ArticlePDF Available

A genome-wide association scan identifies the hepatic cholesterol transporter ABCG8 as a susceptibility factor for human gallstone disease

Authors:

Abstract and Figures

With an overall prevalence of 10-20%, gallstone disease (cholelithiasis) represents one of the most frequent and economically relevant health problems of industrialized countries. We performed an association scan of >500,000 SNPs in 280 individuals with gallstones and 360 controls. A follow-up study of the 235 most significant SNPs in 1,105 affected individuals and 873 controls replicated the disease association of SNP A-1791411 in ABCG8 (allelic P value P(CCA) = 4.1 x 10(-9)), which was subsequently attributed to coding variant rs11887534 (D19H). Additional replication was achieved in 728 German (P = 2.8 x 10(-7)) and 167 Chilean subjects (P = 0.02). The overall odds ratio for D19H carriership was 2.2 (95% confidence interval: 1.8-2.6, P = 1.4 x 10(-14)) in the full German sample. Association was stronger in subjects with cholesterol gallstones (odds ratio = 3.3), suggesting that His19 might be associated with a more efficient transport of cholesterol into the bile.
Content may be subject to copyright.
A genome-wide association scan identifies the hepatic
cholesterol transporter ABCG8 as a susceptibility factor
for human gallstone disease
Stephan Buch
1–3,13
, Clemens Schafmayer
3,4,13
, Henry Vo
¨
lzke
5
, Christian Becker
6,7
, Andre Franke
2
,
Huberta von Eller-Eberstein
3
, Christian Kluck
6,7
, Ingelore Ba
¨
ssmann
6,7
, Mario Brosch
1
, Frank Lammert
8
,
Juan Francisco Miquel
9
, Flavio Nervi
9
, Michael Wittig
2
, Dieter Rosskopf
10
, Birgit Timm
3
, Christine Ho
¨
ll
3
,
Marcus Seeger
1
, Abdou ElSharawy
2
,TimLu
11
, Jan Egberts
4
, Fred Fa
¨
ndrich
4
, Ulrich R Fo
¨
lsch
1
,
Michael Krawczak
3,11
, Stefan Schreiber
2,3
, Peter Nu
¨
rnberg
6,12
,Ju
¨
rgen Tepel
4
& Jochen Hampe
1
With an overall prevalence of 10–20%, gallstone disease
(cholelithiasis) represents one of the most frequent and
economically relevant health problems of industrialized
countries
1,2
. We performed an association scan of
4500,000 SNPs in 280 individuals with gallstones and
360 controls. A follow-up study of the 235 most significant
SNPs in 1,105 affected individuals and 873 controls replicated
the disease association of SNP A-1791411 in ABCG8
(allelic P value P
CCA
¼ 4.1 10
–9
), which was subsequently
attributed to coding variant rs11887534 (D19H). Additional
replication was achieved in 728 German (P ¼ 2.8 10
–7
)
and 167 Chilean subjects (P ¼ 0.02). The overall odds ratio
for D19H carriership was 2.2 (95% confidence interval:
1.8–2.6, P ¼ 1.4 10
–14
) in the full German sample.
Association was stronger in subjects with cholesterol
gallstones (odds ratio ¼ 3.3), suggesting that His19 might
be associated with a more efficient transport of cholesterol
into the bile.
Genetic susceptibility to gallstones was first recognized in the 1930s
(ref. 3). Both familial clustering
4
and an increased concordance rate of
the trait in monozygotic twins
5
have since been reported. Genome-
wide studies of gallstone susceptibility in inbred mouse strains
established a number of Lith loci
6–9
. In humans, a genome-wide
linkage analysis of Mexican American families identified a gallstone
susceptibility locus on chromosome 1p
10
, and several suggestive
linkage signals overlapped with the mouse Lith loci.
For the present study, we genotyped over 500,000 SNPs in case-
control panel A (Ta ble 1) using the Affymetrix 500K array. Stone-free
controls were chosen so that the controls had a higher median age (61
years) than the affected individuals (42 years), bearing in mind that
gallstone prevalence increases with age
2
. Genotyping was successful for
464,585 SNPs, as defined by a call rate of 493% using the BRLMM
algorithm. The 382,492 SNPs with a minor allele frequency Z2% and
no significant departure from Hardy-Weinberg equilibrium (HWE)
(P
HWE
4 0.01 in controls) were analyzed further. We estimated that
the genomic control inflation factors in panels A and A* amounted to
a negligible 1.03 and 1.04, respectively, using PLINK (Supplementary
Fig. 1 online).
Association analyses were performed twice: once using all panel A
samples (Tabl e 1) and once using only a subset of affected individuals
(N ¼ 96) and controls (N ¼ 205), denoted panel A*, with a
DM-algorithm call rate Z93% over the whole array. An age at
diagnosis o50 years (median: 29 years) was also required for inclu-
sion into panel A*. A total of 235 SNPs were selected for SNPlex-based
follow-up, a number that was chosen to match the available laboratory
resources. Follow-up markers were taken from one of two distinct
categories. For category I (N ¼ 202), SNPs were first ranked according
to the P value of the genotypic (P
CCG
)orallelic(P
CCA
)case-control
comparison in panels A and A*, and the top-ranking SNPs were
deemed potential lead markers. For a SNP to qualify as an actual lead,
at least one additional association signal, defined as a P value less than
two orders of magnitude larger than that of the possible lead marker,
was required within a distance of 50 kb. As a result, we included lead
© 2007 Nature Publishing Group http://www.nature.com/naturegenetics
Received 23 February; accepted 12 June; published online 15 July 2007; doi:10.1038/ng2101
1
First Department of Medicine,
2
Institute for Clinical Molecular Biology,
3
POPGEN Biobank and
4
Department of General and Thoracic Surgery, University Hospital
Schleswig-Holstein, 24105 Kiel, Germany.
5
Institute for Community Medicine, University Hospital Greifswald, Walther Rathenau Str. 48, 17487 Greifswald, Germany.
6
Cologne Center for Genomics, University of Cologne, Zu
¨
lpicher Strasse 47, 50674 Cologne, Germany.
7
RZPD German Resource Center for Genome Research,
Heubnerweg 6, 14059 Berlin, Germany.
8
Department of Internal Medicine I, University Hospital Bonn, Sigmund Freud-Strasse 25, 53105 Bonn, Germany.
9
Departamento de Gastroenterologia, Facultad de Medicina, Pontificia Universidad Cato
´
lica de Chile, Santiago, Chile.
10
Institute of Pharmacology, Ernst-Moritz-Arndt
University Greifswald, Friedrich Loeffler Str. 23d, 17487 Greifswald, Germany.
11
Institute of Medical Statistics and Informatics, University Hospital Schleswig-
Holstein, 24105 Kiel, Germany.
12
Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Ko
¨
ln, Germany.
13
These authors contributed equally to this work. Correspondence should be addressed to J.H. (jhampe@1med.uni-kiel.de).
NATURE GEN ETICS ADVAN CE ONLINE PUBLICATION 1
LETTERS
SNPs with P values of up to 5 10
–4
in the follow-up. The screening
stage of our study served to ascertain markers for further study
(Supplementary Fig. 2 online) rather than assessing the significance
of their potential disease association. The P value of SNP A-1791411,
corrected for multiple testing (2 382,492 tests), was 0.80, under-
lining the need for independent replication. However, to offset the
possible disregard of regions with low marker density on the 500K
array, a second marker category (II, N ¼ 33) was created after ranking
and prioritizing the remaining SNPs by their allelic and genotypic
P values alone. The full list of follow-up SNPs is provided in
Supplementary Table 1 online, and a subset is shown in Tabl e 2 .
The 235 SNPs selected for follow-up were genotyped in an
independent sample of 1,105 symptomatic gallstone carriers and 873
independent, sex-matched controls (panel B). We used the following
as criteria for formal replication: a P value of 2 10
–4
(corresponding
to P ¼ 0.05 after Bonferroni correction for 235 tests) in the allelic and
genotypic tests as well as in the logistic regression analysis controlling
for age, sex, BMI and diabetes. Only the disease association of the SNP
with Affymetrix identifier A-1791411 in the ABCG8 gene was formally
replicated. After Bonferroni correction, we found P
CCA
¼ 1.1 10
–4
,
P
CCG
¼ 4.1 10
–4
and P
regression
¼ 9.4 10
–3
. We had included
marker A-1791411 in the follow-up because of its significant disease
association in panel A*. In panel A, its call rate was only 87%.
Therefore, we confirmed the chip-based genotyping results for
A-1791411 with TaqMan (98% concordance, retrospective allelic
P ¼ 5.7 10
–6
for panel A).
We selected additional SNPs covering the ABCG8 and ABCG5 gene
region from HapMap data from individuals of European ancestry
(minor allele frequency Z 3%, pairwise r
2
Z 0.8, P
HWE
Z 0.05) using
Haploview
11
. When we genotyped these SNPs in panel B, the most
significant disease association was found for rs4299376 in intron 2 of
the ABCG8 gene (Fig. 1), with P
CCA
¼ 7.5 10
–9
and P
CCG
¼ 1.2
10
–9
(Supplementary Table 2 online).
ABCG5 and ABCG8 have been the subject of extensive mutation
detection efforts
12–14
. Starting from all known coding SNPs in these
two genes in dbSNP and the literature, we genotyped those with a
minor allele frequency 41% in the general population of European
ancestry (Supplementary Table 2). The coding SNPs responsible for
amino acid changes L36P, Q340E, M429V and G575R were mono-
morphic. For the sake of completeness, we also included two sitostero-
lemia SNPs: the SNP in ABCG8 responsible for premature stop
R121X, which was monomorphic, and the SNP responsible for
premature stop W361X in ABCG8, for which we observed one
heterozygote each in panel B controls and affected individuals. Using
P o 0.01 in both the allelic and the genotypic test as a criterion for
statistical significance, only the SNP responsible for D19H in
the ABCG8 gene was significantly associated with gallstone disease
(P
CCA
¼ 8.8 10
–10
, P
CCG
¼ 3.0 10
–9
in panel B). We confirmed
SNPlex genotyping of the SNP responsible for D19H using a TaqMan
assay (100% genotype concordance in panel B). In a logistic regression
analysis of panel B, no other SNP significantly improved the statistical
model in the presence of D19H (P 4 0.05). Haplotype analysis
(Ta bl e 3) further showed that all significant risk haplotypes indeed
carried the H allele of this SNP. We used an additional panel (C) of
independent cases and controls partly overlapping with panel B
(Ta bl e 1 ) for the fine mapping and replication of the D19H associa-
tion. The strength and localization of the association signal, as well as
the haplotype assignment of D19H, were replicated in a panel of
728 independent German affected individuals (panel C, Ta ble 1 ,
P
CCA
¼ 2.8 10
–7
, P
CCG
¼ 9.2 10
–7
) and in 167 affected individuals
and 167 controls from Chile (panel D, Tab le 1, P
CCA
¼ 0.02).
In a combined post hoc analysis of all German affected individuals,
we estimated the odds ratio to be 7.1 (95% confidence interval (c.i.),
0.9–158.6) for HH homozygosity and 2.2 (95% c.i., 1.8–2.6) for
carriership of the H allele. The latter corresponds to a population
attributable risk of B11%. In the Chilean sample, the 19H carriership
odds ratio was 1.9. A sample of individuals with gallstones from
Greifswald, which included an approximately equal numbers of
symptomatic (N ¼ 374) and asymptomatic (N ¼ 354) gallstone
carriers, demonstrated a disease association of 19H carriership for
both groups of affected individuals, with odds ratios of 2.5 (P ¼ 4.8
10
–6
)and2.4(P ¼ 1.7 10
–6
), respectively. From panels A–C and the
Chilean sample, the combined significance of rs11887534 was
calculated as P ¼ 7.74 10
–9
by Fishers technique, taking into
account multiple testing in panel B and excluding overlapping controls
from panel C.
We performed a logistic regression analysis in all German affected
individuals (N ¼ 2,113) and controls (N ¼ 1,524, including 129
hitherto unused male controls) for which information on age, sex,
BMI and type II diabetes status was available. Upon adjustment for
these covariates, the effect of 19H carriership remained highly sig-
nificant (Wald P o 0.0001) and was similar to that seen in the
unadjusted analysis (adjusted odds ratio ¼ 2.2). Furthermore, none of
the interaction terms was significant in the presence of D19H. For 495
affected individuals in panels A and B, stone composition data were
available from previous FTIR spectroscopy
15
. The odds ratio of 19H
carriership was 3.3 (95% c.i., 2.5–4.3) for stones with cholesterol as a
major constituent, defined by a cholesterol content 430% (wt/wt). In
contrast, allele 19H was completely lacking in the 12 affected indivi-
duals with pure pigment stones.
Mutations in ABCG8 have been shown to be responsible for
sitosterolemia, a rare autosomal recessive disorder caused by the
excessive accumulation of plant sterols
16
. In view of the low prevalence
of sitosterolemia, with fewer than 200 affected families reported
© 2007 Nature Publishing Group http://www.nature.com/naturegenetics
Table 1 Summary of study populations
Affected individuals Controls
Panel N Median age Age at diagnosis N Age Percentage male Percentage symptomatic
Screening panel (panel A) 280 42 38 360 61 50% 86
Replication I and fine mapping (panel B) 1,105 54 48 873 63 28% 100
Replication II and fine mapping (panel C) 728 60 n/a 732
a
65 40% 51
Replication III (Chile) (panel D) 167 54 n/a 167 48 0% 72
All samples were sex matched. All subjects in panel A* were symptomatic with gallstone disease. German affected individuals and controls were recruited in northern Germany
through clinical centers at Kiel University, the POPGEN project and the SHIP project at Greifswald University. No notable population genetic differences exist between these two
regions
30
, so we combined patient samples into genotyping panels on the basis of phenotypic criteria alone. Ages are given in years.
a
Controls in this panel include 570 individuals randomly selected from panel B to match the age and sex distribution of the affected individuals.
2 ADVANCE ONLINE PUBLICATION NATURE GENETICS
LETTERS
worldwide, it is clear that causative mutations for this mendelian
disorder cannot at the same time account for 10% of individuals with
a frequent condition such as gallstone disease. Indeed, the previously
characterized sitosterolemia mutations either were not found in our
study subjects or occurred at the same frequency in affected indivi-
duals and controls, thereby confirming that gallstone disease is allelic,
but not genetically identical, to sitosterolemia. On the contrary,
sitosterolemia may even confer some resistance to gallstone disease,
bearing in mind that individuals with sitosterolemia secrete less
cholesterol into the bile than expected from their increased levels of
intestinal cholesterol absorption
17
.
Logistic regression analysis using BMI, sex, age and type II diabetes
status as covariates showed that the observed genetic gallstone risk was
independent of these potential confounders
18
. This result corroborates
previous epidemiological evidence that gallstone susceptibility cannot
be explained by established risk factors for gallstone disease, such as
obesity, for example
18–20
.
A number of human linkage signals
10
colocalize with the association
leads (Tabl e 2 and Supplementary Table 1). The replicated marker
A-1791411 is not localized in one of the linkage regions of the Mexican
American scan. This lack of concordance may be due to population
differences or may reflect the lower power of nonparametric linkage
analysis for a relatively infrequent genetic risk variant like D19H. This
also underlines the need for further studies to elucidate the nature of
the remaining genetic risk in gallstone disease (Supplementary Note
online). With hindsight, the identification of the ABCG5/ABCG8 locus
as a risk factor for human gallstone disease seems almost too obvious
to have required a genome-wide association scan, because the human
ABCG5/ABCG8 locus is homologous to a previously reported mouse
susceptibility locus (Lith9) detected in strain PERA/Ei
9
. However,
earlier studies by our group investigating candidate human genes
© 2007 Nature Publishing Group http://www.nature.com/naturegenetics
Table 2 Replication study (panel B) of the top 235 SNPs from the genome-wide association scan (panels A and A*)
Screening (panel A) Screening (panel A*) Panels A and A* HapMap Replication (panel B)
# Panel dbSNP ID CR P
CCA
P
CCG
CR P
CCA
P
CCG
MAF
(ctrl)
MAF
(case) MAF CR P
CCA
P
CCG
MAF
(ctrl)
MAF
(case) P
REG
24 A* rs6716275
a
0.93 5.12 10
–4
2.89 10
–3
0.97 8.06 10
–5
4.09 10
–4
0.34 0.52 0.41 1 1.67 10
–1
4.15 10
–2
0.43 0.41 5.75 10
–1
25 A* rs2194448
b
0.97 1.15 10
–1
1.82 10
–1
0.99 2.60 10
–4
1.17 10
–4
0.47 0.63 0.47 1 6.72 10
–1
7.83 10
–1
0.49 0.48 8.61 10
–1
26 A rs1550992
b
0.95 3.42 10
–4
1.23 10
–3
0.98 2.77 10
–1
5.07 10
–1
0.21 0.30 0.35 1 6.03 10
–2
1.27 10
–1
0.21 0.24 9.52 10
–2
27 A rs11124408
b
17.15 10
–5
1.64 10
–4
13.55 10
–4
1.49 10
–3
0.09 0.16 0.18 1 2.06 10
–2
2.28 10
–2
0.10 0.13 3.00 10
–2
28 A* rs7571463
b
0.92 6.94 10
–1
7.88 10
–1
0.94 4.91 10
–4
1.81 10
–3
0.23 0.11 0.27 1 5.32 10
–3
2.03 10
–2
0.18 0.21 8.62 10
–3
29 A* n/a
b,c
0.87 5.30 10
–5
6.01 10
–6
0.94 2.11 10
–6
9.84 10
–6
0.05 0.18 n/a
d
1 4.07 10
–9
1.53 10
–8
0.05 0.10 3.43 10
–7
30 A* rs370068 1 1.48 10
–1
1.03 10
–1
13.04 10
–4
6.09 10
–4
0.30 0.45 0.27 1 7.85 10
–1
4.95 10
–1
0.33 0.33 7.45 10
–1
31 A* rs4852395 0.99 6.43 10
–2
5.30 10
–2
11.23 10
–3
3.56 10
–4
0.16 0.27 0.18 0.99 1.15 10
–1
2.86 10
–1
0.15 0.17 3.02 10
–1
32 A rs1517862 13.87 10
–4
2.95 10
–4
11.35 10
–2
1.20 10
–2
0.02 0.06 0.08 1 6.45 10
–1
4.47 10
–1
0.03 0.04 8.49 10
–2
SNPs fell into one of two categories: those selected on the basis of additional flanking marker support (I) and those selected based upon the P value of the lead marker alone (II). Note, all SNPs in this table are category I and are located
on chromosome 2. The analysis was further stratified by whether a SNP was found to rank among the most significant markers in panel A or A*. Markers are reported by chromosome and nucleotide position (NCBI build 35). The call rate
(CR) and P values for the genotype-based (P
CCG
) and allele-based case-control comparisons (P
CCA
) are reported alongside the minor allele frequencies (MAF) in each sample and in the HapMap CEU category, for comparison. Boldface
indicates results that led to the inclusion of a marker in the follow-up. Markers with P o 0.0002 in all of the allele-, genotype- and regression-based tests in panel B are underlined. For replication panel B, a genotypic logistic regression
P value (P
REG
) controlling for age, sex, BMI and type II diabetes status is included. Only markers neighboring A-1791411 are shown; the full table is available as Supplementary Table 1.
a
Localizes to human chromosome 2p24.2, corresponding to one of the linkage regions reported in ref. 10.
b
Corresponds to the mouse Lith locus Lith9, as reported in refs. 6–9.
c
No ‘rs’ identifier exists (probe set ID A-1791411).
d
No HapMap MAF available,
because no ‘rs’ identifier assignment exists.
44.02 Mb
121110 1398765432112345678910111213
ABCG8ABCG5
chr2
43.95 Mb
rs2954804
rs2954802
rs6544718
rs11124950
rs4148221
rs4148222
rs4148219
rs17031742
rs4953028
rs13405698
rs4953027
rs4245795
rs12468591
rs4148217
rs17409589
rs10174731
*
rs10221914
rs4245791
rs4299376
rs10177200
rs3806470
rs4131229
rs4289236
rs4073237
rs4245786
rs1864814
rs10439467
rs10201851
rs10205816
rs10208987
rs4148192
rs4148194
rs6720173
rs10180615
rs17424122
*
rs4953023*
rs4148211*
rs11887534*
rs4148187*
A-1791411*
Figure 1 Overview of the physical and genetic structure of the ABCG5/
ABCG8 region. The physical position of the investigated SNPs and a
schematic representation of the gene structures are shown above. The
D19H variant is marked by an arrow. Coordinates refer to NCBI genome
assembly build 35. Below is the linkage disequilibrium structure of the
locus (D¢), as generated by Haploview
11
from the control genotypes of
panel B. Haplotype blocks derived from the HapMap genotypes from
individuals of European ancestry (CEU) are outlined in black in the
D¢ chart. SNPs used in the haplotype analysis (see Table 3)arein
boldface and are marked by an asterisk (*).
NATURE GEN ETICS ADVAN CE ONLINE PUBLICATION 3
LETTERS
orthologous to mouse Lith1 (ref. 21) and Lith6 (ref. 22) loci were
consistently disappointing. Nevertheless, the ABCG5/ABCG8 region
clearly represented a prime candidate after Abcg5 and Abcg8 (both
located within Lith9) were suggested as possible mouse candidate
genes
9,23
. Furthermore, variants in the ABCG5/ABCG8 genes have
been reported recently to be associated with different plasma lipid
levels in siblings with gallstones, suggesting a potential alteration of
bile cholesterol levels by the ABCG5/ABCG8 genotype
24
.
Although the genetic gallstone disease risk is clearly attributable to
variant D19H in the ABCG8 gene in our study, we cannot exclude the
possibility that other as-yet-unidentified variants at the ABCG5/
ABCG8 locus might also contribute to gallstone susceptibility. The
ABCG8 protein transports cholesterol into both the biliary and the gut
lumen and has been extensively characterized after its identification as
the causative gene for sitosterolemia (see ref. 12 for a recent review).
Others
25
have observed a lower concentration of campesterol in 19H
carriers and hypothesized that the H allele leads to an increased
transporter activity of ABCG8. This hypothesis received further
support by the observation that 19H carriership is associated with
lower total serum cholesterol levels
24,26
. Hypothetically, 19H carrier-
ship may thus increase the efficiency of the cholesterol transport into
the bile lumen, causing cholesterol hypersaturation of the bile and
eventually promoting the formation of cholesterol gallstones
27
.How-
ever, additional functional studies are needed to determine the effect
of the D19H variant on the ABCG8/ABCG5 heterodimer.
METHODS
Study subjects and procedures. Written informed consent was obtained from
all study participants. Study protocols were approved by the ethics committees
of the Kiel and Greifswald University Hospitals. Details of the recruitment and
genotyping technique are provided in Supplementary Methods online.
Statistical analysis. All markers were tested for a possible deviation from HWE
in the controls before inclusion. Single-marker association tests were performed
using Haploview
11
and GENOMIZER
28
with a w
2
test or Fishers exact test for
contingency tables, as appropriate. Haplotype frequency estimates were
obtained from singletons using COCAPHASE
29
. Significance testing of haplo-
type frequency differences was performed on the basis of the COCAPHASE
results, making use of the fact that twice the log-likelihood ratio between two
nested data models approximately follows a w
2
distribution with k degrees of
freedom, where k is the difference in the number of parameters between the two
models. Logistic regression analysis was performed with SPSS version 11.0,
coding individual SNP genotypes as categorical variables.
Note: Supplementary information is available on the Nature Genetics website.
ACKNOWLEDGMENTS
The cooperation of all patients, their families and physicians is gratefully
acknowledged. The authors gratefully acknowledge the support by the following
heads of surgical departments: I. Vogel (Sta
¨
dtisches Krankenhaus Kiel),
H. Dittrich (Rendsburg), J. Belz (Husum), R. Qua
¨
schling (Eckernfo
¨
rde),
H. Shekarriz (Schleswig), V. Mendel (Flensburg), W. Neugebauer (Flensburg),
F. Kallinowski (Heide), J. Klima (Niebu
¨
ll), M. Sailer (Hamburg) and
A. Schafmayer (Lu
¨
neburg). Special thanks are given to C. Fu
¨
rstenau, T. Wesse,
B. Petersen, L. Bossen, T. Henke, S. Ehlers, A. Dietsch and V. Pucken for technical
assistance. This study was supported by the German Ministry of Education and
Research through the POPGEN biobank project (01GS0426, 01GR0468), the
MediGrid project and the National Genotyping Platforms in Kiel and Cologne
and by the German Research Council (Ha 3091/2-1, 4-1, La 997/3-1), Applied
Biosystems, Mucosaimmunologie gGmbH and the Medical Faculty Kiel. The
SHIP recruitment project is funded by the Federal Ministry of Education and
Research (ZZ9603), the Ministry of Cultural Affairs and the Social Ministry of the
Federal State of Mecklenburg-West Pomerania. The Chilean study was supported
by grants from FONDECYT (Fondo Nacional de Desarrollo Cien
´
fico y
Te c n o l o g o
´
gico) (numbers 1040820 (to J.F.M.) and 1030744 (to F.N.)).
AUTHOR CONTRIBUTIONS
S.B. performed the SNP selection, genotyping and data analysis and prepared the
figures and tables; C.S. coordinated the Kiel recruitment, phenotyped patients and
helped write the paper; J.E., H.v.E., C.H., B.T. and M.S. recruited patients and
helped write the paper. C.B., I.B., C.K. and P.N. performed the chip genotyping
and chip data analysis; H.V. and D.R. coordinated the SHIP recruitment and
participated in experimental design; J.F.M., F.N. coordinated the recruitment in
Chile and participated in experimental design; A.F., M.B., A.E., T.L. and M.W.
helped with data analysis and genotyping and F.L., F.F., U.R.F., S.S., P.N. and J.T.
helped design the experiment, supported recruitment and helped write the paper.
M.K. supervised and performed the statistical analysis and edited the paper. J.H.
designed and supervised the experiment, performed data analysis and wrote the
manuscript. All authors have revised the manuscript for intellectual content.
COMPETING INTERESTS STATEMENT
The authors declare no competing financial interests.
Published online at http://www.nature.com/naturegenetics
Reprints and permissions information is available online at http://npg.nature.com/
reprintsandpermissions
1. Lammert, F. & Sauerbruch, T. Mechanisms of disease: the genetic epidemiology of
gallbladder stones. Nat. Clin. Pract. Gastroenterol. Hepatol. 2, 423–433 (2005).
2. Volzke, H. et al. Independent risk factors for gallstone formation in a region with high
cholelithiasis prevalence. Digestion 71,97105(2005).
3. Ko
¨
rner, G. U
¨
ber die famila
¨
re Ha
¨
ufung der Gallenblasenkrankheiten. Zeitschrift fu
¨
r
Menschliche Vererbungs- und Konstitutionslehre 20, 528–582 (1937).
4. Nu
¨
rnberg, D., Berndt, H. & Pannwitz, H. Familia
¨
re Ha
¨
ufung von Gallensteinen. Dtsch.
med. Wschr. 114, 1059–1063 (1989).
5. Katsika, D. et al. Genetic and environmental influences on symptomatic gallstone
disease: a Swedish study of 43,141 twin pairs. Hepatology 41, 1138–1143
(2005).
© 2007 Nature Publishing Group http://www.nature.com/naturegenetics
Table 3 Haplotype analysis of seven SNPs at the ABCG8 locus in panels B and C
Fine mapping, panel B Fine mapping, panel C
Haplotype f
cases
f
controls
OR
case-control
P
COCAPHASE
f
cases
f
controls
OR
case-control
P
COCAPHASE
C-G-A-T-G-T-C 0.340 0.382 0.8 0.01 0.349 0.392 0.8 0.02
T-G-G-T-G-T-G 0.342 0.325 1.1 0.24 0.340 0.325 1.1 0.38
C-G-G-T-G-T-G 0.066 0.071 0.9 0.53 0.077 0.069 1.1 0.42
C-G-A-T-G-T-G 0.067 0.068 1.0 0.89 0.060 0.068 0.9 0.33
C-
C-A-C-A-T-C 0.097 0.045 2.3 7.75 10
–7
0.080 0.038 2.2 8.76 10
–7
C-G-A-T-G-A-C 0.023 0.043 0.5 5.73 10
–4
0.028 0.042 0.7 0.03
T-G-A-T-G-A-C 0.025 0.033 0.8 0.12 0.020 0.033 0.6 0.03
T-G-A-T-G-A-C 0.028 0.024 1.2 0.53 0.036 0.028 1.3 0.305
SNPs included in the haplotype analysis (rs4148187, rs11887534 (D19H), rs4148211 (Y54C), SNP_A-1791411, rs4953023, rs17424122 and rs17409589) are marked by an
asterisk in Figure 1. All analyses were carried out using COCAPHASE
29
. Nonsynonymous SNP rs11887534 (D19H) is in boldface, and the risk allele is underlined. The sole risk
haplotype (C-
C-A-C-A-T-C) is fully defined by rs11887534 allele C, corresponding to histidine at the amino acid sequence level; all other haplotypes are protective and carry allele G.
This haplotype pattern strongly suggests that rs11887534 is the major risk variant at the ABCG8 locus. OR, odds ratio.
4 ADVANCE ONLINE PUBLICATION NATURE GENETICS
LETTERS
6. Khanuja, B. et al. Lith1, a major gene affecting cholesterol gallstone formation
among inbred strains of mice. Proc. Natl. Acad. Sci. USA 92, 7729–7733
(1995).
7. Paigen, B. et al. Quantitative trait loci mapping for cholesterol gallstones in AKR/J and
C57L/J strains of mice. Physiol. Genomics 4, 59–65 (2000).
8. Lammert, F. et al. Lith genes control mucin accumulation, cholesterol crystallization,
and gallstone formation in A/J and AKR/J inbred mice. Hepatology 36, 1145–1154
(2002).
9. Wittenburg, H. et al. FXR and ABCG5/ABCG8 as determinants of cholesterol gallstone
formation from quantitative trait locus mapping in mice. Gastroenterology 125,
868–881 (2003).
10. Puppala, S. et al. A genomewide search finds major susceptibility loci for gallbladder
disease on chromosome 1 in Mexican Americans. Am. J. Hum. Genet. 78, 377–392
(2006).
11. Barrett, J.C., Fry, B., Maller, J. & Daly, M.J. Haploview: analysis and visualization of LD
and haplotype maps. Bioinformatics 21, 263–265 (2005).
12. Hazard, S.E. & Patel, S.B. Sterolins ABCG5 and ABCG8: regulators of whole body
dietary sterols. Pflugers Arch. 453, 745–752 (2007).
13. Hubacek, J.A., Berge, K.E., Cohen, J.C. & Hobbs, H.H. Mutations in ATP-cassette
binding proteins G5 (ABCG5) and G8 (ABCG8) causing sitosterolemia. Hum. Mutat.
18, 359–360 (2001).
14. Iida, A. et al. Catalog of 605 single-nucleotide polymorphisms (SNPs) among 13 genes
encoding human ATP-binding cassette transporters: ABCA4, ABCA7, ABCA8, ABCD1,
ABCD3, ABCD4, ABCE1, ABCF1, ABCG1, ABCG2, ABCG4, ABCG5, and ABCG8.
J. Hum. Genet. 47, 285–310 (2002).
15. Schafmayer, C. et al. Predictors of gallstone composition in 1025 symptomatic
gallstones from Northern Germany. BMC Gastroenterol. 6, 36 (2006).
16. Berge, K.E. et al. Accumulation of dietary cholesterol in sitosterolemia caused by
mutations in adjacent ABC transporters. Science 290, 1771–1775 (2000).
17. Salen, G. et al. Sitosterolemia. J. Lipid Res. 33, 945–955 (1992).
18. Nakeeb, A. et al. Gallstones: genetics versus environment. Ann. Surg. 235, 842–849
(2002).
19. Sarin, S.K., Negi, V.S., Dewan, R., Sasan, S. & Saraya, A. High familial prevalence of
gallstones in the first-degree relatives of gallstone patients. Hepatology 22, 138–141
(1995).
20. Duggirala, R., Mitchell, B.D., Blangero, J. & Stern, M.P. Genetic determinants of
variation in gallbladder disease in the Mexican-American population. Genet. Epide-
miol. 16, 191–204 (1999).
21. Schafmayer, C. et al. Investigation of the Lith1 candidate genes ABCB11 and LXRA in
human gallstone disease. Hepatology 44, 650–657 (2006).
22. Schafmayer, C. et al. Investigation of the Lith6 candidate genes APOBEC1 and PPARG
in human gallstone disease. Liver Int. (in the press).
23. Wittenburg, H. et al. Association of a lithogenic Abcg5/Abcg8 allele on Chromosome
17 (Lith9) with cholesterol gallstone formation in PERA/EiJ mice. Mamm. Genome 16,
495–504 (2005).
24. Acalovschi, M. et al. Are plasma lipid levels related to ABCG5/ABCG8 polymorphisms?
A preliminary study in siblings with gallstones. Eur. J. Intern. Med. 17, 490–494
(2006).
25. Berge, K.E. et al. Heritability of plasma noncholesterol sterols and relationship to DNA
sequence polymorphism in ABCG5 and ABCG8. J. Lipid Res. 43, 486–494 (2002).
26. Gylling, H. et al. Polymorphisms in the ABCG5 and ABCG8 genes associate with
cholesterol absorption and insulin sensitivity. J. Lipid Res. 45, 1660–1665 (2004).
27. Wittenburg, H. & Carey, M.C. Biliary cholesterol secretion by the twinned sterol half-
transporters ABCG5 and ABCG8. J. Clin. Invest. 110, 605–609 (2002).
28. Franke, A. et al. GENOMIZER: an integrated analysis system for genome-wide
association data. Hum. Mutat. 27, 583–588 (2006).
29. Dudbridge, F. Pedigree disequilibrium tests for multilocus haplotypes. Genet. Epide-
miol. 25, 115–121 (2003).
30. Steffens, M. et al. SNP-based analysis of genetic substructure in the German
population. Hum. Hered. 62, 20–29 (2006).
© 2007 Nature Publishing Group http://www.nature.com/naturegenetics
NATURE GEN ETICS ADVAN CE ONLINE PUBLICATION 5
LETTERS
... 8 These mutations have been found closely linked to metabolic risk factors such as aberrations in lipid profiles, elevated body mass index (BMI), and the presence of coronary artery disease. 3,4,18,21 Among Latin Americans, particularly those of Mapuche descent in Chile, the higher prevalence of GS disease and subsequent complications like gallbladder cancer has been attributed to the presence of the ABCG8 D19H (rs1887534) risk variant. Incidence rates are estimated to be 50% in women and 12% in men. ...
Article
Full-text available
Gallstone (GS) disease is common and arises from a combination of genetic and environmental factors. Although genetic abnormalities specifically leading to cholesterol GSs are rare, there are clinically significant gene variants associated with cholesterol GSs. In contrast, most bilirubin GSs can be attributed to genetic defects. The pathogenesis of cholesterol and bilirubin GSs differs greatly. Cholesterol GSs are notably influenced by genetic variants within the ABC protein superfamily, including ABCG8, ABCG5, ABCB4, and ABCB11, as well as genes from the apolipoprotein family such as ApoB100 and ApoE (especially the E3/E3 and E3/E4 variants), and members of the MUC family. Conversely, bilirubin GSs are associated with genetic variants in highly expressed hepatic genes, notably UGT1A1, ABCC2 (MRP2), ABCC3 (MRP3), CFTR, and MUC, alongside genetic defects linked to hemolytic anemias and conditions impacting erythropoiesis. While genetic cases constitute a small portion of GS disease, recognizing genetic predisposition is essential for proper diagnosis, treatment, and genetic counseling.
... The D19H polymorphism in ABCG8 is by far the most prominent genetic determinant of GSD, GWAS data also showed that mutations in some loci of ABCG5/8 were strongly associated with GSD 47 . The link between GSD and cardiovascular disease is thought to arise through ABCG5/8, a transporter protein that promotes the secretion of cholesterol and phytosterols into the bile 34,48 . In animal studies, researchers found that high expression of ABCG5/8 attenuated hypercholesterolemia in Ldlr−/− mice, which in turn significantly ameliorated atherosclerotic lesions in the aorta 49 . ...
Article
Full-text available
Gallstone disease (GSD) is thought to be associated with the risk of coronary heart disease (CHD) or acute myocardial infarction (AMI), which may be due to abnormal cholesterol metabolism. We used multiple Mendelian randomization (MR) methods based on publicly available genome-wide association study data to assess whether this association is genetically causal and to search for loci driving causality. Pooled data for GSD were obtained from FinnGen Biobank and Biobank Japan, while CHD and AMI were obtained as pooled data from the CARDIoGRAMplusC4D consortium. In this MR study, we found a significant negative causal effect of genetic susceptibility to GSD on AMI in the Finnish population, but no causal effect was found on CHD. This causal effect was not confounded by reverse causality and the same findings were obtained in the Japanese population. Furthermore, the negative causal effect of GSD on AMI risk may be driven by the rs4245791-regulated ABCG5/8 protein. In conclusion, the results of this MR study support a negative causal effect of GSD on AMI and suggest that rs4245791 is the causal driver locus of this effect, which provides new ideas and evidence for the prevention and etiologic study of AMI in patients with GSD.
... Out of them, 11 are known variations, while 21 are novel variations. Through a recessive mode of inheritance, one of the novel common variations, rs708686 upstream of FUT6, dramatically increases its association with gallstone disease [120][121][122]. ...
Article
Full-text available
Over the years, several studies have revealed an important link between thyroid disorders and gallstone disease. According to these studies, hypothyroidism and hyperthyroidism are associated with cholesterol gallstone disease. This association between thyroid hormone disorders and cholesterol gallstone disease is due to the importance of thyroid hormones on cholesterol synthesis, bile functioning and content, and gallbladder motility. Several genes and receptors have been found on the thyroid gland, liver, and gallbladder to verify this association. These genes affect thyroid hormone secretion, lipid metabolism, and bile secretion. Defects in these various gene expression and protein functions lead to bile duct diseases. Other causes that lead to cholesterol gallstone disease are supersaturation of the bile with cholesterol and impaired gallbladder motility, which leads to bile stasis. This article has discussed these factors in detail while highlighting the association between thyroid hormones and cholesterol gallstone disease.
... While metabolic syndrome increases cholelithiasis risk, primary prevention through lifestyle changes is also possible. Moreover, common mutations in the hepatic cholesterol transporter ABCG8 represent a major genetic risk factor, accounting for about 25% of total risk [16,18,19]. Although our understanding of the genetics and pathophysiology has improved recently, invasive surgery-based procedures still dominate treatment strategies. ...
Article
Full-text available
Background: Cholelithiasis is a common digestive system disease that imposes major burden on patients and society. Investigating the relationship between dietary factors and cholelithiasis risk can provide a basis for disease prevention. Previous studies on milk intake and cholelithiasis incidence have been limited.Therefore, the aim of our study was to assess the association between milk consumption and the incidence of cholelithiasis in males and females. Methods: We selected 14,722 adults (≥ 18 years old) from National Center for Health Statistics (NHANSE) 2017-2020, and collected general characteristics of patients in the database, such as age, gender, race and body mass index (BMI), as well as dietary information (milk consumption). The occurrence of cholelithiasis was used as the outcome event, and the group was divided into cholelithiasis and non-cholelithiasis groups according to the outcome event. We used logistic regression models in generalized linear model (GLM) functions, controlling for demographic, lifestyle, and dietary factors, to estimate the association between milk intake and the incidence of cholelithiasis in males and females. Results: A total of 14,722 adults were included. In the present study, the overall weighted prevalence of cholelithiasis was 10.96%, with 15.18% and 6.48% prevalence in females and males, respectively. Compared to infrequent milk intake, frequent milk intake (once a week or more) in females was associated with reduced cholelithiasis risk (OR 0.74, 95% CI 0.61-0.90). Daily milk intake in males was also related to lower cholelithiasis risk (OR 0.69-0.82). As adjusted variables increased in the models, predictive performance was improved (AUC 0.711 in females, 0.730 in males). Conclusions: Appropriate milk intake may correlate with decreased cholelithiasis risk. Our study provides a basis for dietary interventions against gallstones, but prospective studies are needed to verify the results.
Article
Background Gallstone disease (GD) is common but remains asymptomatic in most cases. However, gallstones can lead to complications like choledocholithiasis or gallbladder cancer. In this study, we analyse the common genetic risk factor for GD, the p.D19H variant in the sterol transporter ABCG8 , in Polish patients with gallstones and gallbladder cancer. Methods Three adult cohorts were prospectively recruited: 65 patients with gallbladder cancer, 170 obese individuals scheduled for bariatric surgery and 72 patients who underwent endoscopic retrograde cholangiopancreatography due to recurrent choledocholithiasis. The control cohort consisted of 172 gallstone‐free adults. The ABCG8 p.D19H (rs11887534) polymorphism was genotyped using TaqMan assays. Results The minor allele frequency (MAF) of the ABCG8 p.D19H polymorphism was significantly ( p = .02) higher among cases with either gallstones or gallbladder cancer (MAF = 8.4%) as compared to controls (MAF = 4.0%). The highest frequency of the risk allele was detected in patients with gallbladder cancer (18.5%) and obese patients with GD (17.5%), followed by individuals with choledocholithiasis (13.9%). Notably, the p.19H variant was associated with an increased risk of developing gallbladder cancer (OR 2.76, 95% CI 1.16–6.54, p = .01) and an increased risk of GD in obese individuals scheduled for bariatric surgery (OR = 2.70, 95% CI 1.05–6.49, p = .03), but did not significantly affect the risk of choledocholithiasis. Conclusions The ABCG8 p.D19H common risk variant increases the risk of developing gallbladder cancer in Central Europeans and enhances the risk of gallstones in the obese. Carriers of the p.D19H variant might benefit from personalized preventive strategies, particularly regarding gallbladder cancer.
Article
Acute cholecystitis is a pathology in which the gallbladder wall becomes inflamed. Gallstones are the primary triggering factor of cholecystitis; they are present in more than 10% of the population and their presence increases with age. The main factors for the formation of gallstones are estrogen, pregnancy, cirrhosis, diabetes melitis, obesity and hemolytic disease. Gallstone disease is a common reason for non-gynecologic operations during pregnancy and is the major non-obstetric cause for hospitalization. Pregnancy may accentuate gallbladder stone formation. Alterations in hepatobiliary function occur during pregnancy to create a lithogenic environment. Conclusion our study is to investigate the prevalence of acute calculus cholecystitis among female childbearing age. the objectives: To investigate the prevalence of the most common age acute calculus cholecystitis among female childbearing age. To identify the risk factors associated with gallstones among pregnant women. To determine the most common predisposing factor to gallstones formation. With the sample size of 72, and the questionnaire was self-administered and also the target population was selected from hospital patients and the data was analyzed and interoperated by SPSS and Excel. Recommends educating of community specially women to reduce the effect of the diseases that can be easily treated. Providing free health care for those women with low income level. Mother should visit the doctor every trimester to check up her health condition and to provide consultation.
Article
Background and Aim A large genetic effect of a novel gallstone‐associated genetic variant, the hepatocyte nuclear factor 4α ( HNF4A ) rs1800961 polymorphism, has been identified through recent genome‐wide association studies. However, this effect has not been validated in Asian populations. We investigated the association between the rs1800961 variant and gallstones among a Taiwanese population. Methods A total of 20 405 participants aged between 30 and 70 years voluntarily enrolled in the Taiwan Biobank. Self‐report questionnaires, physical examinations, biochemical tests, and genotyping were used for analysis. The association of the HNF4A rs1800961 variant and other metabolic risks with gallstone disease was analyzed using multiple logistic regression models. Results The minor T allele of HNF4A rs1800961 was associated with an increased risk of gallstone, and the association remained significant even after adjustment for other risk factors including age, body mass index (BMI), diabetes, hyperlipidemia, hypertension, and cigarette smoking (adjusted odds ratio [OR] = 1.90, 95% confidence interval [CI] = 1.31 to 2.75) in male participants. When further stratified by BMI and age, the lithogenic effect was the most significant in male participants with obesity (adjusted OR = 3.55, 95% CI = 1.92 to 6.56) and who were younger (adjusted OR = 2.45, 95% CI = 1.49 to 4.04). Conclusion The novel gallstone‐associated HNF4A rs1800961 variant was associated with the risk of gallstone in the Taiwanese men. Screening for the rs1800961 polymorphism may be particularly useful in assessing the risk of gallstone formation in younger or obese men.
Chapter
Sitosterolemia is a rare genetic lipid disorder, mainly characterized by the accumulation of dietary xenosterols in plasma and tissues. It is caused by inactivating mutations in either ABCG5 or ABCG8 subunits, a subfamily-G ATP-binding cassette (ABCG) transporters. ABCG5/G8 encodes a pair of ABC half transporters that form a heterodimer (G5G8). This heterodimeric ATP-binding cassette (ABC) sterol transporter, ABCG5/G8, is responsible for the hepatobiliary and transintestinal secretion of cholesterol and dietary plant sterols to the surface of hepatocytes and enterocytes, promoting the secretion of cholesterol and xenosterols into the bile and the intestinal lumen. In this way, ABCG5/G8 function in the reverse cholesterol transport pathway and mediate the efflux of cholesterol and xenosterols to high-density lipoprotein and bile salt micelles, respectively. Here, we review the biological characteristics and function of ABCG5/G8, and how the mutations of ABCG5/G8 can cause sitosterolemia, a loss-of-function disorder characterized by plant sterol accumulation and premature atherosclerosis, among other features.
Article
Full-text available
Sitosterolemia is a rare inherited lipid storage disease characterized chemically by the accumulation of plant sterols and 5 alpha-saturated stanols in plasma and tissues. Very low cholesterol synthesis due to a deficiency of HMG-CoA reductase associated with increased intestinal plant sterol absorption and slow hepatic sterol removal are major biochemical features. Because cholesterol synthesis cannot up-regulate, bile acid malabsorption mobilizes body sterols for bile acid synthesis and dramatically lowers plasma and monocyte sterol concentrations and may halt the progression of the atherosclerotic process.
Article
Full-text available
In healthy individuals, acute changes in cholesterol intake produce modest changes in plasma cholesterol levels. A striking exception occurs in sitosterolemia, an autosomal recessive disorder characterized by increased intestinal absorption and decreased biliary excretion of dietary sterols, hypercholesterolemia, and premature coronary atherosclerosis. We identified seven different mutations in two adjacent, oppositely oriented genes that encode new members of the adenosine triphosphate (ATP)-binding cassette (ABC) transporter family (six mutations in ABCG8 and one in ABCG5) in nine patients with sitosterolemia. The two genes are expressed at highest levels in liver and intestine and, in mice, cholesterol feeding up-regulates expressions of both genes. These data suggest that ABCG5 and ABCG8 normally cooperate to limit intestinal absorption and to promote biliary excretion of sterols, and that mutated forms of these transporters predispose to sterol accumulation and atherosclerosis.
Article
Single-nucleotide polymorphisms (SNPs) at some gene loci are useful as markers of individual risk for adverse drug reactions or susceptibility to complex diseases. We have been focusing on identifying SNPs in and around genes encoding drug-metabolizing enzymes and transporters, and have constructed several high-density SNP maps of such regions. Here we report SNPs at additional loci, specifically 13 genes belonging to the superfamily of ATP-binding cassette transporters (ABCA4, ABCA7, ABCA8, ABCD1, ABCD3, ABCD4, ABCE1, ABCF1, ABCG1, ABCG2, ABCG4, ABCG5, and ABCG8). Sequencing a total of 416 kb of genomic DNA from 48 Japanese volunteers identified 605 SNPs among these 13 loci: 14 in 5′ flanking regions, 5 in 5′ untranslated regions, 37 within coding elements, 529 in introns, 8 in 3′ untranslated regions, and 12 in 3′ flanking regions. By comparing our data with SNPs deposited in the dbSNP database of the National Center for Biotechnology Information (US) and with published reports, we determined that 491 (81%) of the SNPs reported here were novel. We also detected 107 genetic variations of other types among the loci examined (insertion–deletions or mono- di-, or trinucleotide polymorphisms). The high-density SNP maps we constructed on the basis of these data should provide useful information for investigating associations between genetic variations and common diseases or responsiveness to drug therapy.
Article
Von März 1986 bis Juni 1988 wurden 1616 scheinbar gesunde Probanden, 634 Männer und 982 Frauen im Alter von 12-93 Jahren, mittels Ultraschalls auf Gallensteine untersucht. Gleichzeitig wurden sie gefragt, ob bei nahen Blutsverwandten jemals Gallensteine aufgetreten waren oder eine Cholezystektomie durchgeführt worden war. Bei 316 der 1616 Probanden (19,6 %) wurden sonographisch Gallensteine festgestellt, oder sie waren in der Vergangenheit cholezystektomiert worden. Die Prävalenz der Gallensteine stieg mit dem Alter, sie war bei Frauen größer als bei Männern. Bei den 316 Probanden mit nachgewiesenen Gallensteinen oder einer Cholezystektomie in der Anamnese waren in sechs von neun Altersgruppen signifikant (P < 0,01 bzw. P < 0,05) häufiger Gallensteine in der Familie bekannt als bei den Probanden ohne Gallensteine. Diese Befunde sprechen für eine genetische Disposition zur Cholelithiasis, Ernährungseinflüsse sind jedoch nicht völlig ausgeschlossen.
Article
From March 1986 to June 1988 ultrasound examinations for gallstones were performed on 1616 symptom-free persons (982 females and 634 males, aged 12-93 years). They were also asked about gallstones and (or) cholecystectomy among blood relatives. Gallstones were discovered or a previous cholecystectomy was reported in 316 persons (19.6%). The prevalence of gallstones was age-dependent and increased with age. Prevalence was higher in females than in males. There was, in this group of 316, a significant increase in family prevalence of gallstones among six of nine age-groups (P less than 0.05 and P less than 0.01, respectively) compared with those without gallstones. These data suggest a genetic disposition towards cholelithiasis, while dietary factors could not be definitely excluded.
Article
Limited information is available on the prevalence of gallstones in the first-degree relatives of gallstone patients. Three groups of subjects were studied by real-time ultrasound examination: group A, 105 index gallstone patients (male/female; 20:85); group B, 330 first-degree relatives of index patients; group C, matched controls for group A (n = 105) and group B (n = 330) subjects. Dietary, anthropometric, and biochemical investigations were carried out. In 39 of 105 (37%) index cases, one or more additional family members had gallstones (positive-index case). The positive-index cases were younger than the remaining index cases (mean age, 33.1 +/- 14 vs. 44.5 +/- 13.1 years; P < .05). Fifty-one of 330 (15.5%) first-degree relatives had gallstones, nearly four and a half times (95% confidence interval [CI], 2.4 to 8.5) more often than in the matched control population (12 of 330 [3.6%]). Thirty-three of 51 (65%) positive relatives were women; mother (37.3%), sister (17.6%) or daughters (10%) to the index patients. There was no difference in the diet, physical activity, and serum lipid profile between the positive index patients and the remaining gallstone patients and positive relatives and their controls. Our results show that there is a strong familial predisposition for gallstone formation. Female relatives of young gallstone patients should be routinely screened for gallstones.
Article
The prevalence of cholesterol gallstones differs among inbred strains of mice fed a diet containing 15% (wt/wt) dairy fat, 1% (wt/wt) cholesterol, and 0.5% (wt/wt) cholic acid. Strains C57L, SWR, and A were notable for a high prevalence of cholelithiasis; strains C57BL/6, C3H, and SJL had an intermediate prevalence; and strains SM, AKR, and DBA/2 exhibited no cholelithiasis after consuming the diet for 18 weeks. Genetic analysis of the difference in gallstone prevalence rates between strains AKR and C57L was carried out by using the AKXL recombinant inbred strain set and (AKR x C57L)F1 x AKR backcross mice. Susceptibility to gallstone formation was found to be a dominant trait determined by at least two genes. A major gene, named Lith1, mapped to mouse chromosome 2. When examined after 6 weeks on the lithogenic diet, the activity of hepatic 3-hydroxy-3-methylglutaryl-CoA reductase (EC 1.1.1.88) was downregulated as expected in the gallstone-resistant strains, AKR and SJL, but this enzyme failed to downregulate in C57L and SWR, the gallstone-susceptible strains. This suggests that regulation of the rate-limiting enzyme in cholesterol biosynthesis may be pivotal in determining the occurrence and severity of cholesterol hypersecretion and hence lithogenicity of gallbladder bile. These studies indicate that genetic factors are critical in determining gallstone formation and that the genetic resources of the mouse model may permit these factors to be identified.
Article
Since there have not been any studies that quantify the influence of genetic factors on gallbladder disease (GBD) in humans using information from families, we utilized pedigree data to explore the genetic control of variation in liability to GBD. Using an extension of a variance components approach, we performed genetic analyses of GBD using information from 32 low-income Mexican-American families with two slightly different general models incorporating several sex-specific GBD risk factors. After evaluating the relative magnitudes of the covariate effects from these two models, we identified a parsimonious model including only significant predictors of GBD. According to this model, heritability for GBD was high (h2 = 0.44+/-0.18), after accounting for the significant effects of age, leptin in both sexes, total cholesterol, and HDL cholesterol in males only. We have shown quantitatively that variation in GBD is under strong genetic control. However, there are two major limitations to our findings: (1) since GBD was defined by a self-reported clinical history rather than an ultrasound examination, the prevalence of GBD could have been underestimated; and (2) since our design did not allow for shared environmental effects, our estimate of heritability may have been inflated.
Article
Quantitative trait locus (QTL) mapping was used to locate genes that determine the difference in cholesterol gallstone disease between the gallstone-susceptible strain C57L/J and the gallstone-resistant strain AKR/J. Gallstone weight was determined in 231 male (AKR x C57L) F(1) x AKR backcross mice fed a lithogenic diet containing 1% cholesterol, 0.5% cholic acid, and 15% butterfat for 8 wk. Mice having no stones and mice having the largest stones were genotyped at approximately 20-cM intervals to find the loci determining cholesterol gallstone formation. The major locus, Lith1, mapped near D2Mit56 and was confirmed by constructing a congenic strain, AK. L-Lith1(s). Another locus, Lith2, mapped near D19Mit58 and was also confirmed by constructing a congenic strain AK.L-Lith2(s). Other suggestive, but not statistically significant, loci mapped to chromosomes 6, 7, 8, 10, and X. The identification of these Lith genes will elucidate the pathophysiology of cholesterol gallstone formation.
Article
Sitosterolemia is an autosomal recessive disorder caused by mutations in two adjacent genes encoding coordinately regulated ATP binding cassette (ABC) half transporters (ABCG5 and ABCG8). In this paper we describe three novel mutations causing sitosterolemia: 1) a frameshift mutation (c.336-337insA) in ABCG5 that results in premature termination of the protein at amino acid 197; 2) a missense mutation that changes a conserved residue c.1311C>G; N437K) in ABCG5 and 3) a splice site mutation in ABCG8 (IVS1-2A>G). This study expands the spectrum of the ABCG5 and ABCG8 mutations that cause sitosterolemia. Nine nonsynonymous polymorphisms are also reported: I523V, C600Y, Q604E, and M622V in ABCG5; and D19H, Y54C, T400K, A632V, and Y641F in ABCG8.