Báo cáo khoa học: ABCG transporters and disease pdf

11 272 0
Báo cáo khoa học: ABCG transporters and disease pdf

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

MINIREVIEW ABCG transporters and disease Owen M. Woodward 1 , Anna Ko ¨ ttgen 2,3 and Michael Ko ¨ ttgen 2,4 1 Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA 2 Renal Division, University Medical Centre Freiburg, Freiburg, Germany 3 Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA 4 Department of Nephrology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA ABCG family Members of the ABCG family are half transporters with one ABC cassette in the amino terminus followed by six putative transmembrane domains (see also reviews on other ABC transporters in the minireview series in this issue [1–3]). Full transporters contain two ABC cassettes and 12 transmembrane domains. Half transporters assemble to homodimeric and heterodi- meric complexes to form functional transporters. Fig- ure 1 provides an overview of the human ABC transporter superfamily and lists the members of the ABCG or White family, which is most closely related to the ABCA family. Currently, five members of the ABCG subfamily are known to exist in humans: ABCG1, ABCG2, ABCG4, ABCG5 and ABCG8. The ABCG1 gene is located on chromosome 21q22.3 [4]. Its product ABCG1 is found in multiple tissues and has a role in macrophage lipid transport [5]. ABCG2, mapped to chromosome 4q22, was initially identified in placenta tissue [6] and as a xenobiotic transporter from a human breast cancer cell line [7]. It was therefore also termed ‘breast cancer resistance pro- tein’ (BCRP). The ABCG4 gene is located on chromo- some 11q23.3 [8,9]. The gene product ABCG4 shows highest homology to ABCG1, and a role in macro- phage lipid metabolism has also been proposed [9]. The human ABCG5 and ABCG8 genes, located adja- cent to each other on chromosome 2p21, were both identified in the search for genetic causes of a rare autosomal-recessive lipid metabolism disorder, sitoster- olemia [10]. ABCG transporters and disease Members of the ABCG family are known to play a role in lipid transport across membranes. Loss-of-function mutations in ABCG5 or ABCG8 cause sitosterolemia, Keywords ABCG2; gout; GWAS; hyperuricemia; urate Correspondence M. Ko ¨ ttgen, Renal Division, University Medical Centre Freiburg, Freiburg, Germany Fax: +49 (0)761 27063240 Tel: +49 (0)761 27032990 E-mail: michael.koettgen@uniklinik- freiburg.de (Received 17 December 2010, revised 18 February 2011, accepted 6 May 2011) doi:10.1111/j.1742-4658.2011.08171.x ATP-binding cassette (ABC) transporters form a large family of transmem- brane proteins that facilitate the transport of specific substrates across membranes in an ATP-dependent manner. Transported substrates include lipids, lipopolysaccharides, amino acids, peptides, proteins, inorganic ions, sugars and xenobiotics. Despite this broad array of substrates, the physio- logical substrate of many ABC transporters has remained elusive. ABC transporters are divided into seven subfamilies, A–G, based on sequence similarity and domain organization. Here we review the role of members of the ABCG subfamily in human disease and how the identification of dis- ease genes helped to determine physiological substrates for specific ABC transporters. We focus on the recent discovery of mutations in ABCG2 causing hyperuricemia and gout, which has led to the identification of urate as a physiological substrate for ABCG2. Abbreviations ABC, ATP-binding cassette; SNP, single nucleotide polymorphism. FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS 3215 a disorder characterized by the accumulation of plant and fish sterols including cholesterol [10–12]. Clinical characteristics of sitosterolemia are xanthomatosis and premature atherosclerosis, resulting in early onset of cardiovascular disease and lethal myocardial infarction [13]. Mutations in ABCG5 or ABCG8 cause increased intestinal absorption and decreased biliary elimination of plant sterols and cholesterol, leading to a 50- to 200-fold increase in plasma plant sterol concentrations [13,14]. The encoded proteins ABCG5 and ABCG8 form obligate heterodimers that are expressed in the apical membrane of enterocytes and in the canicular membrane of hepatocytes [15]. They limit the absorp- tion of plant sterols and cholesterol by secreting these sterols from enterocytes back into the intestinal lumen, and by excretion of sterols from hepatocytes into bile. Disruption of ABCG5 and ABCG8 in mice results in a 3-fold increase in the fractional absorption of plant sterols, a 30% increase in plasma sitosterol levels, and a reduction in biliary cholesterol levels [16]. Thus these mice display many characteristics seen in patients with sitosterolemia. In accordance with the phenotypes observed upon disrupted function of ABCG5 and ABCG8 in humans or mice, it was recently shown that sterols are the direct substrates of ABCG5 and ABCG8. Inside-out membrane vesicles prepared from Sf9 insect cells overexpressing ABCG5 and ABCG8 or from liver membranes showed ATP- dependent transfer of both cholesterol and sitosterol [17,18]. To date no functional mutations in ABCG1 and ABCG4 have been linked to any monogenic human disease, although ABCG1 has been implicated in car- diovascular disease, obesity and diabetes (reviewed in [19]). Abcg1 ) ⁄ ) mice on a high-cholesterol diet display an attenuated endothelium-dependent arterial vasore- laxation as well as reduced activity of endothelial nitric oxide synthase, consistent with a role of ABCG1 in maintaining endothelial cell function by promoting efflux of cholesterol and 7-oxysterols [20]. In contrast, ABCG4 is highly expressed in the central nervous sys- tem. Detailed studies of the brains of Abcg4 ) ⁄ ) mice (< 1 year old) did not identify any pathological changes, however [19]. Both proteins have been shown to transport lipids including cholesterol, but their pre- cise role in vivo remains to be elucidated. It is of great interest whether future studies will establish a role for these transporters in inherited human disorders. Discovery of ABCG2 variants in association studies of human disease ABCG2 was first identified as a multidrug resistance protein (Fig. 2) [7]. It has been shown to transport a wide range of structurally and functionally diverse sub- strates such as chemotherapeutics, antibiotics and HMG-CoA reductase inhibitors. Yet, physiological substrates and the roles of ABCG2 in vivo had remained elusive until very recently. As was the case for ABCG5 and ABCG8, an important physiological function of ABCG2 was uncovered through genetic studies of human disease. In a series of genetic and physiological studies over the past 3 years, it was established that ABCG2 functions as a novel urate transporter that promotes urate excretion in the human kidney. A genome-wide association study among more than 11 000 individuals of European ancestry, including rep- lication in an additional 11 000 European ancestry and 3800 African American study participants, identified common alleles in ABCG2 as associated with serum urate levels and risk of gout [21]. Gout is a common form of arthritis with a prevalence of about 1–3% in western countries [22,23]. Patients with gout experience very painful attacks caused by the precipitation of monosodium urate crystals in joints, which triggers subsequent inflammation. Elevated serum urate levels are therefore a key risk factor for gout. Earlier studies showed that serum urate levels are highly heritable [24]. In fact, the majority of inter-individual variation of urate levels in a population can be explained by additive genetic effects. A genome-wide association study was initiated among individuals participating in three large, population-based prospective studies (Atherosclerosis Risk in Communities Study, Framing- ham Heart Study, Rotterdam Study) in an effort to discover genes that might explain the genetic effects on serum urate levels. Each study participant had serum urate levels measured and genotyping performed either ABCD ABCB ABCC(I) ABCC(II) ABCG ABCA ABCE ABCF ABCG2 ABCG1 ABCG4 ABCG5 ABCG8 Human ABC family members Human ABCGs Disease phenotype Gout, hyperuricemia Sitosterolemia Sitosterolemia ? ? Fig. 1. Phylogenetic tree of all human ABC genes and specifically the ABCG subgroup of genes (after [19,66]). Disease phenotypes reported include only human diseases associated with specific ABCG mutations, not information from model organisms. ABCG transporters and disease O. M. Woodward et al. 3216 FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS as part of a high-throughput single nucleotide poly- morphism (SNP) chip or as targeted replication geno- typing. Gout status was ascertained by self-report or based on the intake of gout-specific medication [21]. Of more than 500 000 SNPs surveyed, the ABCG2 var- iant with the strongest effect on serum urate concen- trations was the SNP rs2231142: each additional copy of the minor T allele was associated with mean serum urate concentrations approximately 0.25 standard devi- ations higher among individuals of European ancestry (P =3· 10 )60 ), corresponding to approximately 0.30 mgÆdL )1 higher mean serum urate per copy of the T allele (Table 1). The odds of gout were increased by 74% with each copy of the T allele (odds ratio 1.74, 95% confidence interval 1.51–1.99, P =4· 10 )15 ). The association between the risk allele and serum urate and gout was significantly stronger in men than in women [21,25]. Since this first study, the effect of the rs2231142 T allele on mean serum urate levels and the risk of gout has been replicated in many diverse study populations and is consistently observed with comparable effect sizes (Table 1). Replication of a finding in study popu- lations of different ancestry, where risk allele frequency and correlation patterns between nearby genomic vari- ants may differ, is an important feature of a functional genetic variant. Interestingly, the allele frequency of the T risk allele in a Japanese study population was reported as 31% [26], which is approximately three times more common than the T allele frequency observed in individuals of European ancestry. While the prevalence of gout in Japan is lower than in coun- tries where a western diet is consumed, the prevalence of gout among US individuals of Asian ancestry has been reported as three times higher than that of US individuals of European ancestry [27]. R L L A A M A T T T R V S G G G F I T Q R R V K K S G E A D R R V V K K L L G E E E I IN N N D H Q Q R V V V V V L L S G F E N M T T QD D S K R V K L L G F P C Y R K S G F P P C N N A V L L S G G G I N A D R K P P S G G G R V V K K K K L L L L L L S S S GG G PP E E I I II N NN M A A AT DD Y N E A I P E S I D L L F T L S G EIMT D I I P F C LR I H A N T T T T T G L D S S K K K L L L S G G G F F F F Q P P I M M A A A A D H G G L S S S V L L L L L R R R Q Q I I Y Y Y S S H E E A T V V V V L Q I S F I I I I A A L G G Y K F R S S E E I I L G Y Y Y Y V V K H S P C M M D R T I I I L L L F F Y V S S P F N T I A Q Q L L L G F Y Y H S S P R W C N M I I A A A L L G F V V K H W T L I F F C C C D D D A A A Q Q Q Q Q G G G G G G G G G G F F FF F F FF Y Y Y Y Y V V V V V V V V K K K K K K K K E E E E P P P P R W W T T T T T TT T T NN N N N N N N N M M M M L L L L L L L L L L L L I I I I I I A A A A A A A S S S S S S S S S S L L L L L L L L L L V V F G G CC T Q Q Q Q Y Y Y K K KK K H H E E E EEE E EE E P PP P R R R W N N N I I I II I I I A AAA A A A S S S SS S S LL L L L L L V V V V F F F F FF F G G G G C TT T T T T K K K K K KKK N N N LL D D D D DS S 395 469 565 644 414 450 495 505 584 625 Signature Walker A Walker B Q E P M I A V V V FF G G T NN N S S SS P F H E V F G C T T K N N L L D S S A A A I V12M N-terminus C-terminus M M M M M T A A A A L F F Y V V S S S F 524 476 Y Q126X G268R S441N F506fs Q141K 44 288 P P A A D D Fig. 2. Topographical representation of the ABCG2 monomer in the plasma membrane. Transmembrane domains experimentally determined by Wang et al. (2008) [67]; nucleotide binding domain (NBD) begins at Y44 and ends at residue N288 [68]. The Walker A and B and ABC sig- nature motif of the nucleotide binding domain are identified, as are the six human polymorphisms associated with hyperuricemia and gout (in red) [21,41]. Amino acid residues: pink, aromatic; green, + charged; light blue, ) charged; white, nonpolar; yellow, polar residues. O. M. Woodward et al. ABCG transporters and disease FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS 3217 Physiological function of ABCG2 A connection between ABCG2 and urate metabolism or gout had not been described until this first genome- wide association study. It was known, however, that human ABCG2 is expressed in the apical membrane of human proximal tubule cells [28], the main site of urate handling in the human kidney. We therefore investigated whether urate is a physiological substrate of ABCG2, and whether the Q141K variant, encoded by rs2231142, leads to altered urate transport and as a consequence to elevated serum urate levels and increased risk of gout. In order to test whether ABCG2 was a yet unknown urate transporter, ABCG2 was expressed in Xenopus oocytes [29]. Accumulation of radiolabeled urate in oo- cytes expressing ABCG2 was decreased by 75% com- pared with water-injected control oocytes (Fig. 3A). The reduced urate accumulation was caused by ABCG2-mediated urate efflux from cells rather than by the inhibition of urate uptake, as shown in experi- ments monitoring the decrease of intracellular urate over time in oocytes preloaded with radiolabeled urate (Fig. 3B). Although it was known that the major site of urate excretion in humans is the proximal tubule in the kidney, the molecular identity of the transporters mediating urate secretion at the apical membrane of proximal tubular cells had only been poorly under- stood. To study ABCG2 function at this location, urate accumulation and localization of ABCG2 was studied in native LLC-PK1 cells, a porcine proximal tubule cell line. These experiments revealed that ABCG2 mediates the apical secretion of urate in proxi- mal tubule cells (Fig. 3D). A similar function and localization has been shown for MRP4 [30,31], but polymorphisms in MRP4 have not been linked to hyperuricemia and gout in humans. Given the vast literature on ABCG2 with dozens of structurally diverse substrates it appears surprising at first glance that urate was not found to be a physiolog- ical substrate earlier. Notably, ABCG2 knockout mice do not develop gout. One of the reasons for urate stay- ing under the radar of ABCG2 research may be that gout is a complex genetic disease with multiple contrib- uting genetic and environmental factors. More impor- tantly though, there are striking species differences in purine metabolism within the animal kingdom. Urate is the end product of purine metabolism in humans. Humans and higher primates have much higher serum urate levels than other mammals because they lack the enzyme uricase, which converts urate into allantoin [32]. Therefore genetic factors that predispose to hyperuricemia and gout cannot be easily studied in rodent models. Q141K is a functional variant in ABCG2 Several lines of evidence in the initial genome-wide association study by Dehghan et al. [21] suggested that the rs2231142 variant may be functional. First, Table 1. Effect sizes of the ABCG2 rs2231142 (Q141K) variant on risk of gout and mean urate levels in study populations of different ancestry. Study sample ethnicity Sample size Risk allele frequency (T) Odds ratio for gout per T allele, 95% CI Effect on mean serum urate per T allele Ref. European ancestry 22 871 0.11 1.74 0.24 standard deviation changes [21] European ancestry 28 141 0.11 NA 0.17 z-score units [44] European ancestry 28 283 0.11 a 1.86 0.30 mgÆdL )1 [45] European ancestry 4492 0.11–0.12 NA 0.34 mgÆdL )1 [62] European ancestry 2246 0.1 (controls), 0.14 (cases) 1.37 NA [63] African American 3843 0.03 1.71 0.22 standard deviations [21] Japanese 739 0.32 2.5 in a subset of gout patients  0.4 mgÆdL )1 [41] Japanese 3923 0.31 1.37 for genotype TG, 4.37 for genotype TT [26] Japanese 5165 0.23–0.30 NA 0.1 mgÆdL )1 per risk allele [64] New Zealand population Cases ⁄ controls: 185 ⁄ 284 Maori, 173 ⁄ 129 Pacific Islanders, 214 ⁄ 562 Caucasian 1.08 Maori, 2.80 Pacific Islanders, 2.20 Caucasian [65] a highly correlated SNP rs2199936 was studied (r2 = 0.92 in HapMap CEU r22). ABCG transporters and disease O. M. Woodward et al. 3218 FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS the variant is located in exon 5 of ABCG2 and leads to a glutamine-to-lysine amino acid substitution (Q141K) in ABCG2. This substitution is predicted to have a possibly damaging effect by the functional prediction program polyphen-2 [33]. Second, the glu- tamine residue at position 141 is highly conserved across species. No other common variants in the ABCG2 gene region showed association with serum urate levels after accounting for the effect of rs2231142 [21,29]. However, while genome-wide association studies have been extremely successful at establishing associa- tions between common SNPs and a multitude of com- plex diseases [34], these studies cannot establish whether a disease-associated SNP is causally related to the disease or merely a naturally occurring genetic marker that is correlated with another, unknown func- tional variant. To test whether the rs2231142 is such a functional variant, the transport capacity of the encoded Q141K mutation was compared with that of wild-type ABCG2. Oocytes expressing ABCG2 Q141K showed 54% reduced urate transport rates compared with oocytes expressing wild-type ABCG2 (Fig. 3C). This is consistent with previous studies showing impaired transport of chemotherapeutic agents by ABCG2 Q141K [35,36] (and reviewed in [37]). While it is difficult to compare the results from different trans- port assays and substrates, the reduction of transport of the Q141K variant compared with wild-type ABCG2 appears to be of similar magnitude. The Q141 residue is located in the nucleotide binding domain of ABCG2 (Fig. 2), and Q141K ABCG2 expression is sig- nificantly lower than wild-type when overexpressed in mammalian cells [35,36,38,39]. Interestingly, the F508 mutation in CFTR, a related ABC transporter, is located right next to this position in the nucleotide binding domain and is commonly mutated in cystic fibrosis patients [40]. And like the Q141K ABCG2 mutation, expression of the deleted F508 CFTR mutant is significantly lower than wild-type suggesting a common pathophysiology (Woodward, unpublished observations). 0.0 0.5 1.0 1.5 Urate accumulation pmol per oocyte·120 min –1 Urate accumulation pmol per oocyte·120 min –1 H 2 O ABCG2 ∗∗ A 0204060 0.4 0.6 0.8 1.0 Relative urate remaining Time (min) ∗∗ ∗∗ ∗∗ ∗∗ ∗∗ B Lumen Blood 0.0 0.3 0.6 0.9 WT Q141K ∗∗ C Others 3 Others 4 SLC2A9 URAT1 SLC2A9 Others 1 Others 2 U- U- U- U- U- U- U- U- U- U- U- U- U- U- U- U- U- U- U- D ABCG2 Fig. 3. ABCG2 is a urate transporter. (A) C-14 urate accumulation from Xenopus oocytes injected with mRNA coding for either ABCG2 or H 2 O controls. (B) Urate efflux in oocytes incubated overnight in 500 lM C-14 urate as relative efflux over time (blue, control; red, ABCG2). (C) Urate accumulation in oocytes expressing either the wild-type ABCG2 or the mutant Q141K ABCG2. (**P < 0.01, ± SEM) (A–C originally from [29]; ª 2009 by the National Academy of Sciences of the USA). (D) Model of urate transport in the proximal tubule of the human kidney overlying fluorescent micrograph of LLCPK-1 proximal tubule cell with endogenous ABCG2 labeled in green and the nucleus in blue. Proteins influencing urate absorption and secretion and with significance for human diseases are shown with the direction of urate transport indicated [21,69,70]. Other transporters expressed in the human kidney and shown to transport urate in model systems: 1 OAT4; 2 OAT1, OAT3; 3 MRP4; 4 OAT1, OAT3 [71,72]. O. M. Woodward et al. ABCG transporters and disease FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS 3219 The role of ABCG2 as a urate transporter with mutations leading to hyperuricemia and gout was recently confirmed and further investigated by Matsuo et al. [41]. The investigators of this study identified sev- eral non-synonymous coding variants in ABCG2 through sequencing of the ABCG2 gene in 90 hyperuri- cemia patients in a Japanese population. In addition to Q141K, Q126X was identified as a novel loss-of-func- tion variant. Q126X was assigned to a different haplo- type than Q141K and shown to increase gout risk (odds ratio 5.97) to an even greater extent than the Q141K variant. In addition, 10% of the gout patients studied had genotype combinations of the Q141K and Q126X variants that resulted in more than a 75% reduction of ABCG2 function compared with patients that were homozygous for the non-risk allele at both variants (odds ratio 25.8, 95% confidence interval 10.3–64.6). Many additional SNPs and their role in ABCG2 function have been analyzed [37,42], but these studies have not addressed the impact of other SNPs in urate transport and gout. Future studies will have to test whether additional functional SNPs also affect serum urate concentrations in humans. Urate transport is complex: in the kidney, urate transport is bidirectional and involves multiple differ- ent transport and regulatory proteins [32,43]. This is reflected in the complex genetic architecture of serum urate levels and risk of gout: two recent large gen- ome-wide association studies identified variants in multiple genes associated with serum urate concentra- tions (SLC2A9, ABCG2, SLC17A1, SLC22A11, SLC22A12, SLC16A9, GCKR, LRRC16A, PDZK1, the R3HDM2–INHBC region and RREB1) [44,45]. The effect of the individual common risk alleles in these genes on mean serum urate concentrations and the risk of gout is modest. The range of the pheno- typic variation in serum urate levels in the studied populations that could be explained by the individual genetic variants ranged from 0.1% to 3.5%. However, the effect of urate-increasing alleles at different geno- mic loci can add up: Yang et al. [45] estimated from several large population-based studies that mean urate levels increased from approximately 4.5 to 6.2 mgÆdL )1 across a genetic score composed of the risk alleles at eight different genomic loci. Similarly, the prevalence of gout increased from 2% to more than 20% at the upper extreme of the risk score. Some of the genes identified in the two large studies mentioned above encode for known urate transporters (SLC2A9, ABCG2, SLC17A1, SLC22A11, SLC22A12) or regulators thereof (PDZK1). For the remaining genes, little is known about a possible connection of the gene product to urate metabolism in humans and therefore this constitutes a new area for future research. ABCG2 function in other tissues ABCG2’s physiological function has been difficult to identify because of the large number of known sub- strates and varied tissue expression. Suggested physio- logical roles include functioning as a xenobiotic transporter, conferring xenobiotic protection in tissues like the liver, intestine, placenta and CNS [37]; and as a transporter of heme and other porphyrins, prevent- ing their accumulation in erythrocytes and stem cells [46,47]. As noted above ABCG2 plays a significant role in urate transport in the human kidney, but does ABCG2 expression in other tissues fit with this newly postulated function? Here we would like to discuss the putative physiological role of ABCG2-mediated urate transport in other tissues. In addition to the kidney, ABCG2 is expressed at high levels in the liver, at the blood–brain barrier, in the placenta and in mammary glands. An examination of ABCG2 at each of these locations suggests that ABCG2 expression is consistent with sites of urate transport. In human hepatocytes, ABCG2 is expressed in the basolateral membrane [48] oriented to mediate efflux into the biliary canaliculus. Though ABCG2 is effectively situated to remove drugs and toxins from the liver, it is also well situated to export urate out of the liver via the biliary system, a known urate excretion pathway [49]. ABCG2, in addi- tion to the urate transporter MRP4 [31], are the only identified urate transporters positioned to secrete urate into the biliary system, and thus ABCG2 could be playing a substantial role in the liver-mediated urate excretion pathway. At the blood–brain barrier, ABCG2 is expressed on the luminal membrane of endothelial cells, seemingly well positioned to protect the brain from accumulating xenotoxins [50]. However, there is also ample evidence that misregulation of urate at the blood–brain barrier has profound effects on brain function and health. Cerebrospinal fluid urate levels and serum urate levels are correlated [51,52] but urate concentration in cerebrospinal fluid is only 7% of that in serum [52], suggesting an important role for urate secretion from the cerebrospinal fluid. Higher serum urate levels are associated with cognitive dys- function [53] but are also protective against developing Parkinson’s disease [52]. Thus a tight regulation of cerebrospinal fluid urate appears important. High expression of ABCG2 at the blood–brain barrier may help maintain appropriate urate concentrations in the brain and the cerebrospinal fluid. ABCG transporters and disease O. M. Woodward et al. 3220 FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS Pregnancy has a profound effect on ABCG2 expression at two sites. First, ABCG2 is expressed highly in the apical membrane of placental syncytio- trophoblasts and is hypothesized to aid in the protec- tion of the fetus from toxins or to regulate fetal estrogen levels by transporting estrogen precursor molecules [54]. However, ABCG2-mediated efflux of urate from the placenta may be critical for normal fetal development. It was recently reported that high urate levels in amniotic fluid correlated with lower birth weights, finding a 2 mgÆdL )1 decrease in amni- otic urate results in a 120 g increase in birth weight [55]. Second, pregnancy and lactation increases ABCG2 expression in mammary gland alveolar epi- thelial cells. This can result in the concentrating of xenotoxins, if present in the mother, into breast milk [56], a seemingly undesirable outcome for a nursing infant. This apparent contradiction prompted the pro- posal that ABCG2 may be mostly transporting non- toxic substitutes like riboflavin [57]. Yet ABCG2 knockout models show no reduction of this vitamin in breast milk [58]. In contrast, there is some evidence that human breast milk plays an important role in delivering antioxidants, including urate, to infants [59]. Interestingly, while human breast milk contains urate, it does not contain orotic acid, which is found in high concentrations in other mammalian milk [60,61]. Orotic acid is a strong uricosuric compound, and its disappearance from human milk is consistent with the evolutionarily conserved loss of uricase rs2231142 P = 4*10 –27 0 20 40 60 30 25 20 15 10 5 0 IBSP MEPE SPP1 PKD2 ABCG2 PPM1K Disease GWAS Physiology Treatment ? 0 2 46810 0.00 0.05 0.10 Efflux: (pmol per oocyte·min –1 ) Internal oocyte concentration (µM) Fig. 4. The cycle of translational research can begin with the description of a disease phenotype like the destruction of joints that occurs in patients with gout from urate crystal deposition. Genome-wide association studies allow the identification of genes that associate with ele- vated serum urate levels and gout. Subsequent in-depth physiological characterization of the gene and its protein product lays the foundation for an improved understanding of physiology and pathophysiology and may reveal a therapeutic target. Finally, drug development can be attempted in order to better treat hyperuricemia or gout (X-ray kindly provided by Janet Maynard). O. M. Woodward et al. ABCG transporters and disease FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS 3221 function to increase urate levels in humans. In sum- mary, a role of ABCG2-mediated urate secretion in several non-renal tissues is conceivable and needs to be investigated in more detail. Pharmacological modulation of ABCG2, both inhi- bition and activation, has been proposed as therapeutic strategies for numerous human diseases. For instance, inhibition of ABCG2 has been tested to overcome multidrug resistance in cancer therapy. However, based on the function of ABCG2 in urate excretion, one pos- sible side effect of ABCG2 inhibitors could be increased serum urate concentrations and gout attacks. Further studies on ABCG2 are needed to learn more about its function in different tissues and the relevance of additional physiological substrates. These studies may help to predict therapeutic effects as well as side effects of drugs targeting ABCG2. Future perspectives and conclusion In summary, mutations in members of the ABCG family have led to the identification of physiological substrates and functions of these transporters. We anticipate that future studies will continue to uncover additional novel physiological substrates and functions for ABC transporters and define additional roles in human disease. The powerful combination of genetic and physiological approaches not only may identify novel mechanisms but may also help to identify novel therapeutic targets. ABCG2 represents an attractive drug target since pharmacological acti- vation of ABCG2 may help to promote urate excre- tion from the body. The discovery of ABCG2 as a novel urate transporter is a prime example for trans- lational research. Hopefully, the fast translation from bedside to bench will eventually lead back to the bedside and benefit patients suffering from gout (Fig. 4). Acknowledgements We acknowledge the work of many others whose work we could not cite due to space constraints. O.M.W. was supported by NIDDK: DK032753-25A1, A.K. was supported by the Emmy Noether programme of DFG and M.K. was supported by DFG KFO 201 and by Alfried Krupp von Bohlen und Halbach Foundation. References 1 Nagao K, Tomioka M & Ueda K (2011) Function and regulation of ABCA1 – membrane meso-domain organization and reorganization. FEBS J 278, 3190–3203. 2 Pollock NL & Callaghan R (2011) The lipid translo- case, ABCA4: seeing is believing. FEBS J 278, 3204– 3214. 3 Chen Z-S (2011) Multidrug resistance proteins (MRPs ⁄ ABCCs) in cancer chemotherapy and genetics disease. FEBS J 278, 3226–3245. 4 Chen H, Rossier C, Lalioti MD, Lynn A, Chakravarti A, Perrin G & Antonarakis SE (1996) Cloning of the cDNA for a human homologue of the Drosophila white gene and mapping to chromosome 21q22.3. Am J Hum Genet 59, 66–75. 5 Klucken J, Buchler C, Orso E, Kaminski WE, Porsch- Ozcurumez M, Liebisch G, Kapinsky M, Diederich W, Drobnik W, Dean M et al. (2000) ABCG1 (ABC8), the human homolog of the Drosophila white gene, is a reg- ulator of macrophage cholesterol and phospholipid transport. Proc Natl Acad Sci USA 97, 817–822. 6 Allikmets R, Schriml LM, Hutchinson A, Romano- Spica V & Dean M (1998) A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res 58, 5337–5339. 7 Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK & Ross DD (1998) A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci USA 95, 15665–15670. 8 Annilo T, Tammur J, Hutchinson A, Rzhetsky A, Dean M & Allikmets R (2001) Human and mouse orthologs of a new ATP-binding cassette gene, ABCG4. Cytogenet Cell Genet 94, 196–201. 9 Engel T, Lorkowski S, Lueken A, Rust S, Schluter B, Berger G, Cullen P & Assmann G (2001) The human ABCG4 gene is regulated by oxysterols and retinoids in monocyte-derived macrophages. Biochem Biophys Res Commun 288, 483–488. 10 Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, Kwiterovich P, Shan B, Barnes R & Hobbs HH (2000) Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science 290, 1771–1775. 11 Dean M (2005) The genetics of ATP-binding cassette transporters. Methods Enzymol 400, 409–429. 12 Lee MH, Lu K, Hazard S, Yu H, Shulenin S, Hidaka H, Kojima H, Allikmets R, Sakuma N, Pegoraro R et al. (2001) Identification of a gene, ABCG5, important in the regulation of dietary cholesterol absorption. Nat Genet 27, 79–83. 13 Sudhop T & von Bergmann K (2004) Sitosterolemia – a rare disease. Are elevated plant sterols an additional risk factor? Z Kardiol 93, 921–928. 14 Berge KE (2003) Sitosterolemia: a gateway to new knowledge about cholesterol metabolism. Ann Med 35, 502–511. ABCG transporters and disease O. M. Woodward et al. 3222 FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS 15 Graf GA, Li WP, Gerard RD, Gelissen I, White A, Cohen JC & Hobbs HH (2002) Coexpression of ATP- binding cassette proteins ABCG5 and ABCG8 permits their transport to the apical surface. J Clin Invest 110, 659–669. 16 Yu L, Hammer RE, Li-Hawkins J, Von Bergmann K, Lutjohann D, Cohen JC & Hobbs HH (2002) Disrup- tion of Abcg5 and Abcg8 in mice reveals their crucial role in biliary cholesterol secretion. Proc Natl Acad Sci USA 99, 16237–16242. 17 Wang J, Sun F, Zhang DW, Ma Y, Xu F, Belani JD, Cohen JC, Hobbs HH & Xie XS (2006) Sterol transfer by ABCG5 and ABCG8: in vitro assay and reconstitu- tion. J Biol Chem 281, 27894–27904. 18 Wang J, Zhang DW, Lei Y, Xu F, Cohen JC, Hobbs HH & Xie XS (2008) Purification and reconstitution of sterol transfer by native mouse ABCG5 and ABCG8. Biochemistry 47, 5194–5204. 19 Tarr PT, Tarling EJ, Bojanic DD, Edwards PA & Baldan A (2009) Emerging new paradigms for ABCG transporters. Biochim Biophys Acta 1791, 584–593. 20 Terasaka N, Yu S, Yvan-Charvet L, Wang N, Mzhavia N, Langlois R, Pagler T, Li R, Welch CL, Goldberg IJ et al. (2008) ABCG1 and HDL protect against endothe- lial dysfunction in mice fed a high-cholesterol diet. J Clin Invest 118, 3701–3713. 21 Dehghan A, Kottgen A, Yang Q, Hwang SJ, Kao WL, Rivadeneira F, Boerwinkle E, Levy D, Hofman A, Astor BC et al. (2008) Association of three genetic loci with uric acid concentration and risk of gout: a gen- ome-wide association study. Lancet 372, 1953–1961. 22 Choi HK, Mount DB & Reginato AM (2005) Patho- genesis of gout. Ann Intern Med 143, 499–516. 23 Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA, Gabriel S, Hirsch R, Hochberg MC, Hunder GG et al. (2008) Estimates of the preva- lence of arthritis and other rheumatic conditions in the United States. Part II. Arthritis Rheum 58, 26–35. 24 Yang Q, Guo CY, Cupples LA, Levy D, Wilson PW & Fox CS (2005) Genome-wide search for genes affecting serum uric acid levels: the Framingham Heart Study. Metabolism 54, 1435–1441. 25 Tanaka Y, Slitt AL, Leazer TM, Maher JM & Klaassen CD (2005) Tissue distribution and hormonal regulation of the breast cancer resistance protein (Bcrp ⁄ Abcg2) in rats and mice. Biochem Biophys Res Commun 326, 181–187. 26 Yamagishi K, Tanigawa T, Kitamura A, Kottgen A, Folsom AR & Iso H (2010) The rs2231142 variant of the ABCG2 gene is associated with uric acid levels and gout among Japanese people. Rheumatology (Oxford) 49, 1461–1465. 27 Krishnan E, Lienesch D & Kwoh CK (2008) Gout in ambulatory care settings in the United States. J Rheumatol 35, 498–501. 28 Huls M, Brown CD, Windass AS, Sayer R, van den Heuvel JJ, Heemskerk S, Russel FG & Masereeuw R (2008) The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. Kidney Int 73, 220–225. 29 Woodward OM, Kottgen A, Coresh J, Boerwinkle E, Guggino WB & Kottgen M (2009) Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci USA 106, 10338–10342. 30 van Aubel RA, Smeets PH, Peters JG, Bindels RJ & Russel FG (2002) The MRP4 ⁄ ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: putative efflux pump for urinary cAMP and cGMP. J Am Soc Nephrol 13, 595–603. 31 Van Aubel RA, Smeets PH, van den Heuvel JJ & Russel FG (2005) Human organic anion transporter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple allosteric substrate bind- ing sites. Am J Physiol Renal Physiol 288, F327–333. 32 Anzai N, Kanai Y & Endou H (2007) New insights into renal transport of urate. Curr Opin Rheumatol 19, 151–157. 33 Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS & Sunyaev SR (2010) A method and server for predicting damaging missense mutations. Nat Methods 7, 248–249. 34 Manolio TA (2010) Genomewide association studies and assessment of the risk of disease. N Engl J Med 363, 166–176. 35 Mizuarai S, Aozasa N & Kotani H (2004) Single nucle- otide polymorphisms result in impaired membrane localization and reduced atpase activity in multidrug transporter ABCG2. Int J Cancer 109, 238–246. 36 Morisaki K, Robey RW, Ozvegy-Laczka C, Honjo Y, Polgar O, Steadman K, Sarkadi B & Bates SE (2005) Single nucleotide polymorphisms modify the transporter activity of ABCG2. Cancer Chemother Pharmacol 56, 161–172. 37 Polgar O, Robey RW & Bates SE (2008) ABCG2: structure, function and role in drug response. Expert Opin Drug Metab Toxicol 4, 1–15. 38 Furukawa T, Wakabayashi K, Tamura A, Nakagawa H, Morishima Y, Osawa Y & Ishikawa T (2009) Major SNP (Q141K) variant of human ABC transporter ABCG2 undergoes lysosomal and proteasomal degrada- tions. Pharm Res 26, 469–479. 39 Imai Y, Nakane M, Kage K, Tsukahara S, Ishikawa E, Tsuruo T, Miki Y & Sugimoto Y (2002) C421A poly- morphism in the human breast cancer resistance protein gene is associated with low expression of Q141K protein and low-level drug resistance. Mol Cancer Ther 1, 611–616. 40 Fuller CM & Benos DJ (1992) Cftr!. Am J Physiol 263, C267–286. O. M. Woodward et al. ABCG transporters and disease FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS 3223 41 Matsuo H, Takada T, Ichida K, Nakamura T, Nakayama A, Ikebuchi Y, Ito K, Kusanagi Y, Chiba T, Tadokoro S et al. (2009) Common defects of ABCG2, a high-capacity urate exporter, cause gout: a function-based genetic analysis in a Japanese popula- tion. Sci Transl Med 1, 5ra11. 42 Koshiba S, An R, Saito H, Wakabayashi K, Tamura A & Ishikawa T (2008) Human ABC transporters ABCG2 (BCRP) and ABCG4. Xenobiotica 38, 863–888. 43 So A & Thorens B (2010) Uric acid transport and disease. J Clin Invest 120, 1791–1799. 44 Kolz M, Johnson T, Sanna S, Teumer A, Vitart V, Perola M, Mangino M, Albrecht E, Wallace C, Farrall M et al. (2009) Meta-analysis of 28,141 individuals identifies common variants within five new loci that influence uric acid concentrations. PLoS Genet 5, e1000504. 45 Yang Q, Kottgen A, Dehghan A, Smith AV, Glazer NL, Chen MH, Chasman DI, Aspelund T, Eiriksdottir G, Harris TB et al. (2010) Multiple genetic loci influ- ence serum urate and their relationship with gout and cardiovascular disease risk factors. Circ Cardiovasc Genet 3, 523–530. 46 Jonker JW, Buitelaar M, Wagenaar E, Van Der Valk MA, Scheffer GL, Scheper RJ, Plosch T, Kuipers F, Elferink RP, Rosing H et al. (2002) The breast cancer resistance protein protects against a major chlorophyll- derived dietary phototoxin and protoporphyria. Proc Natl Acad Sci USA 99, 15649–15654. 47 Krishnamurthy P, Ross DD, Nakanishi T, Bailey-Dell K, Zhou S, Mercer KE, Sarkadi B, Sorrentino BP & Schuetz JD (2004) The stem cell marker Bcrp ⁄ ABCG2 enhances hypoxic cell survival through interactions with heme. J Biol Chem 279, 24218–24225. 48 Ieiri I, Higuchi S & Sugiyama Y (2009) Genetic polymorphisms of uptake (OATP1B1, 1B3) and efflux (MRP2, BCRP) transporters: implications for inter-indi- vidual differences in the pharmacokinetics and pharma- codynamics of statins and other clinically relevant drugs. Expert Opin Drug Metab Toxicol 5, 703–729. 49 Kountouras J, Magoula I, Tsapas G & Liatsis I (1996) The effect of mannitol and secretin on the biliary transport of urate in humans. Hepatology 23 , 229–233. 50 Scherrmann JM (2005) Expression and function of multidrug resistance transporters at the blood–brain barriers. Expert Opin Drug Metab Toxicol 1, 233–246. 51 Bowman GL, Shannon J, Frei B, Kaye JA & Quinn JF (2010) Uric acid as a CNS antioxidant. J Alzheimers Dis 19, 1331–1336. 52 Schlesinger I & Schlesinger N (2008) Uric acid in Parkinson’s disease. Mov Disord 23, 1653–1657. 53 Vannorsdall TD, Jinnah HA, Gordon B, Kraut M & Schretlen DJ (2008) Cerebral ischemia mediates the effect of serum uric acid on cognitive function. Stroke 39, 3418–3420. 54 Mao Q (2008) BCRP ⁄ ABCG2 in the placenta: expres- sion, function and regulation. Pharm Res 25, 1244– 1255. 55 Gao T, Zablith NR, Burns DH, Skinner CD & Koski KG (2008) Second trimester amniotic fluid transferrin and uric acid predict infant birth outcomes. Prenat Diagn 28, 810–814. 56 Jonker JW, Merino G, Musters S, van Herwaarden AE, Bolscher E, Wagenaar E, Mesman E, Dale TC & Schinkel AH (2005) The breast cancer resistance protein BCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. Nat Med 11, 127–129. 57 van Herwaarden AE & Schinkel AH (2006) The func- tion of breast cancer resistance protein in epithelial bar- riers, stem cells and milk secretion of drugs and xenotoxins. Trends Pharmacol Sci 27, 10–16. 58 van Herwaarden AE, Wagenaar E, Merino G, Jonker JW, Rosing H, Beijnen JH & Schinkel AH (2007) Mul- tidrug transporter ABCG2 ⁄ breast cancer resistance pro- tein secretes riboflavin (vitamin B2) into milk. Mol Cell Biol 27, 1247–1253. 59 Aycicek A, Erel O, Kocyigit A, Selek S & Demirkol MR (2006) Breast milk provides better antioxidant power than does formula. Nutrition 22, 616–619. 60 Ferreira IM (2003) Quantification of non-protein nitro- gen components of infant formulae and follow-up milks: comparison with cows’ and human milk. Br J Nutr 90, 127–133. 61 Indyk HE & Woollard DC (2004) Determination of orotic acid, uric acid, and creatinine in milk by liquid chromatography. J AOAC Int 87, 116–122. 62 Brandstatter A, Lamina C, Kiechl S, Hunt SC, Coassin S, Paulweber B, Kramer F, Summerer M, Willeit J, Kedenko L et al. (2010) Sex and age interaction with genetic association of atherogenic uric acid concentra- tions. Atherosclerosis 210, 474–478. 63 Stark K, Reinhard W, Grassl M, Erdmann J, Schunkert H, Illig T & Hengstenberg C (2009) Common polymor- phisms influencing serum uric acid levels contribute to susceptibility to gout, but not to coronary artery disease. PLoS ONE 4, e7729. 64 Tabara Y, Kohara K, Kawamoto R, Hiura Y, Nishim- ura K, Morisaki T, Kokubo Y, Okamura T, Tomoike H, Iwai N et al. (2010) Association of four genetic loci with uric acid levels and reduced renal function: the J- SHIPP Suita study. Am J Nephrol 32, 279–286. 65 Phipps-Green AJ, Hollis-Moffatt JE, Dalbeth N, Merri- man ME, Topless R, Gow PJ, Harrison AA, Highton J, Jones PB, Stamp LK et al. (2010) A strong role for the ABCG2 gene in susceptibility to gout in New Zealand Pacific Island and Caucasian, but not Maori, case and control sample sets. Hum Mol Genet 19, 4813–4819. 66 Dean M (2002) The human ATP-binding cassette (ABC) transporter superfamily [Internet]. National Library of Medicine (US)(ID:NBK3), NCBI, Bethesda, ABCG transporters and disease O. M. Woodward et al. 3224 FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation ª 2011 FEBS [...]... anion transport- ABCG transporters and disease ers in the tubular transport of indoxyl sulfate and the induction of its nephrotoxicity J Am Soc Nephrol 13, 1711–1720 70 Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CN, Knott SA, Kolcic I, Polasek O, Graessler J et al (2008) SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout Nat Genet... (2008) Membrane topology of the human breast cancer resistance protein (BCRP ⁄ ABCG2 ) determined by epitope insertion and immunofluorescence Biochemistry 47, 13778–13787 68 Li YF, Polgar O, Okada M, Esser L, Bates SE & Xia D (2007) Towards understanding the mechanism of action of the multidrug resistance-linked half-ABC transporter ABCG2 : a molecular modeling study J Mol Graph Model 25, 837–851 69 Enomoto... concentration, urate excretion and gout Nat Genet 40, 437–442 71 Enomoto A & Endou H (2005) Roles of organic anion transporters (OATs) and a urate transporter (URAT1) in the pathophysiology of human disease Clin Exp Nephrol 9, 195–205 72 Wright AF, Rudan I, Hastie ND & Campbell H (2010) A ‘complexity’ of urate transporters Kidney Int 78, 446–452 FEBS Journal 278 (2011) 3215–3225 ª 2011 The Authors Journal compilation . measured and genotyping performed either ABCD ABCB ABCC(I) ABCC(II) ABCG ABCA ABCE ABCF ABCG2 ABCG1 ABCG4 ABCG5 ABCG8 Human ABC family members Human ABCGs Disease. humans: ABCG1 , ABCG2 , ABCG4 , ABCG5 and ABCG8 . The ABCG1 gene is located on chromosome 21q22.3 [4]. Its product ABCG1 is found in multiple tissues and has a

Ngày đăng: 22/03/2014, 15:21

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan