Skip to main content
Erschienen in: BMC Pediatrics 1/2019

Open Access 01.12.2019 | Research article

SLCO1B1 c.388A > G variant incidence and the severity of hyperbilirubinemia in Indonesian neonates

verfasst von: Radhian Amandito, Rinawati Rohsiswatmo, Michelle Halim, Vanessa Tirtatjahja, Amarila Malik

Erschienen in: BMC Pediatrics | Ausgabe 1/2019

Abstract

Objective

It has been established that genetic factors play a substantial role in the development of neonatal hyperbilirubinemia. The population of Indonesia and other Southeast Asian countries has similar, yet different genetic makeup compared to the rest of Asia. Aside from UGT1A1, variants of SLCO1B1 have also been known to contribute to the severity of neonatal hyperbilirubinemia in Asian populations. Since there has been no report on SLCO1B1 polymorphism in relation with hyperbilirubinemia in Indonesia, this study aims to explore incidence of SLCO1B1*1B polymorphism in Indonesia based on 3 hospitals from different provinces and population, and their association with hyperbilirubinemia severity.

Methods

Our study included 88 neonates with mild and moderate-severe hyperbilirubinemia from 3 NICU in hospitals representing homogenous and heterogenous populations: Biak General Hospital Papua, Cipto Mangunkusumo Hospital (Jakarta), and M Yunus Hospital (Bengkulu). We collected samples between November 2016 and September 2017. DNA was obtained from existing samples of the patients from previous studies and were subjected to Polymerase Chain Reaction – Restriction Fragment Length Polymorphism (PCR-RFLP). We analyzed the *1B variant located in exon 5 of SLCO1B1 with TaqI restriction endonuclease. Clinical, demographic, and laboratory data was also collected from medical records and parents’ interviews.

Results

The most dominant variant of SLCO1B1*1B in our population is the homozygous G/G (68.18%), followed by heterozygous A/G (26.14%), and wild type A/A (5.68%). The heterozygous A/G had an Odds Ratio (OR) of 0.73 (95% CI 0.10–5.2) and homozygous G/G with OR of 0.51 (95%CI 0.08–3.27), both were not significant. Genotypic distribution across the different centers were also similar and not significant. The significant risk factors for moderate-severe hyperbilirubinemia were the population the neonate originated from (p = < 0.001) and the delivery location (p = 0.001), while SLCO1B1*1B was not associated with the different severity of hyperbilirubinemia.

Conclusions

SLCO1B1*1B is not associated with higher bilirubin levels among neonates with hyperbilirubinemia in Indonesia. Further study is needed to find other potentially important genetic polymorphisms in the development of severe hyperbilirubinemia in Indonesia.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CI
Confidence Interval
CMH
Cipto Mangunkusumo Hospital
PCR–RFLP
polymerase chain reaction–restriction fragment length polymorphism
SD
Standard deviation
SLCO1B1
Solute Carrier Organic Anion Transporter Family Member 1b1
SPSS
Statistical package for the social sciences
TSB
Total serum bilirubin
UGT1A1
UDP-glucoronosyltransferase 1A1

Introduction

Neonatal hyperbilirubinemia (jaundice) occurs in about 60% of all term infants [1]. In general, most of the increase in bilirubin levels is physiological and usually will improve by the end of the first week of life. An increase in total serum bilirubin level above 5 mg/dL can cause jaundice that is visible to observers [2]. This condition can be severe if the infant was born prematurely, where the incidence increases to 80% [2]. In these infants, hyperbilirubinemia can develop into severe neonatal jaundice (SNNJ) that can cause bilirubin-induced neurological damage (BIND) and ultimately lead to irreversible neurodevelopmental impairment or even death [3].
Aside from prematurity, genetic polymorphisms have been reported as an important factor in the development of SNNJ [46]. The most commonly-studied gene is the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) that is closely associated with Crigler-Najjar’s syndrome type I and II, and Gilberts syndrome. These conditions are frequently found in Caucasian and African populations [7, 8]. Another gene that has been gaining attention, one that is mostly found in Asian populations, is OATP2/SLCO1B1 [9]. SLCO1B1 (solute carrier organic anion transporter family member 1b1) encodes the solute carrier organic anion transporter family member 1B1 and is responsible for the absorption of unconjugated and conjugated bilirubin in the liver. This gene, located in chromosome 12p12, consists of 15 exons (1 coding exon and 1 non-coding exon) and 14 introns, as well as containing 2073 nucleotides in the gene-coding area [9]. Neonates carrying this gene polymorphism are associated with reduced bilirubin clearance which induces hyperbilirubinemia [10]. Rotor Syndrome, an autosomal recessive disease associated with elevated conjugated bilirubin, is an example of a disease frequently associated with mutations in SLCO1B1 and SLCO1B3. The most frequent polymorphisms found in this gene are SLCO1B1*1B (388A > G), SLCO1B1*4 (463C > A), and SLCO1B1*5 (521 T > C), and the most commonly associated with hyperbilirubinemia is the SLCO1B1*1B (388A > G) [9, 1113].
Based on previous studies, there is an association between SLCO1B1 with racial variety in regard to the severity of unconjugated neonatal hyperbilirubinemia. In Indian, Japanese, Taiwanese, and Chinese neonates, the incidence of polymorphism is higher in infants with higher bilirubin levels compared to those with lower bilirubin levels [9, 11, 12, 14]. However, in studies of Malaysian and Thai hyperbilirubinemic neonates, this SNP was not frequently found [15, 16]. In Indonesia, polymorphism studies on neonatal hyperbilirubinemia has yet to be conducted extensively. Therefore, this study provides valuable insight into the association of genetic factors especially SLCO1B1 with the severity of neonatal hyperbilirubinemia in Indonesia.

Materials and methods

Study population

We conducted a cross-sectional study of patients in neonatal intensive care unit across three hospitals; M. Yunus General Hospital Bengkulu, Biak General Hospital Papua, and Cipto Mangunkusumo Hospital (CMH) Jakarta based on total population sampling. We collected samples and clinical data from November 2016 to September 2017, followed by molecular analysis in 2018. Diagnosis of hyperbilirubinemia was based on clinical jaundice as observed by a neonatologist based on Kramer’s index between day 3–7 since birth before receiving phototherapy, and peak Total Serum Bilirubin (TSB) was then taken prior to initiation of phototherapy. Mild hyperbilirubinemia was considered below 13 mg/dL, and moderate to severe was considered above and equal to 13 mg/dL [17]. Both inborn and outborn, preterm and term infants were included. Patients with hemolytic anemia, cephalhematoma, neonatal sepsis, ABO and rhesus incompatibility, maternal diabetes, and other comorbidities that would affect the total serum bilirubin level were excluded through clinical and laboratory approach. Clinical and sociodemographic variables were obtained through interviews and medical records. Informed consent was obtained from both parents prior to inclusion into the study. This study was approved by the Ethics Committee of Faculty of Medicine, Universitas Indonesia.

Laboratory investigations

Capillary blood samples were obtained from 88 infants and were used to measure their peak total serum bilirubin using the ADVIA Chemistry Total Bilirubin 2 device (diazo method) [18] taken between day 3 and 7 before phototherapy. A blood sample was also collected from each infant and stored at − 20 °C prior to DNA extraction.

DNA extraction and manipulation

DNA extraction from blood samples was conducted using the QIAmp Blood DNA Mini Kit (QIAGEN, Germany) according to the manufacturer’s protocol as previously described by Amandito et al. [19]. Following extraction, the DNA concentration was then measured by Nano-spectrophotometer (Thermo-Scientific) for use in polymerase chain reaction - restriction fragment length polymorphism (PCR-RFLP) analysis for determining the SNP in the exon 5 of SLCO1B1.
As much as 2uL of DNA was amplified using KOD FX Neo Kit (Toyobo) with each 25uL mix containing 60 ng extracted DNA, 12.5 uL KOD FX Neo Buffer, 5uL dNTPs (2 mM each of dATP, dCTP, dGTP, dTTP), 0.75uL of each primer (final concentration 8 uM), and 0.7 uL KOD FX Neo DNA polymerase (Novagen, Germany). Amplification was carried out with the following parameters: initial denaturation for 2 min at 95 °C, followed by 35 cycles of 1 min denaturation at 94 °C, annealing for 30 s at 57 °C, extension for 30 s at 68 °C, and final polymerisation for 5 min at 68 °C, and hold at 8 °C for 60 min (Biometra, Germany). All PCR products were visualized by agarose gel electrophoresis in 2% (wt/vol) agarose gels with TBE buffer.
To detect the 388A > G SNP (rs2306283), as according to a previous study, we used the primers 388F 5′-ATA ATG GTG CAA ATA AAG GGG-3′ (IDTDNA) for Forward Primer and 388R 5′-ACT ATC TCA GGT GAT GCT CTA-3′ (IDTDNA) for Reverse Primer [15]. For digestion, we used the restriction endonuclease TaqI (NEB) and CutSmart buffer (NEB). The digestion procedure consisted of incubation at 65 °C, followed by enzyme inactivation in 80 °C in a Thermal Cycler PCR MJ Mini (Bio-Rad). The sequenced products that were digested were analyzed by electrophoresis on 3% agarose gels with TBE buffer (Fig. 1).

Statistical analysis

We used SPSS version 24 for data management and statistical analysis. We conducted descriptive analysis for our study prior to inclusion for analytical study. We conducted bivariate analysis between independent variables and dependent variables using parametric and non-parametric methods, as appropriate. Variables with p value < 0.25 were included in the multivariate analysis using multiple logistic regression. We used two-sided p values in our analysis. Gender, population, exclusive breastfeeding, delivery method, delivery location, sibling requiring phototherapy, and SNPs were categorical data, while gestational age, birth weight, and mother’s age were numerical data.

Results

Clinical characteristics

We enrolled 88 hyperbilirubinemic neonates from three different hospitals; Bengkulu (a west coast city on Sumatra Island), Jakarta (capital of Indonesia located on the island of Java), and Biak (a far-eastern remote town on Papua Island), consisting of 47 in the control group (mild hyperbilirubinemia), and 41 in the case group (moderate-severe hyperbilirubinemia). Clinical data of all the patient groups based on the severity of hyperbilirubinemia are shown in Table 1. The mean bilirubin level of all neonates was 13.96 mg/dL with a SD of 6.57, mean gestational age of 33.23 weeks for control and 35.76 weeks for case group, median birth weight of 1580 g for the control group and 2500 g for the case group, and comparable gender between the two groups. Population, delivery location, gestational age, and birth weight were statistically-significant risk factors for moderate-severe hyperbilirubinemia. On the other hand, gender, exclusive breastfeeding, delivery method, sibling requiring phototherapy, and age of the mother were not significant risk factors, and therefore these factors were not included in the multivariate analysis.
Table 1
Demographic and clinical data of control vs. case neonates enrolled in the study
Factor in study
Control group
Case group
P
Gender (n = 88)
 Male
24 (51.1%)
17 (41.5%)
NS*
 MFemale
23 (48.9%)
24 (58.5%)
 
Population (n = 88)
 Jakarta
42 (89.4%)
14 (34.1%)
< 0.001*
 Bengkulu
2 (4.3%)
24 (58.5%)
 
 Papua
3 (6.4%)
3 (7.3%)
 
Exclusive Breastfeeding (n = 88)
 Yes
27 (57.4%)
17 (41.5%)
NS*
 No
20 (42.6%)
24 (58.5%)
 
Delivery Method (n = 88)
 Vaginal
14 (29.8%)
18 (43.9%)
NS*
 Caesarean
33 (70.2%)
23 (56.1%)
 
Delivery Location (n = 88)
 Midwife
2 (4.3%)
12 (29.3%)
0.001*
 Hospital
45 (95.7%)
29 (70.7%)
 
Sibling Requiring Phototherapy (n = 88)
 Yes
3 (6.4%)
3 (7.3%)
NS*
 No
44 (93.6%)
38 (92.7%)
 
Gestational age (n = 88)
 (Mean + SD, wk)
33.23 + 3.3
35.76 + 3.2
0.001**
Birth weight (n = 88)
 (Median (Min-Max), g)
1580 (700–3660)
2500 (940–4100)
< 0.001***
Mother’s age (n = 88)
 (Median (Min-Max), y)
29 (18–46)
28 (19–43)
NS***
*Chi-square
**Student’s t-test
***Mann-Whitney U-test
For the multiple logistic regression analysis (Table 2), the most significant factor was population with OR of 4.62 (95% CI 1.79–11.97) and p value of < 0.001, while delivery location, gestational age, and birth weight were no longer significant.
Table 2
Variables in the logistic regression analysis (n = 88)
Factor in study
OR (95% CI)
P
Population
4.62 (1.79–11.97)
< 0.001
Delivery location
4.26 (0.77–23.5)
NS
Gestational age
1 (0.79–1.31)
NS
Birth weight
1.01 (1–1.02)
NS

Detection of SLCO1B1*1B polymorphism

Table 3 shows the genotypic frequencies of SLCO1B1*1B variants due to the SNP in exon 5. In the control group, most of the SNPs were homozygote G/G with 34 neonates (72.3%), and wildtype A/A was the least common with only 2 neonates (4.3%). The same is in the case group with 26 neonates (63.4%) homozygote G/G and 3 neonates (7.3%) were wild-type A/A. No statistical significance was achieved when analyzing the two groups based on SLCO1B1 SNP variant. Table 4 shows the analysis between SNPs and bilirubin level which shows no statistical significance.
Table 3
Genotypic frequencies in SLCO1B1*1B in control and case groups (n = 88)
Factor in study
Control group
Case group
Odds Ratio (95% CI)
P
Wildtype A/A
2 (4.3%)
3 (7.3%)
Reference
NS*
Heterozygote A/G
11 (23.4%)
12 (29.3%)
0.727 (0.102–5.2)
 
Homozygote G/G
34 (72.3%)
26 (63.4%)
0.51 (0.079–3.27)
 
*Fisher’s exact test
Table 4
Correlation between bilirubin level and genetic polymorphism (n = 88)
SNP
TSB ((Median (Min-Max), mg/dL))
P
Wildtype A/A
14.98 (8.3–18.6)
NS*
Heterozygote A/G
13.2 (6.41–29)
 
Homozygote G/G
12.39 (5.55–26.7)
 
*Kruskal-Wallis
Table 5 highlights the distribution of SLCO1B1*1B between the three centers which represents three different ethnicities and environments, with CMH, Jakarta being the metropolitan capital city with heterogenous ethnicity, M. Yunus, Bengkulu an isolated west coast city with a homogenous ethnicity, and Biak, a remote east town on Papua, also with a homogenous ethnicity. In all three centers, homozygote variant of SLCO1B*1B was the most dominant (75, 57.7, and 50%, respectively), with the heterozygotic variant being the second-most dominant and the wild-type being the least dominant. There was no significant association between the population and the pattern of SLCO1B1*1B polymorphic variance. Table 6 shows the demographic data across the three different hospitals. Between them, only gender (p = 0.004) and ethnicity (p < 0.001) were significantly different.
Table 5
Genotypic distribution of SLCO1B1*1B across three centers (n = 88)
Center
Wildtype A/A
Heterozygote A/G
Homozygote G/G
P
CMH, Jakarta
2 (3.6%)
12 (21.4%)
42 (75%)
NS*
M. Yunus, Bengkulu
2 (7.7%)
9 (34.6%)
15 (57.7%)
 
Biak, Papua
1 (16.7%)
2 (33.3%)
3 (50%)
 
*Chi-square
Table 6
Demographic data across the three hospital populations
Factor in study
CMH, Jakarta
M. Yunus, Bengkulu
Biak, Papua
P
Gender (n = 88)
 Male
32 (57%)
5 (19%)
4 (67%)
0.004*
 Female
24 (43%)
21 (81%)
2 (33%)
 
 Exclusive Breastfeeding (n = 88)
  
NS*
 Yes
31 (55%)
10 (38%)
3 (50%)
 
 No
25 (45%)
16 (62%)
3 (50%)
 
 Delivery Location (n = 88)
  
NS*
 Midwife
52 (93%)
17 (65%)
5 (83%)
 
 Hospital
4 (7%)
9 (35%)
1 (17%)
 
 Ethnicity (n = 88)
< 0.001*
 Javanese
19 (34%)
0 (0%)
0 (0%)
 
 Betawi
15 (27%)
0 (0%)
0 (0%)
 
 Sundanese
13 (24%)
0 (0%)
0 (0%)
 
 Minangkabau
6 (10%)
0 (0%)
0 (0%)
 
 Indo-Chinese
3 (5%)
0 (0%)
0 (0%)
 
 Bengkulu
0 (0%)
26 (100%)
0 (0%)
 
 Papuan
0 (0%)
0 (0%)
6 (100%)
 
 Gestational age (n = 88)
  
NS**
  (Mean + SD, wk)
33.66 + 3.26
35.42 + 3.35
37 + 4.65
 
 Birth weight (n = 88)
  
NS**
 (Median (Min-Max), g)
1887 (910–4100)
2500 (1200–3700)
2890 (700–3500)
 
 Mother’s age (n = 88)
  
NS**
 (Median (Min-Max), y)
31.5 (18–43)
26 (19–38)
28 (21–46)
 
*Chi-square
**Kruskal-Wallis
We conducted additional analysis from both groups and within each group to find any association between SLCO1B1*1B and bilirubin level and found there was no significant association.

Discussion

Physiological conditions such as the short neonatal red blood cell’s lifespan, absorption of bilirubin by the liver and a non-optimal conjugation process of bilirubin can cause hyperbilirubinemia [1, 2]. Premature birth carries a higher risk of severe hyperbilirubinemia because of the immaturity of the red blood cells and an impaired hepatic cell function responsible for neonatal the bilirubin metabolism in neonates [2, 20]. The cause for this condition also varies between populations due to suspected contributions of genetic factors towards the pathogenesis of hyperbilirubinemia.
SLCO1B1 is a sinusoidal membrane protein expressed at the basolateral membrane of hepatocytes and is putatively involved in facilitating the hepatic uptake of unconjugated bilirubin [9, 13]. The exon 5 mutation of 388G > A is a nonsynonymous SNP with a missense mutation that causes a single amino acid substitution (asparagine to aspartic acid) encoded by codons 130 of SLCO1B1. This change disrupts the function of the SLCO1B1 transporter in transporting bilirubin from blood to the liver, leading to bilirubin concentration remaining high in the blood, which, in turn, causes hyperbilirubinemia [12].
Aside from the well-established risk factors of low gestational age and low birth weight, we found a significant association between the severity of hyperbilirubinemia and difference of populations across Jakarta, Bengkulu, and Papua, as well as with delivery location.
SLCO1B1*1B in both the heterozygous and homozygous variants was predominantly found (94.32%) in the neonates that we studied. Compared with the study of UGT1A1*60 and UGT1A1*6 from our previous study, SCLO1B1*1B was similar to *60 which showed a high incidence albeit with no significant association with the higher bilirubin level (severity) in neonatal hyperbilirubinemia [19]. Other studies on SCLO1B1 in different countries also showed similar findings to our study concerning the incidence of polymorphisms. In a study in India, there were 77.7% hyperbilirubinemic neonates having the variant *1B in hyperbilirubinemic neonates, and 87.7% in Malaysia. All these studies demonstrate that more than half of the studied population has the *1B variant [14, 16, 21]. Both Asian countries share a lot in common in terms of genetic makeup with Indonesia.
Regarding the association between SLCO1B1*1B and higher bilirubin level in infants, there are still inconclusive evidence from different studies of different populations. In a study by Huang et al. in 2004, they reported the *1B SNP to be more frequent among hyperbilirubinemic infants with an OR of 2.01. In the study by Liu et al., the Chinese variant of *1B was also found to be associated with hyperbilirubinemia in the Guangdong population, but not in the Yunnan population [9]. Whereas other studies in Fujian and Taiwan also failed to prove the association of *1B with neonatal hyperbilirubinemia, one study in Taiwan showed that Taiwanese neonates with the minor allele of *1B were at high risk to develop severe hyperbilirubinemia [2224].
Some ethnicities are shown to be more at risk for severe hyperbilirubinemia than others such as African-American and Asian [25]. This is most likely caused by the different prevalence of genetic polymorphisms responsible for metabolism or transport pathways [26]. Variations of specific genes can also differ between the individuals in a smaller population group. An example of this is the intra-ethnic differences of UGT1A1 polymorphism in the Chinese populations of Han, She, and Dong origin [27]. Global analysis of SLCO1B1 also suggests that SLCO1B1 diversity is greater within populations than between populations [28]. Conversely, we did not find any intra-ethnic differences in our three populations for SLCO1B1 polymorphism.
From the comparison of genotypic distribution and clinical factor among the three different centers comprising population of different sub-ethnic origins, we observed a significant association with the severity of hyperbilirubinemia independent from the history of exclusive breastfeeding and other established clinical risk factors. This suggests the presence of a potentially undiscovered important risk factor for the development severe hyperbilirubinemia in the Indonesian population. Further research effort in Indonesia is warranted to investigate other potential genetic factors in different pathways leading to hyperbilirubinemia, including BLVRA and HMOX1 [2932].
Our study is limited by the fact that we used a bank of DNA from a larger project, in which some subjects’ DNA samples were not sufficient for genetic analysis. Therefore, we were unable to study the whole population that we originally collected from the three centers. We also did not use consistently homogenous populations in the comparisons, and this could have obscured certain ethnicities where a different polymorphism variant could be dominant instead of *1B. The method of SNP analysis using PCR-RFLP is also less efficient and effective compared to other techniques such as DNA sequencing. However, we believe it is still sufficient in order to obtain national preliminary data that would have been otherwise severely lacking.

Conclusion

SLCO1B1*1B was found to be a common occurrence in Indonesian hyperbilirubinemic neonates. There was no statistically significant differences between occurrence of the SLCO1B1*1B variant and severity of neonatal hyperbilirubinemia. There was, however, a significant difference in the severity depending on the center where the neonate was enrolled. Future studies are required that includes a more diverse population to represent the entirety of the ethnically-diverse Indonesian population and exploring additional genetic risk factors is highly recommended.

Acknowledgements

We would like to thank the laboratory staff Bude Narti and NICU nursing staff of CMH, M. Yunus hospital, and Biak hospital for their contribution to this study. We would extend our utmost gratitude to Prof. Claudio Tiribelli, MD, PhD of Liver Research Center, Italian Liver Foundation for his scientific expertise. We would also give our appreciation to Alex Qinyang Liu, MBBS of Department of Surgery, Prince of Wales Hospital, Hong Kong and Nick Heng, PhD of Faculty of Dentistry, University of Otago for their invaluable English polishing assistance of our manuscript.
All authors hereby declare that all experiments have been examined and approved by the Ethics Committee of the Faculty of Medicine, Universitas Indonesia (No. 995/UN2.F1/ETIK/2016, protocol number 16–11-432) and have therefore been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. A written informed consent was obtained from the parents before inclusion into the study.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Porter ML, Dennis BL. Hyperbilirubinemia in the term newborn. Am Fam Physician. 2002;65(344):599–605.PubMed Porter ML, Dennis BL. Hyperbilirubinemia in the term newborn. Am Fam Physician. 2002;65(344):599–605.PubMed
2.
Zurück zum Zitat Lauer BJ, Spector ND. Hyperbilirubinemia in the newborn. Pediatr Rev. 2011;32(8):341–9.CrossRef Lauer BJ, Spector ND. Hyperbilirubinemia in the newborn. Pediatr Rev. 2011;32(8):341–9.CrossRef
3.
Zurück zum Zitat Watchko JF, Tiribelli C. Bilirubin-induced neurologic damage—mechanisms and management approaches. N Engl J Med. 2013;369(21):2021–30.CrossRef Watchko JF, Tiribelli C. Bilirubin-induced neurologic damage—mechanisms and management approaches. N Engl J Med. 2013;369(21):2021–30.CrossRef
4.
Zurück zum Zitat Rohsiswatmo R, Amandito R. Hiperbilirubinemia pada neonatus> 35 minggu di Indonesia; pemeriksaan dan tatalaksana terkini. Sari Pediatri. 2018;20(2):115–22.CrossRef Rohsiswatmo R, Amandito R. Hiperbilirubinemia pada neonatus> 35 minggu di Indonesia; pemeriksaan dan tatalaksana terkini. Sari Pediatri. 2018;20(2):115–22.CrossRef
5.
Zurück zum Zitat Maruo Y, Nishizawa K, Sato H, Doida Y, Shimada M. Association of neonatal hyperbilirubinemia with bilirubin UDP-glucuronosyltransferase polymorphism. Pediatrics. 1999;103(6):1224–7.CrossRef Maruo Y, Nishizawa K, Sato H, Doida Y, Shimada M. Association of neonatal hyperbilirubinemia with bilirubin UDP-glucuronosyltransferase polymorphism. Pediatrics. 1999;103(6):1224–7.CrossRef
6.
Zurück zum Zitat Travan L, Lega S, Crovella S, Montico M, Panontin E, Demarini S. Severe neonatal hyperbilirubinemia and UGT1A1 promoter polymorphism. J Pediatr. 2014;165(1):42–5.CrossRef Travan L, Lega S, Crovella S, Montico M, Panontin E, Demarini S. Severe neonatal hyperbilirubinemia and UGT1A1 promoter polymorphism. J Pediatr. 2014;165(1):42–5.CrossRef
7.
Zurück zum Zitat Kaplan M, Slusher T, Renbaum P, et al. n UDP-glucuronosyltransferase 1A1 promoter polymorphism in Nigerian neonates. Pediatr Res. 2008;63(1):109–11.CrossRef Kaplan M, Slusher T, Renbaum P, et al. n UDP-glucuronosyltransferase 1A1 promoter polymorphism in Nigerian neonates. Pediatr Res. 2008;63(1):109–11.CrossRef
8.
Zurück zum Zitat Beutler E, Gelbart T, Demina A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci. 1998;95(14):8170–4.CrossRef Beutler E, Gelbart T, Demina A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci. 1998;95(14):8170–4.CrossRef
9.
Zurück zum Zitat Liu J, Long J, Zhang S, Fang X, Luo Y. The impact of SLCO1B1 genetic polymorphisms on neonatal hyperbilirubinemia: a systematic review with meta-analysis. J Pediatr. 2013;89(5):434–43.CrossRef Liu J, Long J, Zhang S, Fang X, Luo Y. The impact of SLCO1B1 genetic polymorphisms on neonatal hyperbilirubinemia: a systematic review with meta-analysis. J Pediatr. 2013;89(5):434–43.CrossRef
10.
Zurück zum Zitat Johnson AD, Kavousi M, Smith AV, et al. Genome-wide association meta-analysis for total serum bilirubin levels. Hum Mol Genet. 2009;18(14):2700–10.CrossRef Johnson AD, Kavousi M, Smith AV, et al. Genome-wide association meta-analysis for total serum bilirubin levels. Hum Mol Genet. 2009;18(14):2700–10.CrossRef
11.
Zurück zum Zitat Kim S-R, Saito Y, Sai K, et al. Genetic variations and frequencies of major haplotypes in SLCO1B1 encoding the transporter OATP1B1 in Japanese subjects: SLCO1B1* 17 is more prevalent than* 15. Drug Metab Pharmacokinet. 2007;22(6):456–61.CrossRef Kim S-R, Saito Y, Sai K, et al. Genetic variations and frequencies of major haplotypes in SLCO1B1 encoding the transporter OATP1B1 in Japanese subjects: SLCO1B1* 17 is more prevalent than* 15. Drug Metab Pharmacokinet. 2007;22(6):456–61.CrossRef
12.
Zurück zum Zitat Liu J, Long J, Zhang S, Fang X, Luo Y. Polymorphic variants of SLCO1B1 in neonatal hyperbilirubinemia in China. Ital J Pediatr. 2013;39(1):49.CrossRef Liu J, Long J, Zhang S, Fang X, Luo Y. Polymorphic variants of SLCO1B1 in neonatal hyperbilirubinemia in China. Ital J Pediatr. 2013;39(1):49.CrossRef
13.
Zurück zum Zitat Lu A, Zhong D, Xie X. Relationship between SLCO1B1 polymorphism and neonatal hyperbilirubinemia. Guangxi Med J. 2015;37:592–6. Lu A, Zhong D, Xie X. Relationship between SLCO1B1 polymorphism and neonatal hyperbilirubinemia. Guangxi Med J. 2015;37:592–6.
14.
Zurück zum Zitat D'Silva S, Colah RB, Ghosh K, Mukherjee MB. Combined effects of the UGT1A1 and OATP2 gene polymorphisms as major risk factor for unconjugated hyperbilirubinemia in Indian neonates. Gene. 2014;547(1):18–22.CrossRef D'Silva S, Colah RB, Ghosh K, Mukherjee MB. Combined effects of the UGT1A1 and OATP2 gene polymorphisms as major risk factor for unconjugated hyperbilirubinemia in Indian neonates. Gene. 2014;547(1):18–22.CrossRef
15.
Zurück zum Zitat Prachukthum S, Nunnarumit P, Pienvichit P, et al. Genetic polymorphisms in Thai neonates with hyperbilirubinemia. Acta Paediatr. 2009;98(7):1106–10.CrossRef Prachukthum S, Nunnarumit P, Pienvichit P, et al. Genetic polymorphisms in Thai neonates with hyperbilirubinemia. Acta Paediatr. 2009;98(7):1106–10.CrossRef
16.
Zurück zum Zitat Wong F, Boo N, Othman A. Risk factors associated with unconjugated neonatal hyperbilirubinemia in Malaysian neonates. J Trop Pediatr. 2013;59(4):280–5.CrossRef Wong F, Boo N, Othman A. Risk factors associated with unconjugated neonatal hyperbilirubinemia in Malaysian neonates. J Trop Pediatr. 2013;59(4):280–5.CrossRef
17.
Zurück zum Zitat Paludetto R, Mansi G, Raimondi F, et al. Moderate hyperbilirubinemia induces a transient alteration of neonatal behavior. Pediatrics. 2002;110(4):e50.CrossRef Paludetto R, Mansi G, Raimondi F, et al. Moderate hyperbilirubinemia induces a transient alteration of neonatal behavior. Pediatrics. 2002;110(4):e50.CrossRef
18.
Zurück zum Zitat Farrell C-JL, Carter AC. Serum indices: managing assay interference. Ann Clin Biochem. 2016;53(5):527–38.CrossRef Farrell C-JL, Carter AC. Serum indices: managing assay interference. Ann Clin Biochem. 2016;53(5):527–38.CrossRef
19.
Zurück zum Zitat Amandito R, Putradista R, Jikesya C, et al. UGT1A1 gene and neonatal hyperbilirubinemia: a preliminary study from Bengkulu, Indonesia. BMC Res Notes. 2018;11(1):172.CrossRef Amandito R, Putradista R, Jikesya C, et al. UGT1A1 gene and neonatal hyperbilirubinemia: a preliminary study from Bengkulu, Indonesia. BMC Res Notes. 2018;11(1):172.CrossRef
20.
Zurück zum Zitat Mishra S, Agarwal R, Deorari AK, Paul VK. Jaundice in the newborns. Indian J Pediatr. 2008;75(2):157.CrossRef Mishra S, Agarwal R, Deorari AK, Paul VK. Jaundice in the newborns. Indian J Pediatr. 2008;75(2):157.CrossRef
21.
Zurück zum Zitat Kohlrausch FB, de Cássia Estrela R, Barroso PF, Suarez-Kurtz G. The impact of SLCO1B1 polymorphisms on the plasma concentration of lopinavir and ritonavir in HIV-infected men. Br J Clin Pharmacol. 2010;69(1):95–8.CrossRef Kohlrausch FB, de Cássia Estrela R, Barroso PF, Suarez-Kurtz G. The impact of SLCO1B1 polymorphisms on the plasma concentration of lopinavir and ritonavir in HIV-infected men. Br J Clin Pharmacol. 2010;69(1):95–8.CrossRef
22.
Zurück zum Zitat Weng Y-H, Chiu Y-W, Cheng S-W, Yang C-Y. Risk assessment of gene variants for neonatal hyperbilirubinemia in Taiwan. BMC Pediatr. 2016;16(1):144.CrossRef Weng Y-H, Chiu Y-W, Cheng S-W, Yang C-Y. Risk assessment of gene variants for neonatal hyperbilirubinemia in Taiwan. BMC Pediatr. 2016;16(1):144.CrossRef
23.
Zurück zum Zitat Zhou J, Yang C, Zhu W, et al. Identification of genetic risk factors for neonatal hyperbilirubinemia in Fujian Province, southeastern China: a case-control study. Biomed Res Int. 2018;2018. Zhou J, Yang C, Zhu W, et al. Identification of genetic risk factors for neonatal hyperbilirubinemia in Fujian Province, southeastern China: a case-control study. Biomed Res Int. 2018;2018.
24.
Zurück zum Zitat Lin R, Wang X, Wang Y, et al. Association of polymorphisms in four bilirubin metabolism genes with serum bilirubin in three Asian populations. Hum Mutat. 2009;30(4):609–15.CrossRef Lin R, Wang X, Wang Y, et al. Association of polymorphisms in four bilirubin metabolism genes with serum bilirubin in three Asian populations. Hum Mutat. 2009;30(4):609–15.CrossRef
25.
Zurück zum Zitat Setia S, Villaveces A, Dhillon P, Mueller BA. Neonatal jaundice in Asian, white, and mixed-race infants. Arch Pediatr Adolesc Med. 2002;156(3):276–9.CrossRef Setia S, Villaveces A, Dhillon P, Mueller BA. Neonatal jaundice in Asian, white, and mixed-race infants. Arch Pediatr Adolesc Med. 2002;156(3):276–9.CrossRef
26.
Zurück zum Zitat Ramamoorthy A, Pacanowski M, Bull J, Zhang L. Racial/ethnic differences in drug disposition and response: review of recently approved drugs. Clin Pharmacol Ther. 2015;97(3):263–73.CrossRef Ramamoorthy A, Pacanowski M, Bull J, Zhang L. Racial/ethnic differences in drug disposition and response: review of recently approved drugs. Clin Pharmacol Ther. 2015;97(3):263–73.CrossRef
27.
Zurück zum Zitat Zhang A, Xing Q, Qin S, et al. Intra-ethnic differences in genetic variants of the UGT-glucuronosyltransferase 1A1 gene in Chinese populations. Pharmacogenomics J. 2007;7(5):333.CrossRef Zhang A, Xing Q, Qin S, et al. Intra-ethnic differences in genetic variants of the UGT-glucuronosyltransferase 1A1 gene in Chinese populations. Pharmacogenomics J. 2007;7(5):333.CrossRef
28.
Zurück zum Zitat Pasanen MK, Neuvonen PJ, Niemi M. Global analysis of genetic variation in SLCO1B1; 2008.CrossRef Pasanen MK, Neuvonen PJ, Niemi M. Global analysis of genetic variation in SLCO1B1; 2008.CrossRef
29.
Zurück zum Zitat Huang CS, Huang MJ, Chen ES, Chang PF, Chen WC. Glucose-6-phosphate dehydrogenase deficiency, the UDP-glucuronosyl transferase 1A1 gene, and neonatal hyperbilirubinemia. Gastroenterology. 2002;123(1):127–33.CrossRef Huang CS, Huang MJ, Chen ES, Chang PF, Chen WC. Glucose-6-phosphate dehydrogenase deficiency, the UDP-glucuronosyl transferase 1A1 gene, and neonatal hyperbilirubinemia. Gastroenterology. 2002;123(1):127–33.CrossRef
30.
Zurück zum Zitat Tiwari PK, Sethi A, Basu S, Raman R, Kumar A. Heme oxygenase-1 gene variants and hyperbilirubinemia risk in north Indian newborns. Eur J Pediatr. 2013;172(12):1627–32.CrossRef Tiwari PK, Sethi A, Basu S, Raman R, Kumar A. Heme oxygenase-1 gene variants and hyperbilirubinemia risk in north Indian newborns. Eur J Pediatr. 2013;172(12):1627–32.CrossRef
31.
Zurück zum Zitat Chou H-C, Chen M-H, Yang H-I, et al. 211 G to a variation of UDP-glucuronosyl transferase 1A1 gene and neonatal breastfeeding jaundice. Pediatr Res. 2011;69(2):170–4.CrossRef Chou H-C, Chen M-H, Yang H-I, et al. 211 G to a variation of UDP-glucuronosyl transferase 1A1 gene and neonatal breastfeeding jaundice. Pediatr Res. 2011;69(2):170–4.CrossRef
32.
Zurück zum Zitat Li Y, Wu T, Chen L, Zhu Y. Associations between G6PD, OATP1B1 and BLVRA variants and susceptibility to neonatal hyperbilirubinaemia in a Chinese Han population. J Paediatr Child Health. 2019;55(1):1–7. Li Y, Wu T, Chen L, Zhu Y. Associations between G6PD, OATP1B1 and BLVRA variants and susceptibility to neonatal hyperbilirubinaemia in a Chinese Han population. J Paediatr Child Health. 2019;55(1):1–7.
Metadaten
Titel
SLCO1B1 c.388A > G variant incidence and the severity of hyperbilirubinemia in Indonesian neonates
verfasst von
Radhian Amandito
Rinawati Rohsiswatmo
Michelle Halim
Vanessa Tirtatjahja
Amarila Malik
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Pediatrics / Ausgabe 1/2019
Elektronische ISSN: 1471-2431
DOI
https://doi.org/10.1186/s12887-019-1589-1

Weitere Artikel der Ausgabe 1/2019

BMC Pediatrics 1/2019 Zur Ausgabe

Endlich: Zi zeigt, mit welchen PVS Praxen zufrieden sind

IT für Ärzte Nachrichten

Darauf haben viele Praxen gewartet: Das Zi hat eine Liste von Praxisverwaltungssystemen veröffentlicht, die von Nutzern positiv bewertet werden. Eine gute Grundlage für wechselwillige Ärzte und Psychotherapeuten.

Durch übermäßige Internetnutzung wird oft die Schule verpasst

Häufige Fehlzeiten in der Schule können durch physische und psychische Probleme verursacht werden. Wie in einer Studie aus Finnland nun belegt wird, führt auch die exzessive Nutzung des Internets gehäuft zu Abwesenheiten.

Kinder mit anhaltender Sinusitis profitieren häufig von Antibiotika

30.04.2024 Rhinitis und Sinusitis Nachrichten

Persistieren Sinusitisbeschwerden bei Kindern länger als zehn Tage, ist eine Antibiotikatherapie häufig gut wirksam: Ein Therapieversagen ist damit zu über 40% seltener zu beobachten als unter Placebo.

Neuer Typ-1-Diabetes bei Kindern am Wochenende eher übersehen

23.04.2024 Typ-1-Diabetes Nachrichten

Wenn Kinder an Werktagen zum Arzt gehen, werden neu auftretender Typ-1-Diabetes und diabetische Ketoazidosen häufiger erkannt als bei Arztbesuchen an Wochenenden oder Feiertagen.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.