid. J. Lipid Res. 21: 751–759.
99. Ogawa, S., K. Mitamura, S. Ikegawa, M. D. Krasowski, L. R. Hagey, 121. Cohen, B. I., A. K. Singhal, J. Mongelli, M. A. Rothschild, C. K.
and A. F. Hofmann. 2011. Chemical synthesis of the (25R)- and McSherry, and E. H. Mosbach. 1983. Hydroxylation of secondary
(25S)-epimers of 3␣,7␣,12␣-trihydroxy-5␤-cholestan-27-oic acid as bile acids in the perfused prairie dog liver. Lipids. 18: 909–912.
well as their corresponding glycine and taurine conjugates. Chem. 122. Kakiyama, G., H. Tamegai, T. Iida, K. Mitamura, S. Ikegawa, T.
Phys. Lipids. 164: 368–377. Goto, N. Mano, J. Goto, P. Holz, L. R. Hagey, et al. 2007. Isolation
100. Hammarsten, O. 1898. Ueber eine neue Gruppe gepaarter of a major novel dominant biliary bile acid in the wombat: 15␣-
Gallensäure. Z. Phys. Chem. 24: 322–350. hydroxylithocholic acid. J. Lipid Res. 48: 2682–2692.
101. Kazuno, T., T. Masui, and K. Okuda. 1965. Stero-bile acids and 123. Shefer, S., G. Salen, S. Hauser, B. Dayal, and A. K. Batta.
bile sterols. LXIV. The isolation and the chemistry of a new bile 1982. Metabolism of iso-bile acids in the rat. J. Biol. Chem. 257:
alcohol, 3-alpha, 7-alpha, 12-alpha, 24-xi, 26-pentahydroxybisho- 1401–1406.
mocholane from Rana catesbiana bile. J. Biochem. 57: 75–80. 124. Marschall, H. U., U. Broome, C. Einarsson, G. Alvelius, H. G.
102. Haslewood, G. A. D. 1964. Comparative studies of bile salts. 19. Thomas, and S. Matern. 2001. Isoursodeoxycholic acid: metabo-
The chemistry of ranol. Biochem. J. 90: 309–313. lism and therapeutic effects in primary biliary cirrhosis. J. Lipid
103. Haslewood, G. A., and L. Tokes. 1969. Comparative studies of bile Res. 42: 735–742.
salts. Bile salts of the lamprey Petromyzon marinus L. Biochem. J. 114: 125. Hagey, L. R., T. Iida, S. Ogawa, Y. Adachi, M. Une, K. Mushiake,
179–184. M. Maekawa, M. Shimada, N. Mano, and A. F. Hofmann. 2011.
104. Hoye, T. R., V. Dvomikovs, J. M. Fine, K. R. Anderson, C. S. Biliary bile acids in birds of the Cotingidae family: taurine-con-
Jeffrey, D. C. Muddiman, F. Shao, P. W. Sorensen, and J. Wang. jugated (24R,25R)-3␣,7␣,24-trihydroxy-5␤-cholestan-27-oic acid
2007. Details of the structure determination of the sulfated and two epimers (25R and 25S) of 3␣,7␣-dihydroxy-5␤-cholestan-
steroids PSDS and PADS: new components of the sea lamprey 27-oic acid. Steroids. 76: 1126–1135.
(Petromyzon marinus) migratory pheromone. J. Org. Chem. 72: 126. Haslewood, G. A. D., and L. Tokès. 1972. Comparative studies
7544–7550. of bile salts. A new type of bile salt from Arapaima gigas (Cuvier)
105. Hagey, L., R. K. Takagi, C. D. Schteingart, H-T. Ton-Nu, and A. (family Osteoglossidae). Biochem. J. 126: 1161–1170.
F. Hofmann. 1994. C24 bile acid reduction into bile alcohols: a 127. Kuroki, S., C. D. Schteingart, L. R. Hagey, B. I. Cohen, E. H.
novel, ubiquitous minor pathway of cholesterol elimination in Mosbach, S. S. Rossi, A. F. Hofmann, N. Matoba, M. Une, T.
vertebrates. Hepatology. 20: A665. Hoshita, et al. 1988. Bile salts of the West Indian manatee,
106. Elliott, W. H. 1971. Allo bile acids. In The Bile Acids: Chemistry, Trichechus manatus latirostris: novel bile alcohol sulfates and ab-
Physiology, Metabolism. Vol. 1. P. P. Nair and D. Kritchevsky, edi- sence of bile acids. J. Lipid Res. 29: 509–522.
tors. Plenum Press, New York, NY. 47–93. 128. Ishida, H., H. Miyamoto, T. Kajino, H. Nakayasu, H. Nukaya, and
107. Hofmann, A. F., and E. H. Mosbach. 1964. Identification of al- K. Tsuji. 1996. Study on the bile salt from Megamouth shark. I.
lodeoxycholic acid as the major component of gallstones induced The structures of a new bile alcohol, 7-deoxyscymnol, and its new
in the rabbit by 5␣ cholestan-3␤-ol. J. Biol. Chem. 239: 2813–2821. sodium sulfates. Chem. Pharm. Bull. (Tokyo). 44: 1289–1292.
108. Mendoza, M. E., M. J. Monte, M. A. Serrano, M. Pastor-Anglada, 129. Hammarsten, O. 1909. Untersuchung über die Galle einiger
B. Stieger, P. J. Meier, M. Medarde, and J. J. Marin. 2003. Polartiere. III. Mitteilung über die Galle des Wallrosses. Hoppe
Physiological characteristics of allo-cholic acid. J. Lipid Res. 44: Seylers Z. Physiol. Chem. 61: 454–496.
84–92. 130. Windaus, A., and A. van Schoor. 1928. Uber die ␤-Phocae-
109. Setchell, K. D. R., J. M. Street, and J. Sjövall. 1988. Fecal bile ac- cholsäure. Hoppe Seylers Z. Physiol. Chem. 143: 312–320.
ids. In The Bile Acids: Chemistry, Physiology, Metabolism. Vol. 4. 131. Pellicciari, R., B. Natalini, A. Roda, M. Iracema, L. Machdao, and
K. D. R. Setchell, D. Kritchevsky, and P. P. Nair, editors. Plenum M. Marinzzi. 1989. Preparation and physicochemical properties
Press, New York, NY. 441–570. of natural (23R)-hydroxy trihydroxylated bile acids and their
110. Ridlon, J. M., D. J. Kang, and P. B. Hylemon. 2006. Bile salt bio- (23S) epimers. J. Chem. Soc. Perkin Trans. 1: 1289–1296.
transformation by human intestinal bacteria. J. Lipid Res. 47: 132. Merrill, J. R., C. D. Schteingart, L. R. Hagey, Y. Peng, H-T. Ton-Nu,
241–259. E. Frick, M. Jirsa, and A. F. Hofmann. 1996. Hepatic biotrans-
111. Katona, B. W., N. P. Rath, S. Anant, W. F. Stenson, and D. F. formation in rodents and physicochemical properties of 23(R)-
Covey. 2007. Enantiomeric deoxycholic acid: total synthesis, char- hydroxychenodeoxycholic acid, a natural ␣-hydroxy bile acid. J.
acterization, and preliminary toxicity toward colon cancer lines. J. Lipid Res. 37: 98–112.
Org. Chem. 72: 9298–9307. 133. Hoshita, T., S. Hirofuji, T. Sasaki, and T. Kazuno. 1967. Stero-bile
112. Strecker, A. 1848. Untersuchung der Ochsengalle. Annalen der acids and bile alcohols. LXXXVII. Isolation of a new bile acid,
Chem. und Pharmacie. 67: 1–60. haemulcholic acid from the bile of Parapristipoma trilineatum. J.
113. Mylius, F. 1886. Ueber die Cholsäure. Ber. 19: 374. Biochem. 61: 136–141.
114. Otto, R. 1869. Ueber die Gänsegalle und die Chenotaurocholsäure. 134. Kihira, K., Y. Morioka, and T. Hoshita. 1981. Synthesis of (22R
Annalen der Chem. und Pharmacie. 149: 185–201. and 22S)-3 alpha, 7 alpha, 22-trihydroxy-5-beta-cholan-24-oic ac-
115. Marschall, H-U., M. Wagner, K. Bodin, G. Zöllner, P. Fickert, J. ids and structure of haemulcholic acid, a unique bile acid isolated
Gumhold, D. Silbert, A. Fuchsbichler, J. Sjövall, and M. Trauner. from fish bile. J. Lipid Res. 22: 1181–1187.
2006. FXR(-/-) mice adapt to biliary obstruction by enhanced 135. Roda, A., B. Grigolo, A. Minutello, R. Pellicciari, and B. Natalini.
phase I detoxification and renal elimination of bile acids. J. Lipid 1990. Physicochemical and biological properties of natural and syn-
Res. 47: 582–592. thetic C-22 and C-23 hydroxylated bile acids. J. Lipid Res. 31: 289–298.
116. Wang, R., L. Liu, J. A. Sheps, D. Forrest, A. F. Hofmann, L. R. 136. Rodrigues, C. M. P., B. T. Kren, C. J. Steer, and K. D. R. Setchell.
Hagey, and V. Ling. 2013. Defective canalicular transport and 1996. Formation of ⌬22-bile acids in rats is not gender specific and
toxicity of dietary ursodeoxycholic acid in the abcb11-/- mouse: occurs in the peroxisome. J. Lipid Res. 37: 540–550.
transport and gene expression studies. Am. J. Physiol. Gastrointest. 137. Kakiyama, G., T. Iida, A. Yoshimuta, T. Goto, N. Mano, J. Goto,
Liver Physiol. 305: G286–G294. T. Nambara, L. R. Hagey, and A. F. Hofmann. 2004. Chemical
117. Hofmann, A. F., J. Sjövall, G. Kurz, A. Radominska, C. D. synthesis of (22E)-3␣,6␤.7␤-trihydroxy-5␤-chol-22-ene-24-oic acid
Schteingart, G. S. Tint, Z. R. Vlahcevic, and K. D. R. Setchell. 1992. and its taurine and glycine conjugates: a major bile acid in the rat.
A proposed nomenclature for bile acids. J. Lipid Res. 33: 599–604. J. Lipid Res. 45: 567–573.
118. Bokkenheuser, V. D., J. Winter, P. B. Hylemon, N. K. N. 138. Une, M., F. Nagai, K. Kihira, T. Kuramoto, and T. Hoshita.
Ayengar, and E. H. Mosbach. 1981. Dehydroxylation of 16␣- 1983. Synthesis of four diastereoisomers at carbons 24 and 26 of
hydroxyprogesterone by fecal flora of man and rat. J. Lipid Res. 3␣,7␣,12␣.24-tetrahydroxy-5␤-cholestan-26-oic acid, intermedi-
22: 95–102. ates of bile acid biosynthesis. J. Lipid Res. 24: 924–929.
119. Hofmann, A. F., V. Loening-Baucke, J. E. Lavine, L. R. Hagey, J. 139. Hagey, L. R., S. Ogawa, N. Kato, R. Satoh née Okihara, M. Une,
H. Steinbach, C. A. Packard, T. L. Griffin, and D. A. Chatfield. K. Mitamura, S. Ikegawa, A. F. Hofmann, and T. Iida. 2012. A
2008. Altered bile acid metabolism in childhood functional con- novel varanic acid epimer–(24R,25S)-3␣,7␣,12␣,24-tetrahydroxy-
stipation: inactivation of secretory bile acids by sulfation in a sub- 5␤-cholestan-27-oic acid–is a major biliary bile acid in two varanid
set of patients. J. Pediatr. Gastroenterol. Nutr. 47: 598–606. lizards and the Gila monster. Steroids. 77: 1510–1521.

History of bile acid research 1587


140. Windaus, A. W. B., and G. König. 1930. Uber einige Versuche mit bile salts shuttle through hepatocyte peroxisomes for taurine con-
Scymnol. Z. Phys. Chem. 189: 148–154. jugation. Hepatology. 52: 2167–2176.
141. Asikari, H. 1939. Uber die Galle des “Akajei” Fisches (Dasyatis aka- 161. Huijghebaert, S. M., and A. F. Hofmann. 1986. Influence of the
jei) und die Konstitution des Scymnols. J. Biochem. 29: 319–324. amino acid moiety on deconjugation of bile acid amidates by
142. Bergmann, W., and W. T. Pace. 1943. Scymnol. J. Am. Chem. Soc. cholylglycine hydrolase or human fecal cultures. J. Lipid Res. 27:
65: 477. 742–752.
143. Bridgwater, R. J., T. Briggs, and G. A. D. Haslewood. 1962. 162. Hepner, G. W., J. A. Sturman, A. F. Hofmann, and P. J. Thomas.
Comparative studies of ‘bile salts’. 14. Isolation from shark bile 1973. Metabolism of steroid and amino acid moieties of conju-
and partial synthesis of scymnol. Biochem. J. 82: 285–290. gated bile acids in man. III. Cholyltaurine (taurocholic acid). J.
144. Bridgwater, R. J., G. A. D. Haslewood, and J. R. Watt. 1963. Clin. Invest. 52: 433–440.
Comparative studies of ‘bile salts’. 17. A bile alcohol from Chimaera 163. Yoon, Y. B., L. R. Hagey, A. F. Hofmann, D. Gurantz, E. L.
monstrosa. Biochem. J. 87: 28–31. Michelotti, and J. H. Steinbach. 1986. Effect of side chain short-
145. Cross, A. D. 1961. Scymnol sulphate and anhydroscymnol. J. Chem. ening on the physiological properties of bile acids: hepatic trans-
Soc. 2817–2821. port and effect on biliary secretion of 23-Nor-ursodeoxycholate
146. Ishida, H., S. Kinoshita, R. Natsuyama, H. Nukaya, K. Tsuji, T. in rodents. Gastroenterology. 90: 837–852.
Kosuge, and K. Yamaguchi. 1991. Study on the bile salt, sodium 164. Yeh, H-Z., C. D. Schteingart, L. R. Hagey, H-T. Ton-Nu, U.
scymnol sulfate, from Rhizoprionodon acutus. II. The structures of Bolder, M. A. Gavrilkina, J. H. Steinbach, and A. F. Hofmann.
scymnol, anhydroscymnol, and sodium scymnol sulfate. Chem. 1997. Effect of side chain length on biotransformation, hepatic
Pharm. Bull. (Tokyo). 39: 3153–3156. transport, and choleretic properties of chenodeoxycholyl ho-
147. Ishida, H., H. Nakayasu, H. Miyamoto, H. Nukaya, and K. Tsuji. mologues in the rodent: studies with dinorchenodeoxycholic
1998. Study on the bile salts from Sunfish, Mola mola L. I. The acid, norchenodeoxycholic acid, and chenodeoxycholic acid.
structures of sodium cyprinol sulfates, the sodium salt of a new Hepatology. 26: 374–385.
bile acid conjugated with taurine, and a new bile alcohol and its 165. Rossi, S. S., J. L. Converse, and A. F. Hofmann. 1987. High pres-
new sodium sulfates. Chem. Pharm. Bull. (Tokyo). 46: 12–16. sure liquid chromatographic analysis of conjugated bile acids in
148. Dayal, B., A. K. Batta, S. Shefer, G. S. Tint, G. Salen, and E. H. human bile: simultaneous resolution of sulfated and unsulfated
Mosbach. 1978. Preparation of 24(R)- and 24(S)-5beta-cholestane- lithocholyl amidates and the common conjugated bile acids. J.
3alpha,7alpha,24-triols and 25(R)- and 25(S)-5beta-cholestane- Lipid Res. 28: 589–595.
3alpha,7alpha,26-triols by a hydroboration procedure. J. Lipid Res. 166. Schwenk, M., A. F. Hofmann, G. L. Carlson, J. A. Carter, F. Coulston,
19: 191–196. and H. Greim. 1978. Bile acid conjugation in the chimpanzee: ef-
149. Satoh née Okihara, R., T. Saito, H. Ogata, A. Ohsaki, T. Iida, fective sulfation of lithocholic acid. Arch. Toxicol. 40: 109–118.
K. Asahina, K. Mitamura, S. Ikegawa, A. F. Hofmann, and L. R. 167. Raedsch, R., B. H. Lauterburg, and A. F. Hofmann. 1981. Altered
Hagey. 2014. N-methyltaurine N-acyl amidated bile acids and de- bile acid metabolism in primary biliary cirrhosis. Dig. Dis. Sci. 26:
oxycholic acid in the bile of angelfish (Pomacanthidae): a novel 394–401.
bile acid profile in Perciform fish. Steroids. 80: 15–23. 168. Stiehl, A. 1977. Disturbances of bile acid metabolism in cholesta-
150. Une, M., T. Goto, K. Kihira, T. Kuramoto, K. Hagiwara, T. sis. Clin. Gastroenterol. 6: 45–67.
Nakajima, and T. Hoshita. 1991. Isolation and identification of 169. Barnes, S., P. G. Burhol, R. Zander, G. Haggstrom, R. L.
bile salts conjugated with cysteinolic acid from bile of the red Settine, and B. I. Hirschowitz. 1979. Enzymatic sulfation of
seabream, Pagrosomus major. J. Lipid Res. 32: 1619–1623. glycochenodeoxycholic acid by tissue fractions from adult ham-
151. Batta, A. K., G. Salen, and S. Shefer. 1984. Substrate specificity of sters. J. Lipid Res. 20: 952–959.
cholyl glycine hydrolase for the hydrolysis of bile acid conjugates. 170. Almé, B., A. Nordén, and J. Sjövall. 1978. Glucuronides of un-
J. Biol. Chem. 259: 15035–15039. conjugated 6-hydroxylated bile acids in urine of a patient with
152. Schmassmann, A., M. A. Angellotti, H-T. Ton Nu, C. D. malabsorption. Clin. Chim. Acta. 86: 251–259.
Schteingart, S. N. Marcus, S. S. Rossi, and A. F. Hofmann. 1990. 171. Sacquet, E., M. Parquet, M. Riottot, A. Raizman, P. Jarrige, C.
Transport, metabolism and effect of chronic feeding of cholyl- Huguet, and R. Infante. 1983. Intestinal absorption, excretion,
sarcosine, a conjugated bile acid resistant to deconjugation and and biotransformation of hyodeoxycholic acid in man. J. Lipid
dehydroxylation. Gastroenterology. 98: 163–174. Res. 24: 604–613.
153. Schmassmann, A., A. F. Hofmann, M. A. Angellotti, H-T. Ton-Nu, 172. Radominska-Pyrek, A., P. Zimniak, Y. M. Irshaid, R. Lester, T.
C. D. Schteingart, C. Clerici, S. S. Rossi, M. A. Rothschild, B. I. R. Tephly, and J. S. Pyrek. 1987. Glucuronidation of 6 alpha-
Cohen, et al. 1990. Prevention of ursodeoxycholate hepatotox- hydroxy bile acids by human liver microsomes. J. Clin. Invest. 80:
icity in the rabbit by conjugation with N-methyl amino acids. 234–241.
Hepatology. 11: 989–996. 173. Matschiner, J. T., T. A. Mahowald, S. L. Hsia, E. A. Doisy, Jr., W.
154. Lillienau, J., C. D. Schteingart, and A. F. Hofmann. 1992. H. Elliott, and E. A. Doisy. 1957. Bile acids. IV. The metabolism of
Physicochemical and physiological properties of cholylsarcosine: hyodeoxycholic acid-24-C14. J. Biol. Chem. 225: 803–810.
a potential replacement detergent for bile acid deficiency states in 174. Kirkpatrick, R. B., M. D. Green, L. R. Hagey, A. F. Hofmann, and
the small intestine. J. Clin. Invest. 89: 420–431. T. R. Tephly. 1988. Effect of side chain length on bile acid con-
155. Russell, D. W. 2003. The enzymes, regulation, and genetics of bile jugation: glucuronidation, sulfation, and CoA formation of nor
acid synthesis. Annu. Rev. Biochem. 72: 137–174. bile acids and their natural C24 homologues by human rat liver
156. Hylemon, P. B., P. M. Bohdan, A. E. Sirica, D. M. Heuman, fractions. Hepatology. 8: 353–357.
and Z. R. Vlahcevic. 1990. Cholesterol and bile acid metabo- 175. Hofmann, A. F., S. F. Zakko, M. Lira, C. Clerici, L. R. Hagey, K.
lism in cultures of primary rat bile ductular cells. Hepatology. K. Lambert, J. H. Steinbach, C. D. Schteingart, P. Olinga, and G.
11: 982–988. M. M. Groothuis. 2005. Novel biotransformation and physiologi-
157. Ikegawa, S., H. Ishikawa, H. Oiwa, M. Nagata, J. Goto, T. Kozaki, cal properties of norursodeoxycholic acid in man. Hepatology. 42:
M. Gotowda, and N. Asakawa. 1999. Characterization of cholyl- 1391–1398.
adenylate in rat liver microsomes by liquid chromatography/ 176. Borgström, B., J. Barrowman, L. Krabish, M. Lindstrom, and J.
electrospray ionization-mass spectrometry. Anal. Biochem. 266: Lillienau. 1986. Effects of cholic acid, 7 beta-hydroxy and 12 beta-
125–132. hydroxy-isocholic acid on bile flow, lipid secretion, and bile acid
158. Falany, C. N., M. R. Johnson, S. Barnes, and R. B. Diasio. 1994. synthesis in the rat. Scand. J. Clin. Lab. Invest. 46: 167–175.
Glycine and taurine conjugation of bile acids by a single en- 177. Marschall, H. U., W. J. Griffiths, U. Gotze, J. Zhang, H. Wietholtz,
zyme. Molecular cloning and expression of human liver bile N. Busch, J. Sjövall, and S. Matern. 1994. The major metabolites
acid CoA:amino acid N-acyltransferase. J. Biol. Chem. 269: of ursodeoxycholic acid in human urine are conjugated with
19375–19379. N-acetylglucosamine. Hepatology. 20: 845–853.
159. Kase, B. F., and I. Björkhem. 1989. Peroxisomal bile acid- 178. Iida, T., G. Kakiyama, Y. Hibiya, S. Miyata, T. Inoue, K. Ohno,
CoA:amino-acid N-acyltransferase in rat liver. J. Biol. Chem. 264: T. Goto, N. Mano, J. Goto, T. Nambara, et al. 2006. Chemical
9220–9223. synthesis of the 3-sulfooxy-7-N-acetylglucosaminyl-24-amidated
160. Rembacz, K. P., J. Woudenberg, M. Hoekstra, E. Z. Jonkers, F. A. conjugates of 3␤,7␤-dihydroxy-5-cholen-24-oic acid, and related
van den Heuvel, M. Buist-Homan, T. E. Woudenberg-Vrenken, J. compounds: unusual, major metabolites of bile acid in a patient
Rohacova, M. L. Marin, M. A. Miranda, et al. 2010. Unconjugated with Niemann-Pick disease type C1. Steroids. 71: 18–29.

1588 Journal of Lipid Research Volume 55, 2014


179. Mitamura, K., N. Hori, T. Iida, M. Suzuki, T. Shimizu, H. Nittono, 202. Hofmann, A. F. 1964. Thin-layer chromatographic mobilities of
K. Takaori, A. F. Hofmann, and S. Ikegawa. 2011. The identifica- bile acids. In New Biochemical Separations. L. J. Morris and A. T.
tion of S-acyl glutathione conjugates of bile acids in human bile by James, editors. Van Nostrand, London. 261–282.
means of LC/ESI/MS. Steroids. 76: 1609–1614. 203. Snyder, F., and H. Kimble. 1965. An automatic zonal scraper
180. Dietschy, J. M., and S. D. Turley. 2002. Control of cholesterol and sample collector for radioassay of thin-layer chromatograms.
turnover in the mouse. J. Biol. Chem. 277: 3801–3804. Anal. Biochem. 11: 510–518.
181. Moschetta, A., F. Xu, L. R. Hagey, G. P. van Berge-Henegouwen, 204. Linnet, K., J. R. Andersen, and P. Hesselfeldt. 1984. Concentrations
K. J. van Erpecum, J. F. Brouwers, J. C. Cohen, M. Bierman, H. H. of glycine- and taurine-conjugated bile acids in portal and sys-
Hobbs, J. H. Steinbach, et al. 2005. A phylogenetic survey of bili- temic venous serum in man. Scand. J. Gastroenterol. 19: 575–578.
ary lipids in vertebrates. J. Lipid Res. 46: 2221–2232. 205. Roda, A., A. M. Gioacchini, C. Cerrè, and M. Baraldini. 1995. High-
182. Erb, W., and E. Böhle. 1969. Fecal excretion of lipids in infec- performance liquid chromatographic-electrospray mass spectro-
tious hepatitis, liver cirrhosis, and obstructive jaundice. Klin. metric analysis of bile acids in biological fluids. J. Chromatogr. B
Wochenschr. 47: 249–256. Biomed. Appl. 665: 281–294.
183. van der Velde, A. E., C. L. Vrins, K. van den Oever, I. Seemann, R. 206. Kakiyama, G., A. Muto, H. Takei, H. Nittono, T. Murai, T.
P. Oude Elferink, M. van Eck, F. Kuipers, and A. K. Groen. 2008. Kurosawa, A. F. Hofmann, W. M. Pandak, and J. S. Bajaj. 2014. A
Regulation of direct transintestinal cholesterol excretion in mice. simple and accurate HPLC method for fecal bile acid profile in
Am. J. Physiol. Gastrointest. Liver Physiol. 295: G203–G208. healthy and cirrhotic subjects: validation by GC-MS and LC-MS. J.
184. Fini, A., and A. Roda. 1987. Chemical properties of bile acids. IV. Lipid Res. 55: 978–990.
Acidity constants of glycine-conjugated bile acids. J. Lipid Res. 28: 207. Iwata, T., and K. Yamasaki. 1964. Enzymatic determination and
755–759. thin-layer chromatography of bile acids in blood. J. Biochem. 56:
185. Huijghebaert, S. M., and A. F. Hofmann. 1986. Pancreatic car- 424–431.
boxypeptidase hydrolysis of bile acid amino acid conjugates: selec- 208. Hurlock, B., and P. Talalay. 1957. Principles of the enzymatic
tive resistance of glycine and taurine amidates. Gastroenterology. 90: measurement of steroids. J. Biol. Chem. 227: 37–52.
306–315. 209. Turley, S. D., and J. N. Dietschy. 1978. Re-evaluation of the 3␣-
186. Makino, I., S. Nakagawa, and K. Mashimo. 1969. Conjugated and hydroxysteroid dehydrogenase assay for total bile acids in bile. J.
unconjugated serum bile acid levels in patients with hepatobiliary Lipid Res. 19: 924–928.
diseases. Gastroenterology. 56: 1033–1039. 210. Beher, W. T., S. Stradnieks, and G. J. Lin. 1983. Rapid photomet-
187. Setchell, K. D. R., A. M. Lawson, E. J. Blackstock, and G. P. ric determination of free and esterified 3 alpha-hydroxy fecal bile
Murphy. 1982. Diurnal changes in serum unconjugated bile acids acids. Steroids. 41: 729–740.
in normal man. Gut. 23: 637–642. 211. Roda, A., L. J. Kricka, M. DeLuca, and A. F. Hofmann. 1982.
188. Hagey, L. R., and M. D. Krasowski. 2013. Microbial biotransforma- Bioluminescent measurement of primary bile acids using immo-
tions of bile acids as detected by electrospray mass spectrometry. bilized 7␣-hydroxysteroid dehydrogenase: application to serum
Adv. Nutr. 4: 29–35. bile acids. J. Lipid Res. 23: 1354–1361.
212. Rossi, S. S., M. A. Angellotti, K. D. Setchell, and A.F. Hofmann.
189. Hayakawa, S. 1982. Microbial transformation of bile acids. A uni-
1991. Measurement of total fasting-state serum bile acids: compar-
fied scheme for bile acid degradation and hydroxylation of bile
ison of a solid-phase bioluminescence enzymatic assay, a homo-
acids. Z. Allg. Mikrobiol. 22: 309–326.
geneous fluorescence enzymatic assay, and isotope dilution gas
190. Goto, T., F. Holzinger, L. R. Hagey, C. Cerrè, H. T. Ton-Nu, C. D.
chromatography-mass spectrometry. Clin. Chim. Acta. 200: 63–66.
Schteingart, J. H. Steinbach, B. L. Shneider, and A. F. Hofmann.
213. Simmonds, W. J., M. G. Korman, V. L. W. Go, and A. F. Hofmann.
2003. Physicochemical and physiological properties of 5␣-
1973. Radioimmunoassay of conjugated cholyl bile acids in se-
cyprinol sulfate, the toxic bile salt of cyprinid fish. J. Lipid Res. 44:
rum. Gastroenterology. 65: 705–711.
1643–1651.
214. LaRusso, N. F., M. G. Korman, N. E. Hoffman, and A. F. Hofmann.
191. Sjövall, J. 1953. On the separation of bile acids by partition chro- 1974. Dynamics of the enterohepatic circulation of bile acids.
matography. Acta Physiol. Scand. 29: 232–240. Postprandial serum concentrations of conjugates of cholic acid
192. Haslewood, G. A. D., and J. Sjövall. 1954. Comparative studies of in health, cholecystectomized patients, and patients with bile acid
‘Bile Salts’ 8. Preliminary examination of bile salts by paper chro- malabsorption. N. Engl. J. Med. 291: 689–692.
matography. Biochem. J. 57: 126–130. 215. Schalm, S. W., G. P. van Berge-Henegouwen, A. F. Hofmann, A.
193. Sandberg, D. H., J. Sjövall, K. Sjövall, and D. A. Turner. 1965. E. Cowen, and J. Turcotte. 1977. Radioimmunoassay of bile acids:
Measurement of human serum bile acids by gas-liquid chroma- development, validation and preliminary application of an as-
tography. J. Lipid Res. 6: 182–192. Q26

say for conjugates of chenodeoxycholic acid. Gastroenterology. 73:


194. Eneroth, P., G. Gordon, R. Ryhage, and J. Sjövall. 1966. 285–290.
Identification of mono- and dihydroxy bile acids in human feces 216. Schalm, S. W., N. F. LaRusso, A. F. Hofmann, N. E. Hoffman, G. P.
by gas-liquid chromatography and mass spectrometry. J. Lipid Res. van Berge Henegouwen, and M. G. Korman. 1978. Diurnal serum
7: 511–523. levels of primary conjugated bile acids. Assessment by specific ra-
195. Eneroth, P., and J. Sjövall. 1971. Extraction, purification, and dioimmunoassays for conjugates of cholic and chenodeoxycholic
chromatographic analysis of bile acids in biological materials. In acid. Gut. 19: 1006–1014.
The Bile Acids: Chemistry, Physiology, and Metabolism. Vol. 1. P. 217. van der Velden, L. M., M. V. Golynskiy, I. T. G. W. Bijmans, S. W.
P. Nair and D. Kritchevsky, editors. Plenum Press, New York, NY. C. van Mil, L. W. J. Klomp, M. Merix, and S. F. J. van deGraaf.
121–68. 2013. Monitoring bile acid transport in single living cells using
196. Stahl, E. 1962. Dünnschichtchromatographie. Springer Verlag, a genetically encoded Förster resonance energy transfer sensor.
Berlin, Germany. Hepatology. 57: 740–752.
197. Malins, D. C., and H. Mangold. 1960. Analysis of complex lipid 218. Cravetto, C., G. Molino, A. M. Biondi, A. Cavanne, P. Avagnina,
mixtures by thin-layer chromatography and complementary and S. Frediani. 1985. Evaluation of the diagnostic value of serum
methods. J. Am. Oil Chem. Soc. 37: 576–578. bile acid in the detection and functional assessment of liver dis-
198. Hofmann, A. F. 1962. Thin layer adsorption chromatography on eases. Ann. Clin. Biochem. 22: 596–605.
microscope slides. Anal. Biochem. 3: 145–149. 219. Mishler, J. M., L. Barbosa, L. J. Mihalko, and H. McCarter. 1981.
199. Hofmann, A. F. 1962. Thin-layer adsorption chromatography Serum bile acids and alanine aminotransferase concentrations.
of free and conjugated bile acids on silicic acid. J. Lipid Res. 3: Comparison of efficacy as indirect means of identifying carriers of
127–128. non-A, Non-B hepatitis agents and of onset, severity, and duration
200. Lepri, L., D. Heimler, and P. G. Desiderio. 1984. Reversed-phase of posttransfusion non-A, non-B hepatitis in recipients. JAMA.
high-performance thin-layer chromatography of free and conju- 246: 2340–2344.
gated bile acids. J. Chromatogr. 288: 461–468. 220. Ennulat, D., M. Magid-Slav, S. Rehm, and K. S. Tatsuoka. 2010.
201. Momose, T., M. Mure, T. Iida, J. Goto, and T. Nambara. 1998. Diagnostic performance of traditional hepatobiliary biomarkers
Method for the separation of the unconjugates and conjugates of of drug-induced liver injury in the rat. Toxicol. Sci. 116: 397–412.
chenodeoxycholic acid and deoxycholic acid by two-dimensional 221. Glantz, A., H. U. Marschall, and L. A. Mattsson. 2004. Intrahepatic
reversed-phase thin layer chromatography with methyl beta-cyclo- cholestasis of pregnancy: Relationships between bile acid levels
dextrin. J. Chromatogr. A. 811: 171–180. and fetal complication rates. Hepatology. 40: 467–474.

History of bile acid research 1589


222. Cowen, A. E., M. G. Korman, A. F. Hofmann, O. W. Cass, and S. hepatocyte Na+/bile acid cotransport system. Proc. Natl. Acad. Sci.
B. Coffin. 1975. Metabolism of lithocholate in healthy man. II. USA. 88: 10629–10633.
Enterohepatic circulation. Gastroenterology. 69: 67–76. 248. Geyer, J., T. Wilke, and E. Petzinger. 2006. The solute carrier fam-
223. Tappeiner, A. J. F. H. 1878. Ueber die Aufsaugung der Gallsäuren ily SLC10: more than a family of bile acid transporters regarding
alkaline im Dunndarme. Wien. Akad. Sitzber. 77: 281–304. function and phylogenetic relationships. Naunyn Schmiedebergs
224. Verzar, F. 1936. Absorption from the Intestine. Longmans, Green Arch. Pharmacol. 372: 413–431.
and Co., London. 249. Hagenbuch, B., and B. Stieger. 2013. The SLCO (former SLC21)
225. Fröhlicher, E. 1936. Die Resorption von gallensäuren aus ver- superfamily of transporters. Mol. Aspects Med. 34: 396–412.
schiedenen Dunndarmabschnitten. Biochem. Z. 283: 273–279. 250. Klaassen, C. D., and L. M. Aleksunes. 2010. Xenobiotic, bile acid,
226. Baker, R. D., and G. W. Searle. 1960. Bile salt absorption at various and cholesterol transporters: function and regulation. Pharmacol.
levels of rat small intestine. Proc. Soc. Exp. Biol. Med. 105: 521–523. Rev. 62: 1–96.
227. Wilson, T. H., and G. Wiseman. 1954. The use of sacs of everted 251. Cowen, A. E., M. G. Korman, A. F. Hofmann, and P. J. Thomas.
small intestine for the study of the transference of substances 1975. Plasma disappearance of radioactivity after intravenous in-
from the mucosal to the serosal surface. J. Physiol. 123: 116–125. jection of labeled bile acids in man. Gastroenterology. 68: 1567–1573.
228. Crane, R. K. 1960. Intestinal absorption of sugars. Physiol. Rev. 40: 252. Norman, A. 1970. Metabolism of glycocholic acid in man. Scand.
789–825. J. Gastroenterol. 5: 231–236.
229. Weiner, I. M., and L. Lack. 1962. Absorption of bile salts from the 253. Hepner, G. W., A. F. Hofmann, and P. J. Thomas. 1972.
small intestine in vivo. Am. J. Physiol. 202: 155–157. Q27 Metabolism of steroid and amino acid moieties of conjugated bile
230. Holt, P. R. 1964. Intestinal absorption of bile salts in the rat. Am. acids in man. II. Glycine conjugated dihydroxy bile acids. J. Clin.
J. Physiol. 207: 1–7. Invest. 51: 1898–1905.
231. Lücke, H., G. Stange, R. Kinne, and H. Murer. 1978. Taurocholate- 254. Csanaky, I. L., H. Lu, Y. Zhang, K. Ogura, S. Choudhuri, and C.
sodium co-transport by brush-border membrane vesicles isolated D. Klaassen. 2011. Organic anion-transporting polypeptide 1b2
from rat ileum. Biochem. J. 174: 951–958. (Oatp1b2) is important for the hepatic uptake of unconjugated
232. Kramer, W., and G. Kurz. 1983. Photolabile derivatives of bile bile acids: studies in Oatp1b2-null mice. Hepatology. 53: 272–281.
salts. Synthesis and suitability for photoaffinity labeling. J. Lipid 255. Hubbard, B., H. Doege, S. Punreddy, H. Wu, X. Huang, V. K.
Res. 24: 910–923. Kashik, R. L. Mozell, J. J. Bynes, A. Stricker-Krongrad, C. J. Chou,
233. Kramer, W., S. B. Nicol, F. Girbig, U. Gutjahr, S. Kowalewski, and et al. 2006. Mice depleted for fatty acid transport protein 5 have
H. Fasold. 1992. Characterization and chemical modification of defective bile acid conjugation and are protected from obesity.
the Na(+)-dependent bile-acid transport system in brush-border Gastroenterology. 130: 1259–1269.
membrane vesicles from rabbit ileum. Biochim. Biophys. Acta. 1111: 256. Steinberg, S. J., S. J. Mihalik, D. G. Kim, D. A. Cuebas, and P. A.
93–102. Watkins. 2000. The human liver-specific homolog of very long-
234. Heidrich, H-G., R. Kinne, E. Kinne-Saffran, and K. Hannig. 1972. chain acyl-CoA synthetase is cholate:CoA ligase. J. Biol. Chem. 275:
The polarity of the proximal tubule cell in rat kidney. J. Cell Biol. 15605–15608.
54: 232–245. 257. Clayton, L. M., D. Gurantz, A. F. Hofmann, L. R. Hagey, and C.
235. Hediger, M. A., M. Coady, T. S. Ikeda, and E. Wright. 1987. D. Schteingart. 1989. The role of bile acid conjugation in he-
Expression cloning and cDNA sequencing of the Na+/glucose patic transport of dihydroxy bile acids. J. Pharm. Exp. Ther. 248:
cotransporter. Nature. 330: 379–381. 1130–1137.
236. Wong, M. H., P. Oelkers, A. L. Craddock, and P. A. Dawson. 258. Rius, M., J. Hummel-Eisenbeiss, A. F. Hofmann, and D. Keppler.
1994. Expression cloning and characterization of the hamster 2006. Substrate specificity of human ABCC4 (MRP4)-mediated
ileal sodium-dependent bile acid transporter. J. Biol. Chem. 269: cotransport of bile acids and reduced glutathione. Am. J. Physiol.
1340–1347. Gastrointest. Liver Physiol. 290: G640–G649.
237. Wilson, F. A., G. Burckhardt, H. Murer, G. Rumrich, and K. J. 259. Sorscher, S., J. Lillienau, J. L. Meinkoth, J. H. Steinbach, C. D.
Ulrich. 1981. Sodium-coupled taurocholate transport in the prox- Schteingart, J. Feramisco, and A. F. Hofmann. 1992. Conjugated
imal convolution of the rat kidney in vivo and in vitro. J. Clin. bile acid uptake by Xenopus laevis oocytes induced by microinjec-
Invest. 67: 1141–1150. tion with ileal Poly A+ mRNA. Biochem. Biophys. Res. Commun. 186:
238. Alpini, G., S. S. Glaser, R. Rodgers, J. L. Phinizy, W. E. Robertson, 1455–1462.
J. Lasater, A. Caligiuri, Z. Tretjak, and G. D. LeSage. 1997. 260. Keppler, D. 2011. Cholestasis and the role of basolateral efflux
Functional expression of the apical Na+-dependent bile acid trans- pumps. Z. Gastroenterol. 49: 1553–1557.
porter in large, but not small rat cholangiocytes. Gastroenterology. 261. Duane, W. C. 1975. The intermicellar bile salt concentration
113: 1734–1740. in equilibrium with the mixed micelles of human bile. Biochim.
239. Lazaridis, K. N., L. Pham, P. Tietze, R. A. Marinelli, P. C. deGroen, Biophys. Acta. 398: 275–286.
S. Levine, P. A. Dawson, and N. F. LaRusso. 1997. Rat cholangio- 262. Inoue, M., R. Kinne, T. Tran, L. Biempica, and I. M. Arias. 1983.
cytes absorb bile acids at their apical domain via the ileal sodium- Rat liver canalicular membrane vesicles. Isolation and topological
dependent bile acid transporter. J. Clin. Invest. 100: 2714–2721. characterization. J. Biol. Chem. 258: 5183–5188.
240. Kolhatkar, V., and J. E. Polli. 2012. Structural requirements of bile 263. Meier, P. J., E. S. Sztul, A. Reuben, and J. L. Boyer. 1984. Structural
acid transporters: C-3 and C-7 modifications of steroidal hydroxyl and functional polarity of canalicular and basolateral plasma
groups. Eur. J. Pharm. Sci. 46: 86–99. membrane vesicles isolated in high yield from rat liver. J. Cell Biol.
241. Hu, N. J., S. Iwata, A. D. Cameron, and D. Drew. 2011. Crystal 98: 991–1000.
structure of a bacterial homologue of the bile acid sodium sym- 264. Meier, P. J., A. S. Meier-Abt, C. Barrett, and J. L. Boyer. 1984.
porter ASBT. Nature. 478: 408–411. Mechanisms of taurocholate transport in canalicular and basolateral
242. Döring, B., T. Lütteke, J. Geyer, and E. Petzinger. 2012. The rat liver plasma membrane vesicles. Evidence for an electrogenic
SLC10 carrier family: transport functions and molecular struc- canalicular organic anion carrier. J. Biol. Chem. 259: 10614–10622.
ture. Curr. Top. Membr. 70: 105–168. 265. Inoue, M., R. Kinne, T. Tran, and I. M. Arias. 1984. Taurocholate
243. Hoffman, N. E., D. E. Donald, and A. F. Hofmann. 1975. Effect of transport by rat liver canalicular membrane vesicles. J. Clin. Invest.
primary bile acids on bile lipid secretion from the perfused dog 73: 659–663.
liver. Am. J. Physiol. 229: 714–720. 266. O’Máille, E. R., and A. F. Hofmann. 1986. Relatively high biliary
244. Poupon, R., R. Poupon, M. Dumont, and S. Erlinger. 1976. secretory maximum for non-micelle-forming bile acid: possible
Hepatic storage and biliary transport maximum of taurocholate significance for mechanism of secretion. Q. J. Exp. Physiol. 71:
and taurochenodeoxycholate in the dog. Eur. J. Clin. Invest. 6: 475–482.
431–437. 267. Adachi, Y., H. Kobayashi, Y. Kurumi, M. Shouji, M. Kitano, and T.
245. Van Dyke, R. W., J. E. Stephens, and B. F. Scharschmidt. 1982. Yamamoto. 1991. ATP-dependent taurocholate transport by rat
Bile acid transport in cultured hepatocytes. Am. J. Physiol. 243: liver canalicular vesicles. Hepatology. 14: 655–659.
G484–G492. 268. Nishida, T., Z. Gatmaitan, M. Che, and I. M. Arias. 1991. Rat liver
246. Reichen, J., and G. Paumgartner. 1976. Uptake of bile acids by canalicular membrane vesicles contain an ATP-dependent bile
perfused rat liver. Am. J. Physiol. 231: 734–742. acid transport system. Proc. Natl. Acad. Sci. USA. 88: 6590–6594.
247. Hagenbuch, B., B. Stieger, M. Foguet, H. Lubbert, and P. J. Meier. 269. Childs, S., R. L. Yeh, E. Georges, and V. Ling. 1995. Identification
1991. Functional expression cloning and characterization of the of a sister gene to P-glycoprotein. Cancer Res. 55: 2029–2034.

1590 Journal of Lipid Research Volume 55, 2014


270. Gerloff, T., B. Stieger, B. Hagenbuch, J. Madon, L. Landmann, 290. Lehmann, C. G. 1855. Physiological Chemistry. 2nd edition. Vol.
J. Roth, A. F. Hofmann, and P. J. Meier. 1998. The sister of 1. Blanchard and Lea, Philadelphia.
P-glycoprotein represents the canalicular bile salt export pump of 291. Schiff, M. 1870. Bericht über einige Versuchsreihen. I.
mammalian liver. J. Biol. Chem. 273: 10046–10050. Gallenbildung abhängig von der Aufsäugung der Gallstoffe.
271. Strautnieks, S. S., L. N. Bull, A. S. Knisely, S. A. Kocoshis, N. Dahl, Pflugers Arch. Physiol. 3: 598–613.
H. Arnell, E. Sokal, K. Dahan, S. Childs, V. Ling, et al. 1998. A 292. Carey, M. C., and W. C. Duane. 1994. Enterohepatic circulation.
gene encoding a liver-specific ABC transporter is mutated in pro- In The Liver: Pathobiology. 3rd edition. I. M. Arias, J. L. Boyer, N.
gressive familial intrahepatic cholestasis. Nat. Genet. 20: 233–238. Fausto, et al., editors. Raven Press, Ltd., New York.
272. Akita, H., H. Suzuki, K. Ito, S. Kinoshita, N. Sato, H. Takikawa, 293. Back, P. 1988, Urinary bile acids. In The Bile Acids. vol. 4. K. D.
and Y. Sugiyama. 2001. Characterization of bile acid transport me- R. Setchell, D. Kritchevsky, and P. P. Nair, editors. Plenum Press,
diated by multidrug resistance associated protein 2 and bile salt New York. 405–40.
export pump. Biochim. Biophys. Acta. 1511: 7–16. 294. Hofmann, A. F., and N. E. Hoffman. 1974. Measurement of bile acid
273. Wang, R., M. Salem, I. M. Yousef, B. Tuchweber, P. Lam, S. J. kinetics by isotope dilution in man. Gastroenterology. 67: 314–323.
Childs, C. D. Helgason, C. Ackerley, M. J. Phillips, and V. Ling. 295. Stellaard, F., G. Brufau, R. Boverhof, E. Z. Jonkers, T. Boer, and
2001. Targeted inactivation of sister of P-glycoprotein gene F. Kuipers. 2009. Developments in bile acid kinetic measurements
(spgp) in mice results in nonprogressive but persistent intrahe- using (13)C and (2)H: 10(5) times improved sensitivity during
patic cholestasis. Proc. Natl. Acad. Sci. USA. 98: 2011–2016. the last 40 years. Isotopes Environ. Health Stud. 45: 275–288.
274. Wang, R., H. I. Chen, L. Liu, J. A. Sheps, M. J. Phillips, and V. 296. LaRusso, N. F., P. A. Szczepanik, A. F. Hofmann, and S. B. Coffin.
Ling. 2009. Compensatory role of P-glycoproteins in knockout 1977. Effect of deoxycholic acid ingestion on bile acid metabolism
mice lacking the bile salt export pump. Hepatology. 50: 948–956. and biliary lipid secretion in normal subjects. Gastroenterology. 72:
275. Perwaiz, S., D. Forrest, D. Magnault, B. Tuchweber, M. J. Phillips, 132–140.
R. Wang, V. Ling, and I. M. Yousef. 2003. Appearance of atypical 297. van der Werf, S. D., F. M. Nagengast, G. P. van Berge Henegouwen,
3 alpha, 6 beta, 7 beta, 12 alpha-tetrahydroxy-5 beta-cholan-24-oic A. W. Huijbregts, and J. H. van Tongeren. 1982. Colonic absorp-
acid in spgp knockout mice. J. Lipid Res. 44: 494–502. tion of secondary bile-acids in patients with adenomatous polyps
276. Wang, R., L. Liu, J. Sheps, D. Forrest, A. F. Hofmann, L. R. Hagey, and in matched controls. Lancet. 1: 759–762.
and V. Ling. 2013. Defective canalicular transport and toxicity of 298. Bergström, S., M. Rottenberg, and J. Voltz. 1953. The preparation
dietary ursodeoxycholic acid in the Abcb11⫺/⫺ mouse: trans- of some carboxyl-labeled bile acids. Acta Chem. Scand. 7: 481–484.
port and gene expression studies. Am. J. Physiol. Gastrointest. Liver 299. Hofmann, A. F., P. A. Szczepanik, and P. D. Klein. 1968. Rapid
Physiol. 305: G286–G294. preparation of tritium-labeled bile acids by enolic exchange on
277. Megaraj, V., T. Iida, P. Jungsuwadee, A. F. Hofmann, and M. Vore. basic alumina containing tritiated water. J. Lipid Res. 9: 707–713.
2010. Hepatobiliary disposition of 3␣,6␣,7␣.12␣-tetrahydroxy- 300. LaRusso, N. F., N. E. Hoffman, and A. F. Hofmann. 1974. Validity
cholanoyltaurine: a substrate for multiple canalicular transport- of using 2,4-3H-labeled bile acids to study bile acid kinetics in
ers. Drug Metab. Dispos. 38: 1723–1730. man. J. Lab. Clin. Med. 84: 759–765.
278. Wang, R., P. Lam, L. Liu, D. Forrest, I. M. Yousef, D. Mignault, 301. Ng, P. Y., R. N. Allan, and A. F. Hofmann. 1977. Suitability of
M. J. Phillips, and V. Ling. 2003. Severe cholestasis induced by [11,12-3H2]chenodeoxycholic acid and [11,12-3H2]lithocholic
cholic acid feeding in knockout mice of sister of P-glycoprotein. acid for isotope dilution studies of bile acid metabolism in man. J.
Hepatology. 38: 1489–1499. Lipid Res. 18: 753–758.
279. Zhang, Y., F. Li, A. D. Patterson, Y. Wang, K. W. Krausz, G. Neale, 302. Duane, W. C., C. D. Schteingart, H-T. Ton-Nu, and A. F.
S. Thomas, D. Nachagari, P. Vogel, M. Vore, et al. 2012. Abcb11 Hofmann. 1996. Validation of [22,23-3H]cholic acid as a stable
deficiency induces cholestasis coupled to impaired ␤-fatty acid tracer through conversion to deoxycholic acid in human subjects.
oxidation in mice. J. Biol. Chem. 287: 24784–24794. J. Lipid Res. 37: 431–436.
280. Wang, W., D. J. Seward, L. Li, J. L. Boyer, and N. Ballatori. 2001. 303. Usui, T., and K. Yamasaki. 1963. Studies on the enterohepatic cir-
Expression cloning of two genes that together mediate organic culation of bile acid. J. Biochem. (Tokyo). 55: 593–598.
solute and steroid transport in the liver of a marine vertebrate. 304. Go, V. L. W., A. F. Hofmann, and W. H. J. Summerskill. 1970.
Proc. Natl. Acad. Sci. USA. 98: 9431–9436. Simultaneous measurements of total pancreatic, biliary, and gas-
281. Dawson, P. A., M. Hubbert, J. Haywood, A. L. Craddock, N. tric outputs in man using a perfusion technique. Gastroenterology.
Zerangue, W. V. Christian, and N. Ballatori. 2005. The hetero- 58: 321–328.
meric organic solute transporter ␣-␤, Ost␣-Ost␤, is an ileal baso- 305. Brunner, H., T. C. Northfield, V. L. W. Go, and W. H. J.
lateral bile acid transporter. J. Biol. Chem. 280: 6960–6968. Summerskill. 1974. Gastric emptying and secretion of bile ac-
282. Soroka, C. J., N. Ballatori, and J. L. Boyer. 2010. Organic solute ids, cholesterol, and pancreatic enzymes during digestion: duo-
transporter, OSTalpha-OSTbeta: its role in bile acid transport denal perfusion studies in healthy subjects. Mayo Clin. Proc. 49:
and cholestasis. Semin. Liver Dis. 30: 178–185. 851–860.
283. Rao, A., J. Haywood, A. L. Craddock, M. G. Belinsky, G. D. Kruh, 306. van Berge Henegouwen, G. P., and A. F. Hofmann. 1978.
and P. A. Dawson. 2008. The organic solute transporter alpha- Nocturnal gallbladder storage and emptying in gallstone patients
beta, Ostalpha-Ostbeta, is essential for intestinal bile acid trans- and healthy subjects. Gastroenterology. 75: 879–885.
port and homeostasis. Proc. Natl. Acad. Sci. USA. 105: 3891–3896. 307. Northfield, T. C., and A. F. Hofmann. 1973. Biliary lipid secretion
284. Dawson, P. A., J. Haywood, A. L. Craddock, M. Wilson, M. Tietjent, in gallstone patients. Lancet. 1: 747–748.
K. Kluckman, N. Maeda, and J. S. Parks. 2003. Targeted deletion 308. Angelin, B., I. Björkhem, K. Einarsson, and S. Ewerth. 1982.
of the ileal bile acid transporter eliminates enterohepatic cycling Hepatic uptake of bile acids in man. Fasting and postprandial
of bile acids in mice. J. Biol. Chem. 278: 33920–33927. concentrations of individual bile acids in portal venous and sys-
285. Lan, T., J. Haywood, A. Rao, and P. A. Dawson. 2011. Molecular temic blood serum. J. Clin. Invest. 70: 724–731.
mechanisms of altered bile acid homeostasis in organic solute 309. Korman, M. G., N. F. LaRusso, N. E. Hoffman, and A. F. Hofmann.
transporter-alpha knockout mice. Dig. Dis. 29: 18–22. 1975. Development of an intravenous bile acid tolerance test:
286. Frisch, K., S. Jakobsen, M. Sørenson, O. Lajord Munk, A. K. O. plasma disappearance of cholylglycine in health. N. Engl. J. Med.
Alstrup, P. Ott, A. F. Hofmann, and S. Keiding. 2012. [N-methyl- 292: 1205–1209.
11C]cholylsarcosine, a novel bile acid tracer for PET/CT of he- 310. Pries, J. M., C. A. Sherman, G. C. Williams, and R. F. Hanson.
patic excretory function: radiosynthesis and proof-of-concept 1979. Hepatic extraction of bile salts in conscious dog. Am. J.
studies in pigs. J. Nucl. Med. 53: 772–778. Physiol. 236: E191–E197.
287. Häussinger, D., V. Keitel, and R. Kubitz, editors. 2012. 311. van Berge Henegouwen, G. P., and A. F. Hofmann. 1977. Pha-
Hepatobiliary Transport in Health and Disease. Walter de Gruyter rmacology of chenodeoxycholic acid. II. Absorption and metabo-
GmbH and Company, Berlin/Boston. lism. Gastroenterology. 73: 300–309.
288. Borelli, G. 1681. De Motu animalium, Pars altera. Angeli Bernabo, 312. Marigold, J. H., H. J. Bull, I. T. Gilmore, D. J. Coltart, and R. P.
Rome. English translation by P. Gosse, The Hague, Netherlands. H. Thompson. 1982. Direct measurement of hepatic extraction
1743. Reprinted in 1989 by Springer, Berlin. 354–364. of chenodeoxycholic acid and ursodeoxycholic acid in man. Clin.
289. Liebig, J. 1843. Animal chemistry or chemistry in its applications Sci. 63: 197–203.
to physiology and pathology. 2nd edition. Taylor and Walton, 313. Lamri, Y., S. Erlinger, M. Dumont, A. Roda, and G. Feldmann.
London. 1992. Immunoperoxidase localization of ursodeoxycholic acid in

History of bile acid research 1591


rat biliary epithelial cells. Evidence for a cholehepatic circulation. for dissimilar solution and membrane phenomena. J. Lipid Res.
Liver. 12: 351–354. 21: 72–90.
314. Wheeler, H. O., E. D. Ross, and S. E. Bradley. 1968. Canalicular 335. Josephson, B. A. 1933. Studien über die gallensäuren und ange-
bile production in dogs. Am. J. Physiol. 214: 866–874. horige verbindungen. Doctoral Thesis. Royal Caroline Institute
315. Dumont, M., S. Erlinger, and S. Uchman. 1980. Hypercholeresis (Sweden). Centraltryckeriet, Stockholm.
induced by ursodeoxycholic acid and 7-ketolithocholic acid in 336. Ekwall, P., T. Rosendahl, and B. Löfman. 1957. Studies on bile salt
the rat: possible role of bicarbonate transport. Gastroenterology. 79: solutions. I. The dissociation constants of the cholic and deoxy-
82–89. cholic acids. Acta Chem. Scand. 11: 590–599.
316. Fracchia, M., K. D. R. Setchell, A. Crosignani, M. Podda, N. 337. Bell, R. P. 1969. Acids and bases: their quantitative behavior. 2nd
O’Connell, R. Ferraris, A. F. Hofmann, and G. Galatola. 1996. edition. Methuen, London.
Bile acid conjugation in cholestatic liver disease before and dur- 338. Evans, G. F., G. Pye, R. Bramley, A. G. Clark, T. J. Dyson, and J. D.
ing treatment with ursodeoxycholic acid. Clin. Chim. Acta. 248: Hardcastle. 1988. Measurement of gastrointestinal pH profiles in
175–185. normal ambulant human subjects. Gut. 29: 1035–1041.
317. Bolder, U., N. V. Trang, L. R. Hagey, C. D. Schteingart, H-T. 339. Hofmann, A. F., and K. J. Mysels. 1992. Bile acid solubility and
Ton-Nu, C. Cerrè, R. O. Elferink, and A. F. Hofmann. 1999. precipitation in vitro and in vivo: the role of conjugation, pH and
Sulindac is excreted into bile by a canalicular bile salt pump and Ca2+ ions. J. Lipid Res. 33: 617–626.
undergoes a cholehepatic circulation in rats. Gastroenterology. 117: 340. van Berge-Henegouwen, G. P., A. F. Hofmann, and T. S. Gaginella.
962–971. 1977. Pharmacology of chenodeoxycholic acid. I. Pharmaceutical
318. LaRusso, N. F., N. E. Hoffman, M. G. Korman, A. F. Hofmann, properties. Gastroenterology. 73: 291–299.
and A. E. Cowen. 1978. Determinants of fasting and postprandial 341. Go, V. L. W., J. R. Poley, A. F. Hofmann, and W. H. J. Summerskill.
serum bile acid levels in healthy man. Am. J. Dig. Dis. 23: 385–391. 1970. Disturbances of fat digestion induced by acid jejunal pH due
319. Hofmann, A. F., and J. R. Poley. 1972. Role of bile acid malab- to gastric hypersecretion in man. Gastroenterology. 58: 638–646.
sorption in pathogenesis of diarrhea and steatorrhea in patients 342. Spriggs, M., K. A. Thompson, K. Volle, I. Stasiak, L. Beyea, A.
with ileal resection. I. Response to cholestyramine or replacement Guthrie, D. Barton, J. Talley, A. Roda, C. Camborata, et al.
of dietary long chain triglyceride by medium chain triglyceride. 2014. Gastrolithiasis in prehensile-tailed porcupines (Coendou
Gastroenterology. 62: 918–934. Prehensilis): 9 cases and pathogenesis of stone formation. Am. J.
320. Grundy, S. M., E. H. Ahrens, and G. Salen. 1971. Interruption of Vet. Med. In press.
the enterohepatic circulation of bile acids in man: comparative 343. Gu, J-J., A. F. Hofmann, H-T. Ton-Nu, C. D. Schteingart, and K.
effects of cholestyramine and ileal exclusion on cholesterol me- J. Mysels. 1992. Solubility of calcium salts of unconjugated and
tabolism. J. Lab. Clin. Med. 78: 94–121. conjugated natural bile acids. J. Lipid Res. 33: 635–646.
321. Dowling, R. H., E. Mack, and D. M. Small. 1970. Effects of con- 344. Jones, C., A. F. Hofmann, K. J. Mysels, and A. Roda. 1986. The
trolled interruption of the enterohepatic circulation of bile salts effect of calcium and sodium ion concentration on the proper-
by biliary diversion and by ileal resection on bile salt secretion, ties of dilute aqueous solutions of glycine conjugated bile salts. J.
Colloid Interface Sci. 114: 452–470.
synthesis, and pool size in the Rhesus monkey. J. Clin. Invest. 49:
345. Shiffman, M. L., H. J. Sugermen, J. M. Kellum, and E. W. Moore.
232–242.
1992. Calcium in human gallbladder bile. J. Lab. Clin. Med. 120:
322. Pandak, W. M., D. M. Heuman, P. B. Hylemon, J. Y. Chiang, and
875–884.
Z. R. Vlahcevic. 1995. Failure of intravenous infusion of tauro-
346. Hofmann, A. F. 1984. Animal models of calcium cholelithiasis.
cholate to downregulate cholesterol 7␣-hydroxylase in rats with
Hepatology. 4: 209S–211S.
biliary fistulas. Gastroenterology. 108: 533–544.
347. Palmer, R. H., and Z. Hruban. 1966. Production of bile duct
323. Nagano, M., S. Kuroki, A. Mizuta, M. Furukawa, M. Noshiro, K.
hyperplasia and gallstones by lithocholic acid. J. Clin. Invest. 45:
Chiijiwa, and M. Tanaka. 2004. Regulation of bile acid synthesis
1255–1267.
under reconstructed enterohepatic circulation in rats. Steroids. 69: 348. Zaki, F. G., J. B. Carey, F. W. Hofbauer, and C. Nokolo. 1967.
701–709. Biliary reaction and choledocholithiasis induced in the rat by
324. Shang, Q., G. L. Guo, A. Honda, M. Saumoy, G. Salen, and G. Xu. lithocholic acid. 1967. J. Lab. Clin. Med. 69: 737–748.
2013. Gut hepatic signal FGF15/19 protein levels in the portal 349. Cohen, B. I., N. Ayyad, E. H. Mosbach, C. K. McSherry, N. Matoba,
blood do not reflect changes in the ileal FGF 15/19 or hepatic A. F. Hofmann, H-T. Ton-Nu, Y. Peng, C. D. Schteingart, and R. J.
Cyp7A1 mRNA levels. J. Lipid Res. 54: 2606–2614. Stenger. 1991. Replacement of cholesterol gallstones by muride-
325. Lillienau, J., D. L. Crombie, J. Munoz, S. J. Longmire-Cook, L. R. oxycholyl taurine gallstones in prairie dogs fed murideoxycholic
Hagey, and A. F. Hofmann. 1993. Negative feedback regulation of acid. Hepatology. 14: 158–168.
the ileal bile acid transport system in rodents. Gastroenterology. 104: 350. Hofmann, A. F., and H. S. Mekhjian. 1973. Bile acids and the in-
38–46. testinal absorption of fat and electrolytes in health and disease.
326. Sauer, P., A. Stiehl, B. A. Fitscher, H. D. Riedel, C. Benz, P. In The Bile Acids. Vol. 2. P. P. Nair and D. Kritchevsky, editors.
Klöters-Platchky, S. Stengelin, W. Stremmel, and W. Kramer. Plenum Press, New York. 103–152.
2000. Downregulation of ileal bile acid absorption in bile-duct- 351. Hartley, G. S. 1936. Aqueous solutions of paraffin-chain salts.
ligated rats. J. Hepatol. 33: 2–8. A study in micelle formation. In Actualités Scientifiques et
327. Matsumura, J. S., M. A. Greiner, D. L. Nahrwold, and L. G. Dawes. Industrielles, 387. Hermann and Company, Editeurs, Paris.
1993. Reduced ileal taurocholate absorption with total parenteral 352. McBain, E. L., and E. Hutchinson. 1955. Solubilization and
nutrition. J. Surg. Res. 54: 517–522. Related Phenomena: Physical Chemistry. Academic Press, New
328. Hofmann, A. F., editor. 1984. The physical chemistry of bile acids York.
in health and disease. Hepatology. 4(Suppl): 1S–252S. 353. Bashour, J. T., and L. Bauman. 1937. The solubility of cholesterol
329. Hofmann, A. F., and D. M. Small. 1967. Detergent properties of in bile salt solutions. J. Biol. Chem. 121: 1–3.
bile salts: correlation with physiological function. Annu. Rev. Med. 354. Mellander, S., and E. Stenhagen. 1942. The state of bile salt solu-
18: 333–376. tions. II. Conductivity studies on dilute solutions of sodium tauro-
330. Carey, M. C. 1985. Physical-chemical properties of bile acids and cholate at 25° C. Acta Physiol. Scand. 4: 349–361.
their salts. In Sterols and Bile Acids. H. Danielsson and J. Sjövall, 355. Ekwall, P. 1951. Micelle formation in sodium cholate solutions.
editors. Elsevier, Amsterdam. 345–404. Acta Acad. Aboensis Math. Phys. 17: 3–10.
331. Roda, A., and A. Fini. 1984. Effect of nuclear hydroxyl substituents 356. Fontell, K. 1991. In memoriam. P. Ekwall. Colloid Polym. Sci. 269:
on aqueous solubility and acidic strength of bile acids. Hepatology. 419–420.
4(Suppl): 72S–76S. 357. Norman, A. 1960. The beginning solubilization of 20-methylcho-
332. Hofmann, A. F. 1994. Pharmacology of ursodeoxycholic acid, an lanthrene in aqueous solutions of conjugated and unconjugated
enterohepatic drug. Scand. J. Gastroenterol. Suppl. 204: 1–15. bile salts. Acta Chem. Scand. 14: 1300–1309.
333. Small, D. M. 1971. The physical chemistry of cholanic acids. In 358. Hofmann, A. F. 1963. The function of bile salts in fat absorption:
The Bile Acids: Chemistry, Physiology, and Metabolism. P. P. Nair the solvent properties of dilute micellar solutions of conjugated
and D. Kritchevsky, editors. Plenum Press, New York. 249–356. bile salts. Biochem. J. 89: 57–68.
334. Igimi, H., and M. C. Carey. 1980. pH-solubility relations of che- 359. Kratohvil, J. P. 1986. Size of bile salt micelles: techniques, prob-
nodeoxycholic and ursodeoxycholic acids: physical-chemical basis lems, and results. Adv. Colloid Interface Sci. 26: 131–154.

1592 Journal of Lipid Research Volume 55, 2014


360. Roda, A., A. F. Hofmann, and K. J. Mysels. 1983. The influence of 384. 1989. Biliary cholesterol transport and precipitation. Hepatology
bile salt structure on self association in aqueous solutions. J. Biol. Suppl. S. M. Strasberg and A. F. Hofmann, editors. 1S–244S.
Chem. 258: 6362–6370. 385. Strasberg, S. M., and P. R. C. Harvey. 1990. Biliary cholesterol
361. Iida, T., and F. C. Chang. 1982. Potential bile acid metabolites. 6. transport and precipitation: Introduction and overview of confer-
Stereoisomeric 3,7-dihydroxy-5␤-cholanoic acids. J. Org. Chem. 47: ence. Hepatology. 12: 1S–5S.
2966–2972. 386. Donovan, J. M., and M. C. Carey. 1990. Separation and quantita-
362. Fickert, P., A. Fuchsbichler, M. Wagner, G. Zollner, A. Kaser, H. tion of cholesterol carriers in bile. Hepatology. 12: 94S–104S.
Tilg, R. Krause, F. Lammert, C. Langner, K. Zatloukal, et al. 2004. 387. Schriever, C. E., and D. Jüngst. 1989. Association between choles-
Regurgitation of bile acids from leaky bile ducts causes sclerosing terol-phospholipid vesicles and cholesterol crystals in human bile.
cholangitis in Mdr2 (Abcb4) knockout mice. Gastroenterology. 127: Hepatology. 9: 541–546.
261–274. 388. Lee, P. H., W. I. Higuchi, Y. Adachi, and N. A. Mazer. 1992.
363. Poupon, R., O. Rosmorduc, P. Y. Boelle, Y. Chretien, C. Mechanisms of cholesterol monohydrate dissolution in aqueous
Corbechot, O. Chazouilleres, C. Housset, and V. Barbu. 2013. taurocholate, taurochenodeoxycholate, and tauroursodeoxycho-
Genotype-phenotype relationships in the low-phospholipid as- late-lecithin solutions: correlation between micellar species and
sociated cholelithiasis syndrome: a study of 156 consecutive pa- dissolution rates. J. Colloid Interface Sci. 154: 35–50.
tients. Hepatology. 58: 1105–1110. 389. Hegardt, F. G., and H. Dam. 1971. The solubility of cholesterol in
364. Polonovski, M., and R. Bourillon. 1952. Les phospholipids de la aqueous solutions of bile salts and lecithin. Z. Ernahrungswiss. 10:
bile. Bull. Str. Chim. Biol. 34: 712–719. 223–233.
365. Phillips, G. B. 1960. The lipid composition of human bile. Biochim. 390. Holzbach, R. T., M. Marsh, M. Olszewski, and K. Holan. 1973.
Biophys. Acta. 41: 361–363. Cholesterol solubility in bile: evidence that supersaturated bile is
366. Isaksson, B. 1954. On the dissolving power of lecithin and bile frequent in healthy man. J. Clin. Invest. 52: 1467–1479.
salts for cholesterol in human bladder bile. Acta Soc. Med. Ups. 59: 391. Dam, H., I. Prange, and E. Sondergaard. 1972. Alimentary produc-
296–306. tion of gallstones in hamsters. XXIV. Influence of orally ingested
367. Yoshimuta, S. 1960. Factors affecting solubility of cholesterol in chenodeoxycholic acid and hyodeoxycholic acid on formation of
bile. Fukuoka Acta Med. 51: 510–528. gallstones. Z. Ernahrungswiss. 11: 80–94.
368. Tamesue, N., T. Inoue, and K. Juniper. 1973. Solubility of cho- 392. Dam, H., I. Kruse, I. Prange, H. E. Kallehauge, H. J. Fenger, and
lesterol in bile salt-lecithin model systems. Am. J. Dig. Dis. 18: M. Krogh Jensen. 1971. Studies on human bile. III. Composition
670–678. of duodenal bile from healthy young volunteers compared with
369. Small, D. M., S. A. Penkett, and D. Chapman. 1969. Studies on composition of bladder bile from surgical patients with and
simple and mixed bile salt micelles by nuclear magnetic reso- without uncomplicated gallstone disease. Z. Ernahrungswiss. 10:
nance spectroscopy. Biochim. Biophys. Acta. 176: 178–189. 160–177.
370. Lindblom, G., P-O. Eriksson, and G. Arvidson. 1984. Molecular 393. Carey, M. C. 1978. Critical tables for calculating the cholesterol
organization in phases of lecithin-cholate-water as studied by nu- saturation of native bile. J. Lipid Res. 19: 945–955.
clear magnetic resonance. Hepatology. 4: 129S–133S. 394. Carey, M. C., and G. Ko. 1979. The importance of total lipid con-
371. Nichols, J. W., and J. Ozarowski. 1990. Sizing of lecithin-bile salt centration in determining cholesterol solubility in bile and the
mixed micelles by size-exclusion high performance liquid chro- development of critical tables for calculating “percent cholesterol
matography. Biochemistry. 29: 4600–4606. saturation” with a correction factor for ursodeoxycholate-rich
372. Hjelm, R. P., P. Thiyagarajan, and H. Alkan-Onyuksel. 1992. bile. In Biological Effects of Bile Acids. G. Paumgartner, A. Stiehl,
Organization of phosphatidylcholine and bile salt in rodlike and W. Gerok, editors. MTP Press, Lancaster, UK. 299–308.
mixed micelles. J. Phys. Chem. 96: 8653–8661. 395. Mattson, F. H., and L. W. Beck. 1956. The specificity of pancreatic
373. Marrink, S. J., and A. E. Mark. 2002. Molecular dynamics simu- lipase for the primary hydroxyl groups of glycerides. J. Biol. Chem.
lation of mixed micelles modeling human bile. Biochemistry. 41: 219: 735–740.
5375–5382. 396. Mattson, F. H., and R. A. Volpenhein. 1964. The digestion and
374. Long, M. A., E. W. Kaler, and S. P. Lee. 1994. Structural charac- absorption of triglycerides. J. Biol. Chem. 239: 2772–2777.
terization of the micelle-vesicle transition in lecithin-bile salt solu- 397. Hofmann, A. F., and B. Borgström. 1963. Hydrolysis of long chain
tions. Biophys. J. 67: 1733–1742. monoglycerides in micellar solution by pancreatic lipase. Biochim.
375. Heuman, D. M., R. S. Bajaj, and Q. Lin. 1996. Adsorption of mix- Biophys. Acta. 70: 317–331.
tures of bile salt taurine conjugates to lecithin-cholesterol mem- 398. Svärd, M., P. Schurtenberger, K. Fontell, B. Jonsson, and B.
branes: implications for bile salt toxicity and cytoprotection. J. Lindman. 1988. Micelles, vesicles, and liquid crystals in the mon-
Lipid Res. 37: 562–573. olein-sodium taurocholate-water system: phase behavior, NMR,
376. Admirand, W. H., and D. M. Small. 1968. The physicochemical self-diffusion, and quasi-elastic light scattering studies. J. Phys.
basis of cholesterol gallstone formation in man. J. Clin. Invest. 47: Chem. 92: 2261–2270.
1043–1052. 399. Hofmann, A. F., and B. Borgström. 1964. The intraluminal phase
377. Rozeboom, H. W. B. 1894. Graphische darstellung der hetero- of fat digestion in man: the lipid content of the micellar and oil
genen systeme aus ein bis vier stoffen, mit einschluss der che- phases of intestinal content obtained during fat digestion and ab-
mischen umsetzung. Ztrschr. F. Physik. Chemie. 15: 145–158. sorption. J. Clin. Invest. 43: 247–257.
378. Ahrens, E. H. 1957. Seminar on atherosclerosis: nutritional fac- 400. Staggers, J. E., O. Hernell, R. J. Stafford, and M. C. Carey. 1990.
tors and serum lipid levels. Am. J. Med. 23: 928–952. Physical-chemical behavior of dietary and biliary lipids during intes-
379. Mazer, N. A., G. B. Benedek, and M. C. Carey. 1980. Quasielastic tinal digestion and absorption: 1. Phase behavior and aggregation
light-scattering studies of aqueous biliary lipid systems. Mixed states of model lipid systems patterned after aqueous duodenal con-
micelle formation in bile salt-lecithin solutions. Biochemistry. 19: tents of healthy adult human beings. Biochemistry. 29: 2028–2040.
601–615. 401. Hernell, O., J. E. Staggers, and M. C. Carey. 1990. Physical-
380. Mazer, N. A., and M. C. Carey. 1983. Quasielastic light scatter- chemical behavior of dietary and biliary lipids during intestinal
ing studies of aqueous biliary lipid systems. Cholesterol solubili- digestion and absorption.2. Phase analysis and aggregation states
zation and precipitation in model bile solutions. Biochemistry. 22: of luminal lipids during duodenal fat digestion in healthy adult
426–442. human beings. Biochemistry. 29: 2041–2056.
381. Sömjen, G. J., R. Rosenberg, and T. Gilat. 1990. Gel filtration and 402. Lawrence, A. S. C. 1959. The mechanism of detergence. Nature.
quasielastic light scattering studies of human bile. Hepatology. 12: 103: 1491–1494.
123S–128S. 403. Hjelm, R. P., C. D. Schteingart, A. F. Hofmann, and D. S. Silvia.
382. Halpern, Z., M. A. Dudley, M. P. Lynn, J. M. Nader, A. C. Breuer, 1995. Form and structure of self-assembling particles in mono-
and R. T. Holzbach. 1986. Vesicle aggregation in model systems of olein-bile salt mixtures. J. Phys. Chem. 99: 16395–16400.
supersaturated bile: relation to crystal nucleation and lipid com- 404. Hjelm, R. P., C. D. Schteingart, A. F. Hofmann, and P.
position of the vesicular phase. J. Lipid Res. 27: 295–306. Thiyagarajan. 2000. Structure of conjugated bile salt-fatty acid-
383. Higuchi, W. I., C-L. Liu, Y. Adachi, N. A. Mazer, and P. H. Lee. monoglyceride mixed colloids: studies by small-angle neutron
1990. Equilibrium dialysis studies on aqueous taurocholate-leci- scattering. J. Phys. Chem. B. 104: 197–211.
thin solutions: further validation of the method. Hepatology. 12: 405. Annegers, J. H. 1954. Function of pancreatic juice and bile in as-
45S–49S. similation of dietary triglyceride. AMA Arch. Intern. Med. 93: 9–22.

History of bile acid research 1593


406. Porter, H. P., D. R. Saunders, G. Tytgat, O. Brunser, and C. D. 429. Miyai, K., N. B. Javitt, N. Gochman, H. M. Jones, and D. Baker,
Rubin. 1971. Fat absorption in bile fistula man. A morphological 1982. Hepatotoxicity of bile acids in rabbits: ursodeoxycholic acid
and biochemical study. Gastroenterology. 60: 1008–1019. is less toxic than chenodeoxycholic acid. Lab. Invest. 46: 428–437.
407. Lucassen, J. 1966. Hydrolysis and precipitates in carboxylate soap 430. Wolpers, C., and A. F. Hofmann. 1993. Solitary versus multiple
solutions. J. Phys. Chem. 70: 1824–1830. cholesterol gallbladder stones. Clin. Investig. 71: 423–434.
408. Heydorn, S., P. B. Jeppesen, and P. B. Mortensen. 1999. Bile acid 431. Petroni, M. L., R. P. Jazrawi, P. Pazzi, M. Zuin, A. Lanzini, M.
replacement therapy with cholylsarcosine for short-bowel syn- Fracchia, D. Facchinetti, V. Alvisi, R. Ferraris, J. M. Bland, et al.
drome. Scand. J. Gastroenterol. 34: 818–823. 2000. Risk factors for the development of gallstone recurrence
409. Harkins, R. W., C. H. Whiteside, H. B. Fluckiger, and H. P. Sarett. following medical dissolution. The British-Italian Gallstone Study
1965. Fat utilization in rats fed cholestyramine, a bile acid seques- Group. Eur. J. Gastroenterol. Hepatol. 12: 695–700.
trant. Proc. Soc. Exp. Biol. Med. 118: 399–402. 432. Ammon, H. V., and A. F. Hofmann. 1983. The Langenbuch pa-
410. Johnston, C. G., and F. Nakayama. 1957. Solubility of choles- per. I. An historical perspective and comments of the translators.
terol and gallstones in metabolic material. AMA Arch. Surg. 75: Gastroenterology. 85: 1426–1433.
436–442. 433. Ransohoff, D. F., and W. A. Gracie. 1990. Management of patients
411. Nakayama, F. 1997. Cholelithiasis: Causes and Treatment. Igaku- with symptomatic gallstones: a quantitative analysis. Am. J. Med.
Shoin, Tokyo. 88: 154–160.
412. Heywood, R., A. K. Palmer, C. V. Foll, and M. R. Lee. 1973. Letter: 434. Isselbacher, K. J. 1981. Chenodiol for gallstones: dissolution or
pathological changes in fetal rhesus monkey induced by oral che- disillusion? Ann. Intern. Med. 95: 377–379.
nodeoxycholic acid. Lancet. 302: 1021. 435. Sackmann, M., J. Pauletski, T. Sauerbruch, T. Holl, G. Schelling,
413. Bell, G. D., H. Y. I. Mok, M. Thwe, G. M. Murphy, K. Henry, and R. and G. Paumgartner. 1991. The Munich Gallbladder Lithotripsy
H. Dowling. 1974. Liver structure and function in cholelithiasis: Study. Results of the first five years with 711 patients. Ann. Intern.
effect of chenodeoxycholic acid. Gut. 15: 165–172. Med. 114: 290–296.
414. Cowen, A. E., M. G. Korman, A. F. Hofmann, and O. W. Cass. 1975. 436. Schoenfield, L. J., G. Berci, R. L. Carnovale, W. Casarella, P.
Metabolism of lithocholate in healthy man. I. Biotransformation Caslowitz, D. Chumley, R. C. Davis, J. Y. Gillenwater, A. C. Johnson,
and biliary excretion of intravenously administered lithocholate, R. S. Jones, et al. 1990. The effect of ursodiol on the efficacy and
lithocholylglycine, and their sulfates. Gastroenterology. 69: 59–66. safety of extracorporeal shock-wave lithotripsy of gallstones. The
415. Allan, R. N., J. L. Thistle, and A. F. Hofmann. 1976. Lithocholate Dornier National Biliary Lithotripsy Study. N. Engl. J. Med. 323:
metabolism during chemotherapy for gallstone dissolution. II. 1239–1245.
Absorption and sulphation. Gut. 17: 413–419. 437. Thistle, J. L., G. R. May, C. E. Bender, H. J. Williams, A. J. LeRoy,
416. Gadacz, T. R., R. N. Allan, E. Mack, and A .F. Hofmann. 1976. P. E. Nelson, C. J. Peine, B. T. Petersen, and J. E. McCullough.
Impaired lithocholate sulfation in the rhesus monkey: a possible 1989. Dissolution of cholesterol gallbladder stones by methyl tert-
mechanism for chenodeoxycholate toxicity. Gastroenterology. 70: butyl ether administered by percutaneous transhepatic catheter.
1125–1129. N. Engl. J. Med. 320: 633–639.
417. Webster, K. H., M. C. Lancaster, A. F. Hofmann, D. R. Wease, 438a. Cheslyn-Curtis, S., W. R. Lees, A. R. Hatfield, and R. C. Russell.
and A. H. Baggenstoss. 1975. Influence on primary bile acid feed- 1992. Percutaneous techniques for the management of symptom-
atic gallbladder stones. Dig. Dis. 10: 208–217.
ing on cholesterol metabolism and hepatic function in the rhesus
438b. Holl, J., T. Sauerbruch, M. Sackmann, J. Pauletzski, and G.
monkey. Mayo Clin. Proc. 50: 134–138.
Paumgartner. 1991. Combined treatment pf symptomatic gallblad-
418. Fischer, C. D., N. S. Cooper, M. A. Rothschild, and E. H. Mosbach.
der stones by extracorporeal shock-wave lithotripsy (ESWL) and in-
1974. Effect of dietary chenodeoxycholic acid and lithocholic acid
stillation of methyl tert-butyl ether. Dig. Dis. Sci. 36: 1097–1101.
in the rabbit. Am. J. Dig. Dis. 19: 877–886.
439. Zakko, S. F., and A. F. Hofmann. 1990. Microprocessor-assisted
419. Dyrszka, H., T. Chen, G. Salen, and E. H. Mosbach. 1975. Toxicity
solvent transfer system for effective contact dissolution of gall-
of chenodeoxycholic acid in the rhesus monkey. Gastroenterology.
bladder stones. IEEE Trans. Biomed. Eng. 37: 410–416.
69: 333–337.
440. Esch, O., C. D. Schteingart, D. Pappert, D. Kirby, R. Streich, and
420. Morrissey, K. P., C. K. McSherry, R. L. Swarm, W. H. Nieman, and
A. F. Hofmann. 1993. Increased blood levels of methyl tert-butyl
J. E. Deitrick. 1975. Toxicity of chenodeoxycholic acid in the non-
ether but not of ethyl propionate during gallbladder instillation
human primate. Surgery. 77: 851–860.
421. Schoenfield, L. J., and J. M. Lachin. 1981. Chenodiol (cheno- with contact gallstone dissolution agents in the pig. Hepatology. 18:
deoxycholic acid) for dissolution of gallstones: the National 373–379.
Cooperative Gallstone Study. A controlled trial of efficacy and 441. Hofmann, A. F., A. Amelsberg, O. Esch, C. D. Schteingart, K.
safety. Ann. Intern. Med. 95: 257–282. Lyche, H. Jinich, E. VanSonnenberg, and H. B. D’Agostino. 1997.
422. Palmer, R. H., and M. C. Carey. 1982. Sounding Board. An opti- Successful topical dissolution of cholesterol gallbladder stones us-
mistic view of the National Cooperative Gallstone Study. N. Engl. ing ethyl propionate. Dig. Dis. Sci. 42: 1274–1282.
J. Med. 306: 1171–1174. 442. Phillips, S., R. B. Palmer, and A. Brody. 2008. Epidemiology, toxi-
423. Fisher, R. L., A. F. Hofmann, J. L. Converse, S. S. Rossi, and S. P. cokinetics, and health effects of methyl tert-butyl ether (MTBE).
Lan. 1991. Lack of relationship between hepatotoxicity and litho- J. Med. Toxicol. 4: 115–126.
cholic acid sulfation during chenodiol therapy in the National 443. Esch, O., J. C. Spinosa, R. L. Hamilton, D. L. Crombie, C. D.
Cooperative Gallstone Study. Hepatology. 14: 454–463. Schteingart, J. F. Rondinone, H. B. D’Agostino, J. Lillienau, and
424. Erlinger, S., A. Le Go, J. M. Husson, and J. Fevery. 1984. A. F. Hofmann. 1992. Acute effects on gallbladder histology of
Franco-Belgian cooperative study if ursodeoxycholic acid in the topical methyl tert-butyl ether or ethyl propionate in animals: a
medical dissolution of gallstones: a double-blind randomized comparison of two solvents for contact dissolution of cholesterol
dose-response study and comparison with chenodeoxycholic acid. gallstones. Hepatology. 16: 984–991.
Hepatology. 4: 308–314. 444. Ponchon, T., J. Baroud, B. Pujol, P. J. Valette, and D. Perrot. 1988.
425. Roda, E., D. Festi, A. Roda, C. Sama, R. Aldini, and G. Mazzella. Renal failure during dissolution of gallstones by methyl-tert-butyl
1977. Results of chenic acid therapy in patients with cholesterol ether. Lancet. 2: 276–277.
calculi [article in Italian]. Minerva Med. 68: 3027–3029. 445. Hofmann, A. F., C. D. Schteingart, E. vanSonnenberg, O. Esch,
426. Higuchi, W. I., F. Sjuib, D. Mufson, A. P. Simonelli, and A. F. and S. F. Zakko. 1991. Contact dissolution of cholesterol gallstones
Hofmann. 1973. Dissolution kinetics of gallstones: physical model with organic solvents. Gastroenterol. Clin. North Am. 20: 183–199.
approach. J. Pharm. Sci. 62: 942–945. 446. Clayton, P. T. 2011. Disorders of bile acid synthesis. J. Inherit.
427. Senior, J. R., M. F. Johnson, D. M. DeTurck, F. Bazzoli, and E. Metab. Dis. 34: 593–604.
Roda. 1990. In vivo kinetics of radiolucent gallstones by oral dihy- 447. Mekjian, H. S., S. F. Phillips, and A. F. Hofmann. 1971. Colonic
droxy bile acids. Gastroenterology. 99: 243–251. secretion of water and electrolytes induced by bile acids: perfu-
428. Sarva, R. P., H. Fromm, S. Farivar, R. F. Sembrat, H. Mendelow, H. sion studies in man. J. Clin. Invest. 50: 1569–1577.
Shinozuka, and S. K. Wolfson. 1980. Comparison of the effects be- 448. Keating, N., and S. J. Keely. 2009. Bile acids in regulation of intes-
tween ursodeoxycholic and chenodeoxycholic acid on liver func- tinal physiology. Curr. Gastroenterol. Rep. 11: 375–382.
tion and structure and on bile composition in the rhesus monkey. 449. Kelly, O. B., M. S. Mroz, J. B. Ward, C. Colliva, M. Scharl, R.
Gastroenterology. 79: 629–636. Pellicciari, J. F. Gilmer, P. G. Fallon, A. F. Hofmann, A. Roda,

1594 Journal of Lipid Research Volume 55, 2014


et al. 2013. Ursodeoxycholic acid attenuates colonic epithelial se- 466. Kamen, M. D. 1963. The early history of carbon-14. J. Chem. Educ.
cretory function. J. Physiol. 591: 2307–2318. 40: 234–242.
450. Ao, M., J. Sarathy, J. Dominique, W. A. Airefai, and M. C. Rao. 467. Lindor, K. D. 2011. Farnesoid X receptor agonists for primary bili-
2013. Chenodeoxycholic acid stimulates Cl(-) secretion via cAMP ary cirrhosis. Curr. Opin. Gastroenterol. 27: 285–288.
signaling and increases cystic fibrosis transmembrane conduc- 468. Clinical trials.gov. 2010. The farnesoid X receptor (FXR) ligand
tance regulator phosphorylation in T84 cells. Am. J. Physiol. Cell obeticholic acid in NASH treatment trial (FLINT). ClinicalTrials.
Physiol. 305: C447–C456. gov identifier NCT01265498.
451. Hofmann, A. F., and J. R. Poley. 1969. Cholestyramine treatment 469. Fickert, P., M. J. Pollheimer, D. Silbert, T. Moustafa, E. Halilbasic,
of diarrhea associated with ileal resection. N. Engl. J. Med. 281: E. Krones, F. Durchschein, A. Thüringer, G. Zollner, H. Denk,
397–402. et al.. 2013. Differential effects of norUDCA and UDCA in ob-
452. Walters, J. R., and S. S. Pattni. 2010. Managing bile acid diarrhea. structive cholestasis in mice. J. Hepatol. 58: 1201–1208.
Therap. Adv. Gastroenterol. 3: 349–357. 470. Huff, M. W., D. E. Telford, J. Y. Edwards, J. R. Burnett, P. H.
453. Walters, J. R., A. M. Tasleern, O. S. Omer, W. G. Brydon, T. Dew, Barrett, S. R. Rapp, N. Napawan, and B. T. Keller. 2002. Inhibition
and C. W. Le Roux. 2009. A new mechanism for bile salt diar- of the apical sodium-dependent bile acid transporter reduces
rhea: defective feedback inhibition of bile acid biosynthesis. Clin. LDL cholesterol and apoB by enhanced plasma clearance of LDL
Gastroenterol. Hepatol. 7: 1189–1194. apoB. Arterioscler. Thromb. Vasc. Biol. 22: 1884–1891.
454. Wedlake, L., R. A’Hern, D. Russell, K. Thomas, and J. R. Walters. 471. Chen, L., X. Yao, A. Young, J. McNulty, D. Anderson, Y. Liu, C.
2009. Systematic review: the prevalence of idiopathic bile acid Nystrom, D. Croom, S. Ross, J. Collins, et al. 2012. Inhibition of
malabsorption as diagnosed by SeHCAT scanning in patients apical sodium-dependent bile acid transporter as a novel treat-
with diarrhea predominant irritable bowel syndrome. Aliment. ment for diabetes. Am. J. Physiol. Endocrinol. Metab. 302: E68–E76.
Pharmacol. Ther. 30: 707–717. 472. Simrén, M., A. Bajor, P. G. Gillberg, M. Rudling, and H.
455. Axelson, M., A. Aly, and J. Sjövall. 1988. Levels of 7 alpha-hydroxy- Abrahamsson. 2011. Randomised clinical trial: the ileal bile acid trans-
4-cholesten-3-one in plasma reflect rates of bile acid synthesis in porter inhibitor A3309 vs. placebo in patients with chronic idiopathic
man. FEBS Lett. 239: 324–328. constipation – a double blind study. Aliment. Pharmacol. Ther. 34: 41–50.
456. Sauter, G., F. Berr, U. Beuers, S. Fischer, and G. Paumgartner. 473. Hagey, L. R., N. Vidal, A. F. Hofmann, and M. D. Krasowski. 2010.
1996. Serum concentrations of 7alpha-hydroxy-4-cholesten-3-one Complex evolution of bile salts in birds. Auk. 127: 820–831.
reflect bile acid synthesis in humans. Hepatology. 24: 123–126. 474. Lee, S. P., R. Lester, and J. S. Pyrek. 1987. Vulpecholic acid (1
457. Vijayvargiya, P., M. Camilleri, A. Shin, and A. Saenger. 2013. alpha, 3 alpha, 7 alpha-trihydroxy-5-beta-cholan-24-oic acid): a
Methods for diagnosis of bile acid malabsorption in clinical prac- novel bile acid of a marsupial, Trichosurus vulpecula (Lesson). J.
tice. Clin. Gastroenterol. Hepatol. 11: 1232–1239. Lipid Res. 28: 19–31.
458. Shin, A., M. Camilleri, P. Vijayvargiya, I. Busciglio, D. Burton, M. 475. Hagey, L. R., C. D. Schteingart, H-T. Ton-Nu, and A. F. Hofmann.
Ryks, D. Rhoten, A. Lueke, A. Saenger, A. Girtman, et al. 2013. 1994. Biliary bile acids of fruit pigeons and doves (Columbiformes):
Bowel functions, fecal unconjugated primary and secondary presence of 1␤-hydroxychenodeoxycholic acid and conjugation
bile acids and colonic transit in patients with irritable bowel syn- with glycine as well as taurine. J. Lipid Res. 35: 2041–2048.
drome. Clin. Gastroenterol. Hepatol. 11: 1270–1275. 476. Murai, T., R. Mahara, T. Kurosawa, A. Kimura, and M. Tohma.
459. Okuda, K. I., and I. Björkhem. 1996. Molecular cloning and ex- 1997. Determination of fetal bile acids in biological fluids from
pression of rabbit sterol 12alpha-hydroxylase. J. Biol. Chem. 271: neonates by gas chromatography-negative ion chemical ionization
32269–32275. mass spectrometry. J. Chromatogr. B Biomed. Sci. Appl. 691: 13–22.
460. Higuchi, H., and G. J. Gores. 2003. Bile acid regulation of he- 477. Bi, D., X. Y. Chai, Y. L. Song, Y. Lei, and P. F. Tu. 2009. Novel bile
patic physiology. IV. Bile acids and death receptors. Am. J. Physiol. acids from bear bile power and bile of geese. Chem. Pharm. Bull.
Gastrointest. Liver Physiol. 284: G734–G738. (Tokyo). 57: 528–531.
461. Sokol, R. J., M. S. Straka, R. Dahl, M. W. Devereaux, and B. 478. Kakiyama, G., T. Iida, T. Goto, N. Mano, J. Goto, T. Nambara,
Yerushalmi. 2001. Role of oxidant stress in the permeability tran- L. R. Hagey, and A. F. Hofmann. 2006. Identification of a novel
sition induced in rat hepatic mitochondria by hydrophobic bile bile acid in swans, tree ducks, and geese: 3␣,7␣,15␣-trihydroxy-5␤-
acids. Pediatr. Res. 49: 519–531. cholan-24-oic acid. J. Lipid Res. 47: 1551–1558.
462. Amaral, J. D., R. J. Viana, R. M. Ramalho, C. J. Steer, and C. M. P. 479. Kurosawa, T., Y. Nomura, R. Mahara, T. Yoshimura, A. Kimura, S.
Rodrigues. 2009. Bile acids: regulation of apoptosis by ursodeoxy- Ikegawa, and M. Tohma. 1995. Synthesis of 19-hydroxylated bile
cholic acid. J. Lipid Res. 50: 1721–1734. acids and identification of 3 alpha, 7 alpha, 12 alpha, 19-tetrahy-
463. Allen, K., H. Jaeschke, and B. L. Kopple. 2011. Bile acids in- droxy-5-beta-cholan-24-oic acid in human neonatal urine. Chem.
duce inflammatory genes in hepatocytes: a novel mechanism of Pharm. Bull. (Tokyo). 43: 1551–1557.
inflammation during obstructive cholestasis. Am. J. Pathol. 178: 480. Schiff, L., J. B. Carey, and J. Dietschy. 1969. Bile Salt Metabolism.
175–186. Thomas, Springfield, IL.
464. Gustafsson, B. E., J. A. Gustafsson, and J. Sjövall. 1966. Intestinal 481. Schteingart, C. D., L. R. Hagey, K. D. R. Setchell, and A. F.
and fecal sterols in germfree and conventional rats. Acta Chem. Hofmann. 1993. 5␤-Hydroxylation by the liver: Identification of
Scand. 20: 1827–1835. 3,5,7-trihydroxy nor-bile acids as new major biotrans-formation
465. Devkota, S., Y. Wang, M. W. Musch, V. Leone, H. Fehlner-Peach, products of 3,7-dihydroxy nor-bile acids in rodents. J. Biol. Chem.
A. Nadimpalli, D. A. Antonopoulos, B. Jabri, and E. B. Chang. 268: 11239–11246.
2012. Dietary-fat-induced taurocholic acid promotes pathobiont 482. Reuben, A. 2005. The biliary cycle of Moritz Schiff. Hepatology 42:
expansion and colitis in IL10⫺/⫺ mice. Nature. 487: 104–108. 500–505.

History of bile acid research 1595

Potrebbero piacerti anche