1
|
Stevenson DK, Wells GS, Wong RJ. Is it time for a precision health approach to the management of newborn hyperbilirubinemia? J Perinatol 2024; 44:920-923. [PMID: 38514741 DOI: 10.1038/s41372-024-01941-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Newborn hyperbilirubinemia during the first two weeks of life is one of most common problems requiring management decisions by a pediatrician. However, high bilirubin levels in the circulation have been associated with neurologic injury under a variety of conditions encountered in the newborn infant, such as hemolysis. The risk for developing dangerous hyperbilirubinemia is multifactorial and is determined by a complex set of factors related to a newborn infant's genetic capacities as well as intra- and extrauterine exposures. To this end, a precision health approach based on the integration of prenatal genetic and postnatal diagnostic measures might improve the management of neonatal hyperbilirubinemia.
Collapse
Affiliation(s)
- David K Stevenson
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Grant S Wells
- Stanford Maternal & Child Health Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Division of Neonatal & Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
2
|
Fan J, He HY, Li HH, Wu PL, Tang L, Deng BY, Dong WH, Wang JH. Associations between UGT1A1, SLCO1B1, SLCO1B3, BLVRA and HMOX1 polymorphisms and susceptibility to neonatal severe hyperbilirubinemia in Chinese Han population. BMC Pediatr 2024; 24:82. [PMID: 38279097 PMCID: PMC10811893 DOI: 10.1186/s12887-024-04537-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 01/04/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Severe neonatal hyperbilirubinemia could lead to kernicterus and neonatal death. This study aimed to analyze the association between single nucleotide polymorphisms in genes involved in bilirubin metabolism and the incidence of severe hyperbilirubinemia. METHODS A total of 144 neonates with severe hyperbilirubinemia and 50 neonates without or mild hyperbilirubinemia were enrolled in 3 institutions between 2019 and 2020. Twelve polymorphisms of 5 genes (UGT1A1, SLCO1B1, SLCO1B3, BLVRA, and HMOX1) were analyzed by PCR amplification of genomic DNA. Genotyping was performed using an improved multiplex ligation detection reaction technique based on ligase detection reaction. RESULTS The frequencies of the A allele in UGT1A1-rs4148323 and the C allele in SLCO1B3-rs2417940 in the severe hyperbilirubinemia group (30.2% and 90.6%, respectively) were significantly higher than those in the controls (30.2% vs.13.0%, 90.6% vs. 78.0%, respectively, both p < 0.05). Haplotype analysis showed the ACG haplotype of UGT1A1 were associated with an increased hyperbilirubinemia risk (OR 3.122, p = 0.001), whereas the GCG haplotype was related to a reduced risk (OR 0.523, p = 0.018). CONCLUSION The frequencies of the A allele in rs4148323 and the C allele in rs2417940 are highly associated with the incidence of severe hyperbilirubinemia in Chinese Han neonates. TRIAL REGISTRATION Trial registration number:ChiCTR1800020424; Date of registration:2018-12-29.
Collapse
Affiliation(s)
- Juan Fan
- Department of Neonatology , Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Hua-Yun He
- Department of Neonatology , Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Huan-Huan Li
- Department of Neonatology, Chongqing Jiulongpo People's Hospital, Chongqing, China
| | - Pi-Liu Wu
- Department of Neonatology, Bishan Maternity & Child Hospital of Chongqing, Chongqing, China
| | - Lei Tang
- Department of Pediatrics, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, China
| | - Bo-Yin Deng
- Department of Neonatology , Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Wen-Hui Dong
- Department of Neonatology , Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
| | - Jian-Hui Wang
- Department of Neonatology , Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| |
Collapse
|
3
|
Zhang W, Zhang Q, Cao Z, Zheng L, Hu W. Physiologically Based Pharmacokinetic Modeling in Neonates: Current Status and Future Perspectives. Pharmaceutics 2023; 15:2765. [PMID: 38140105 PMCID: PMC10747965 DOI: 10.3390/pharmaceutics15122765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Rational drug use in special populations is a clinical problem that doctors and pharma-cists must consider seriously. Neonates are the most physiologically immature and vulnerable to drug dosing. There is a pronounced difference in the anatomical and physiological profiles be-tween neonates and older people, affecting the absorption, distribution, metabolism, and excretion of drugs in vivo, ultimately leading to changes in drug concentration. Thus, dose adjustments in neonates are necessary to achieve adequate therapeutic concentrations and avoid drug toxicity. Over the past few decades, modeling and simulation techniques, especially physiologically based pharmacokinetic (PBPK) modeling, have been increasingly used in pediatric drug development and clinical therapy. This rigorously designed and verified model can effectively compensate for the deficiencies of clinical trials in neonates, provide a valuable reference for clinical research design, and even replace some clinical trials to predict drug plasma concentrations in newborns. This review introduces previous findings regarding age-dependent physiological changes and pathological factors affecting neonatal pharmacokinetics, along with their research means. The application of PBPK modeling in neonatal pharmacokinetic studies of various medications is also reviewed. Based on this, we propose future perspectives on neonatal PBPK modeling and hope for its broader application.
Collapse
Affiliation(s)
| | | | | | - Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| |
Collapse
|
4
|
Riskin A, Bravdo Y, Habib C, Maor I, Mousa J, Shahbarat S, Shahak E, Shalata A. The Genetics of Glucose-6-Phosphate-Dehydrogenase (G6PD) and Uridine Diphosphate Glucuronosyl Transferase 1A1 (UGT1A1) Promoter Gene Polymorphism in Relation to Quantitative Biochemical G6PD Activity Measurement and Neonatal Hyperbilirubinemia. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1172. [PMID: 37508669 PMCID: PMC10378156 DOI: 10.3390/children10071172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency and polymorphism in uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) were associated with significant neonatal hyperbilirubinemia (NHB) and increased risk for kernicterus. However, quantitative screening tests for G6PD enzyme activity proved unsatisfactory in estimating the risk for significant NHB, especially in heterozygous females that could present phenotype overlap between normal homozygotes, heterozygotes, and deficient homozygotes, resulting in a continuum of intermediate G6PD activity. OBJECTIVE To examine the association of genotype and phenotype in newborns with decreased G6PD activity and its relation to NHB. STUDY DESIGN Quantitative G6PD enzyme activities were measured on umbilical cord blood samples. After accepting parental consent, samples were analyzed for G6PD mutations and UGT1A1 gene polymorphisms (number of TA repeats in the UGT1A1 promoter). The associations to quantitative G6PD activity and bilirubin levels were assessed. RESULTS 28 females and 27 males were studied. The Mediterranean mutation (NM_001360016.2(G6PD): c.563C>T (p.Ser188Phe)) was responsible for most cases of G6PD deficiency (20 hemizygous males, 3 homozygous and 16 heterozygous females). The association between this mutation, decreased G6PD activity and higher bilirubin levels was confirmed. Heterozygosity to 6/7 TA repeats in the UGT1A1 promoter was associated with increased NHB, especially in female newborns with G6PD deficiency. However, it seems that the interaction between G6PD deficiency, UGT1A1 promoter polymorphism, and NHB is more complex, possibly involving other genetic interactions, not yet described. Despite genotyping females with G6PD deficiency, the overlap between the upper range of borderline and the lower range of normal G6PD activity could not be resolved. CONCLUSIONS The results of this study highlight the possibility for future implementation of molecular genetic screening to identify infants at risk for significant NHB, especially UGT1A1 polymorphism in heterozygous females with borderline G6PD deficiency. However, further studies are needed before such screening could be applicable to daily practice.
Collapse
Affiliation(s)
- Arieh Riskin
- Department of Neonatology, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Yulia Bravdo
- Department of Pediatrics, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Clair Habib
- Department of Pediatrics, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Irit Maor
- Biochemistry Laboratory, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Julnar Mousa
- The Simon Winter Institute for Human Genetics, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Sizett Shahbarat
- The Simon Winter Institute for Human Genetics, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Elena Shahak
- The Simon Winter Institute for Human Genetics, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Adel Shalata
- The Simon Winter Institute for Human Genetics, Bnai Zion Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
5
|
Golden WC. Race and the 2022 American Academy of Pediatrics Hyperbilirubinemia Guideline. JAMA Pediatr 2023; 177:436-437. [PMID: 36780145 DOI: 10.1001/jamapediatrics.2022.5916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Affiliation(s)
- William Christopher Golden
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
6
|
Yang H, Li H, Xia Q, Dai W, Li X, Liu Y, Nie J, Yang F, Sun Y, Feng L, Yang L. UGT1A1 variants in Chinese Uighur and Han newborns and its correlation with neonatal hyperbilirubinemia. PLoS One 2022; 17:e0279059. [PMID: 36520959 PMCID: PMC9754166 DOI: 10.1371/journal.pone.0279059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/17/2022] [Indexed: 12/23/2022] Open
Abstract
To explore the correlation between UGT1A1 variant and neonatal hyperbilirubinemia in Chinese Uighur and Han populations. We conducted this study in Urumqi, China. Umbilical cord blood specimens and clinical information of term infants born in the studied center were collected. Variation status of UGT1A1 was determined by direct sequencing or capillary electrophoresis analysis. 102 Uighur and 99 Han normal term neonates, together with 19 hospitalized term newborns (10 Uighur and 9 Han) due to significant hyperbilirubinemia were enrolled into the final analysis. The incidence of neonates with high-risk transcutaneous bilirubin level (TCB) were much higher in Han newborns than in Uighur newborns(P = 0.01). Also, there was statistically significant difference in (TA) 7 promoter mutation of UGT1A1 between Han and Uighur group(χ2 = 4.675, P = 0.03). Furthermore, exon mutation (c.211 and /or c.1091) in UGT1A1 gene was significantly associated with increased TCB level (ORadj = 1.41, 95%CI: 0.25-2.51, P = 0.002) and higher risk of hyperbilirubinemia in both Han and Uighur infants after adjusted for covariates (ORadj = 2.21, 95%CI: 1.09-4.49, P = 0.03). In conclusion, UGT1A1 promoter polymorphism seem to be an important genetic modulator of plasma bilirubin level and neonatal hyperbilirubinemia risk within ethnic groups. Genetic assessment of UGT1A1 coding variants may be useful for clinical diagnosis of neonatal jaundice.
Collapse
Affiliation(s)
- Hui Yang
- Department of Laboratory Medicine, School of Medicine, Yangtze University, Jingzhou, China
- * E-mail:
| | - Huijun Li
- Department of Gynecology and Obstetrics, Maternity and Child Health Care Hospital, Urumqi Municipality Xinjiang Uighur Autonomous Region, Xinjiang Province, China
| | - Qingyao Xia
- Department of Laboratory Medicine, Western China Women and Child’s Hospital, Sichuan University, Chengdu, China
| | - Wencheng Dai
- Department of Gynecology and Obstetrics, Maternity and Child Health Care Hospital, Urumqi Municipality Xinjiang Uighur Autonomous Region, Xinjiang Province, China
| | - Xin Li
- Department of Laboratory Medicine, School of Medicine, Yangtze University, Jingzhou, China
| | - Yan Liu
- Department of Laboratory Medicine, School of Medicine, Yangtze University, Jingzhou, China
| | - Jie Nie
- Department of Laboratory Medicine, School of Medicine, Yangtze University, Jingzhou, China
| | - Fei Yang
- Department of Laboratory Medicine, School of Medicine, Yangtze University, Jingzhou, China
| | - Yunfeng Sun
- Department of Rehabilitation, Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Urumqi Municipality Xinjiang Uighur Autonomous Region, Xinjiang Province, China
| | - Lei Feng
- Department of Laboratory Medicine, People’s Hospital of Yuxi City, Yuxi, P. R. China
| | - Liye Yang
- Lab for Respiratory Disease, People’s Hospital of Yangjiang, Yangjiang, P. R. China
| |
Collapse
|
7
|
Salinas-Marín R, Murakami Y, González-Domínguez CA, Cruz-Muñoz ME, Mora-Montes HM, Morava E, Kinoshita T, Monroy-Santoyo S, Martínez-Duncker I. Case report: Functional characterization of a de novo c.145G>A p.Val49Met pathogenic variant in a case of PIGA-CDG with megacolon. Front Genet 2022; 13:971473. [PMID: 36324500 PMCID: PMC9619068 DOI: 10.3389/fgene.2022.971473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/23/2022] [Indexed: 12/02/2022] Open
Abstract
A subgroup of congenital disorders of glycosylation (CDGs) includes inherited GPI-anchor deficiencies (IGDs) that affect the biosynthesis of glycosylphosphatidylinositol (GPI) anchors, including the first reaction catalyzed by the X-linked PIGA. Here, we show the first PIGA-CDG case reported in Mexico in a male child with a moderate-to-severe phenotype characterized by neurological and gastrointestinal symptoms, including megacolon. Exome sequencing identified the hemizygous variant PIGA c.145G>A (p.Val49Met), confirmed by Sanger sequencing and characterized as de novo. The pathogenicity of this variant was characterized by flow cytometry and complementation assays in PIGA knockout (KO) cells.
Collapse
Affiliation(s)
- Roberta Salinas-Marín
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | - Yoshiko Murakami
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Carlos Alberto González-Domínguez
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, México
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | | | - Héctor Manuel Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Guanajuato, México
| | - Eva Morava
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, United States
- Department of Medical Genetics, University of Pecs Medical School, Pecs, Hungary
- Frontiers in Congenital Disorders of Glycosylation Consortium, National Institute of Neurological Diseases and Stroke (NINDS), National Institute of Child Health and Human Development (NICHD) and the National Center for Advancing Translational Sciences (NCATS), and the Rare Disorders Clinical Research Network (RDCRN), Bethesda, MD, United States
| | - Taroh Kinoshita
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Susana Monroy-Santoyo
- Centro de Investigación Traslacional, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, México
- *Correspondence: Iván Martínez-Duncker, ; Susana Monroy-Santoyo,
| | - Iván Martínez-Duncker
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, México
- Frontiers in Congenital Disorders of Glycosylation Consortium, National Institute of Neurological Diseases and Stroke (NINDS), National Institute of Child Health and Human Development (NICHD) and the National Center for Advancing Translational Sciences (NCATS), and the Rare Disorders Clinical Research Network (RDCRN), Bethesda, MD, United States
- *Correspondence: Iván Martínez-Duncker, ; Susana Monroy-Santoyo,
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Hyperbilirubinemia is commonly seen in neonates. Though hyperbilirubinemia is typically asymptomatic, severe elevation of bilirubin levels can lead to acute bilirubin encephalopathy and progress to kernicterus spectrum disorder, a chronic condition characterized by hearing loss, extrapyramidal dysfunction, ophthalmoplegia, and enamel hypoplasia. Epidemiological data show that the implementation of universal pre-discharge bilirubin screening programs has reduced the rates of hyperbilirubinemia-associated complications. However, acute bilirubin encephalopathy and kernicterus spectrum disorder are still particularly common in low- and middle-income countries. RECENT FINDINGS The understanding of the genetic and biochemical processes that increase the susceptibility of defined anatomical areas of the central nervous system to the deleterious effects of bilirubin may facilitate the development of effective treatments for acute bilirubin encephalopathy and kernicterus spectrum disorder. Scoring systems are available for the diagnosis and severity grading of these conditions. The treatment of hyperbilirubinemia in newborns relies on the use of phototherapy and exchange transfusion. However, novel therapeutic options including deep brain stimulation, brain-computer interface, and stem cell transplantation may alleviate the heavy disease burden associated with kernicterus spectrum disorder. Despite improved screening and treatment options, the prevalence of acute bilirubin encephalopathy and kernicterus spectrum disorder remains elevated in low- and middle-income countries. The continued presence and associated long-term disability of these conditions warrant further research to improve their prevention and management.
Collapse
Affiliation(s)
- Shuo Qian
- Department of Neurology and Rehabilitation, University of Illinois at Chicago College of Medicine, 912 S Wood St, Chicago, IL, 60612, USA.
| | - Prateek Kumar
- Department of Neurology and Rehabilitation, University of Illinois at Chicago College of Medicine, 912 S Wood St, Chicago, IL, 60612, USA
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois at Chicago College of Medicine, 912 S Wood St, Chicago, IL, 60612, USA
| |
Collapse
|
9
|
Clinical and Genetic Etiologies of Neonatal Unconjugated Hyperbilirubinemia in the China Neonatal Genomes Project. J Pediatr 2022; 243:53-60.e9. [PMID: 34953813 DOI: 10.1016/j.jpeds.2021.12.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/09/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the clinical and genetic causes of neonatal unconjugated hyperbilirubinemia. STUDY DESIGN We included 1412 neonates diagnosed with unconjugated hyperbilirubinemia (total serum bilirubin >95 percentile for age), from the China Neonatal Genomes Project between August 2016 and September 2019, in the current study. Clinical data and targeted panel sequencing data on 2742 genes including known unconjugated hyperbilirubinemia genes were analyzed. RESULTS Among the 1412 neonates with unconjugated hyperbilirubinemia, 37% had severe unconjugated hyperbilirubinemia, with total serum bilirubin levels that met the recommendations for exchange transfusion. Known clinical causes were identified for 68% of patients. The most common clinical cause in the mild unconjugated hyperbilirubinemia group was infection (17%) and in the severe group was combined factors (21%, with infection combined with extravascular hemorrhage the most common). A genetic variant was observed in 55 participants (4%), including 45 patients with variants in genes associated with unconjugated hyperbilirubinemia and 10 patients with variants that were regarded as additional genetic findings. Among the 45 patients identified with unconjugated hyperbilirubinemia-related variants, the genes were mainly associated with enzyme deficiencies, metabolic/biochemical disorders, and red blood cell membrane defects. G6PD and UGT1A1 variants, were detected in 34 of the 45 patients (76%). CONCLUSIONS Known clinical causes, which varied with bilirubin levels, were identified in approximately two-thirds of the patients. Genetic findings were identified in 4% of the patients, including in patients with an identified clinical cause, with G6PD and UGT1A1 being the most common genes in which variants were detected.
Collapse
|
10
|
Deng H, Zhou Y, Wang L, Zhang C. Ensemble learning for the early prediction of neonatal jaundice with genetic features. BMC Med Inform Decis Mak 2021; 21:338. [PMID: 34852805 PMCID: PMC8638201 DOI: 10.1186/s12911-021-01701-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/25/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neonatal jaundice may cause severe neurological damage if poorly evaluated and diagnosed when high bilirubin occurs. The study explored how to effectively integrate high-dimensional genetic features into predicting neonatal jaundice. METHODS This study recruited 984 neonates from the Suzhou Municipal Central Hospital in China, and applied an ensemble learning approach to enhance the prediction of high-dimensional genetic features and clinical risk factors (CRF) for physiological neonatal jaundice of full-term newborns within 1-week after birth. Further, sigmoid recalibration was applied for validating the reliability of our methods. RESULTS The maximum accuracy of prediction reached 79.5% Area Under Curve (AUC) by CRF and could be marginally improved by 3.5% by including genetic variant (GV). Feature importance illustrated that 36 GVs contributed 55.5% in predicting neonatal jaundice in terms of gain from splits. Further analysis revealed that the main contribution of GV was to reduce the false-positive rate, i.e., to increase the specificity in the prediction. CONCLUSIONS Our study shed light on the theoretical and practical value of GV in the prediction of neonatal jaundice.
Collapse
Affiliation(s)
- Haowen Deng
- School of Management, Fudan University, Shanghai, China
| | - Youyou Zhou
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lin Wang
- Shanghai University of Finance and Economics, Shanghai, China
| | - Cheng Zhang
- School of Management, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Yang H, Lin F, Chen ZK, Zhang L, Xu JX, Wu YH, Gu JY, Ma YB, Li JD, Yang LY. UGT1A1 mutation association with increased bilirubin levels and severity of unconjugated hyperbilirubinemia in ABO incompatible newborns of China. BMC Pediatr 2021; 21:259. [PMID: 34074250 PMCID: PMC8167307 DOI: 10.1186/s12887-021-02726-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 05/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neonatal hyperbilirubinemia causing jaundice is common in East Asian population. Uridine diphosphate glucuronosyltransferase isoenzyme (UGT1A1) glucuronidates bilirubin and converts the toxic form of bilirubin to its nontoxic form. METHOD A retrospective study was conducted to review clinical information of ABO hemolysis neonates (ABO HDN) admitted to the Department of Neonatology, referred for neonatal hyperbilirubinemia, in a large general hospital of southern China from 2011 to 2017. Variation status of UGT1A1 was determined by direct sequencing or genotype assays. RESULT Sixty-nine ABO HDNs were included into the final analysis. UGT1A1 c.211 G > A mutation (UGT1A1*6, p.Arg71Gly, rs4148323) was significantly associated with the increased bilirubin level in ABO HDNs, after adjusted by age, sex and feeding method (P = 0.019 for TBIL, P = 0.02 for IBIL). Moreover, heterozygous and/or homozygous UGT1A1 mutations in the coding sequence region were significantly associated with the increased risk of developing hazardous hyperbilirubinemia (as defined by TSB > 427 umol/L) as compared those with a normal UGT1A1 genotype (ORadj = 9.16, 95%CI 1.99-42.08, P = 0.002) in the study cohort. CONCLUSION UGT1A1 variant in coding region is actively involved in the pathogenesis of ABO hemolysis related neonatal hyperbilirubinemia. Genetic assessment of UGT1A1 may be useful for clinical diagnosis of neonatal unconjugated hyperbilirubinemia.
Collapse
Affiliation(s)
- Hui Yang
- Department of Laboratory Medicine, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, People's Republic of China
| | - Fen Lin
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, People's Republic of China
| | - Zi-Kai Chen
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, Guangdong Province, People's Republic of China
| | - Lin Zhang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, People's Republic of China
| | - Jia-Xin Xu
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, People's Republic of China
| | - Yong-Hao Wu
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, People's Republic of China
| | - Jing-Ying Gu
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, People's Republic of China
| | - Yu-Bin Ma
- Department of Pediatrics, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, People's Republic of China
| | - Jian-Dong Li
- Department of Pediatrics, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, People's Republic of China
| | - Li-Ye Yang
- Lab for Respiratory Disease, People's Hospital of Yangjiang, No. 42 Dongshan Road, Yangjiang, 529500, Guangdong Province, People's Republic of China.
| |
Collapse
|
12
|
The Role of Heme Oxygenase-1 Promoter Polymorphisms in Perinatal Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073520. [PMID: 33805292 PMCID: PMC8037596 DOI: 10.3390/ijerph18073520] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 11/19/2022]
Abstract
Heme oxygenase (HO) is the rate-limiting enzyme in the heme catabolic pathway, which degrades heme into equimolar amounts of carbon monoxide, free iron, and biliverdin. Its inducible isoform, HO-1, has multiple protective functions, including immune modulation and pregnancy maintenance, showing dynamic alteration during perinatal periods. As its contribution to the development of perinatal complications is speculated, two functional polymorphisms of the HMOX1 gene, (GT)n repeat polymorphism (rs3074372) and A(-413)T single nucleotide polymorphism (SNP) (rs2071746), were studied for their association with perinatal diseases. We systematically reviewed published evidence on HMOX1 polymorphisms in perinatal diseases and clarified their possible significant contribution to neonatal jaundice development, presumably due to their direct effect of inducing HO enzymatic activity in the bilirubin-producing pathway. However, the role of these polymorphisms seems limited for other perinatal complications such as bronchopulmonary dysplasia. We speculate that this is because the antioxidant or anti-inflammatory effect is not directly mediated by HO but by its byproducts, resulting in a milder effect. For better understanding, subtyping each morbidity by the level of exposure to causative environmental factors, simultaneous analysis of both polymorphisms, and the unified definition of short and long alleles in (GT)n repeats based on transcriptional capacity should be further investigated.
Collapse
|
13
|
Li Z, Song L, Hao L. The role of UGT1A1 (c.-3279 T > G) gene polymorphisms in neonatal hyperbilirubinemia susceptibility. BMC MEDICAL GENETICS 2020; 21:218. [PMID: 33158427 PMCID: PMC7648392 DOI: 10.1186/s12881-020-01155-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022]
Abstract
Background Neonatal hyperbilirubinemia (NNH) is a common disease in newborns. This research study aimed to assess the associations between uridine diphospho-glucuronate-glucuronosyltransferase 1A1 (UGT1A1, c.-3279 T > G) polymorphisms and NNH risk. Methods We searched PubMed, the Cochrane Library, and the Embase electronic databases. All published eligible studies before July 1, 2019, were searched for this meta-analysis. Results We identified 7 independent studies including 1560 cases. The data showed that in the general population, compared with the GT + GG vs TT and GG vs TT, c.-3279 T > G (rs4124874) was significantly related to a higher NNH risk (GG vs TT: OR = 1.865, 95% CI: 1.031–3.373, P = 0.039; GT + GG vs TT: OR = 1.331, 95% CI: 1.055–1.679, P = 0.016). Although not statistically significant, the data showed that c.3279 T > G had a tendency to be associated with NNH under the allele model and GG vs GT + TT in the overall population (G vs T: OR = 1.288, 95% CI: 0.982–1.689, P = 0.067; GG vs TT + GT: OR = 1.583, 95% CI: 0.947–2.647, P = 0.080). Conclusion The UGT1A1 gene c.-3279 T > G (rs4124874) polymorphism increased susceptibility to NNH, especially for the comparison of GT + GG vs TT and GG vs TT. In the future, we can use homozygous state of the UGT1A1 gene c.-3279 T > G (rs4124874) polymorphism for the diagnosis and screening of molecular biomarkers in NNH patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12881-020-01155-2.
Collapse
Affiliation(s)
- Zijin Li
- Department of Internal Medicine, Tianjin Children's Hospital, Tianjin, 300074, China
| | - Li Song
- Department of Neonatology, Tianjin Children's Hospital, No. 238 Longyan Road, Beichen district, Tianjin, 300074, China
| | - Lihong Hao
- Department of Neonatology, Tianjin Children's Hospital, No. 238 Longyan Road, Beichen district, Tianjin, 300074, China.
| |
Collapse
|
14
|
Hansen TWR, Wong RJ, Stevenson DK. Molecular Physiology and Pathophysiology of Bilirubin Handling by the Blood, Liver, Intestine, and Brain in the Newborn. Physiol Rev 2020; 100:1291-1346. [PMID: 32401177 DOI: 10.1152/physrev.00004.2019] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Bilirubin is the end product of heme catabolism formed during a process that involves oxidation-reduction reactions and conserves iron body stores. Unconjugated hyperbilirubinemia is common in newborn infants, but rare later in life. The basic physiology of bilirubin metabolism, such as production, transport, and excretion, has been well described. However, in the neonate, numerous variables related to nutrition, ethnicity, and genetic variants at several metabolic steps may be superimposed on the normal physiological hyperbilirubinemia that occurs in the first week of life and results in bilirubin levels that may be toxic to the brain. Bilirubin exists in several isomeric forms that differ in their polarities and is considered a physiologically important antioxidant. Here we review the chemistry of the bilirubin molecule and its metabolism in the body with a particular focus on the processes that impact the newborn infant, and how differences relative to older children and adults contribute to the risk of developing both acute and long-term neurological sequelae in the newborn infant. The final section deals with the interplay between the brain and bilirubin and its entry, clearance, and accumulation. We conclude with a discussion of the current state of knowledge regarding the mechanism(s) of bilirubin neurotoxicity.
Collapse
Affiliation(s)
- Thor W R Hansen
- Division of Paediatric and Adolescent Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; and Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Ronald J Wong
- Division of Paediatric and Adolescent Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; and Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - David K Stevenson
- Division of Paediatric and Adolescent Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; and Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
15
|
Review of bilirubin neurotoxicity I: molecular biology and neuropathology of disease. Pediatr Res 2020; 87:327-331. [PMID: 31600770 DOI: 10.1038/s41390-019-0608-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022]
Abstract
Despite the availability of successful prevention strategies to prevent excessive hyperbilirubinemia, the neurological sequelae of bilirubin neurotoxicity (BNTx) still occur throughout the world. Kernicterus, encephalopathy due to BNTx, is now understood to be a spectrum of severity and phenotypes known as kernicterus spectrum disorder (KSD). A better understanding of the selective neuropathology and molecular biology of BNTx and using consistent clinical definitions of KSDs as outcome measure can lead to more accurately predicting the risk and causes of BNTx and KSDs. In Part I of our two-part review, we will summarize current and recent advances in the understanding of the selective neuropathology and molecular biology of the disease. Herein we emphasize the role of unbound, free unconjugated bilirubin as well as genetic contributions to the susceptibility BNTx and the development of KSDs. In Part II, we focus on current and possible novel methods to prevent BNTx and ABE and treat ABE and KSDs.
Collapse
|
16
|
Vockley J, Dobrowolski SF, Arnold GL, Guerrero RB, Derks TGJ, Weinstein DA. Complex patterns of inheritance, including synergistic heterozygosity, in inborn errors of metabolism: Implications for precision medicine driven diagnosis and treatment. Mol Genet Metab 2019; 128:1-9. [PMID: 31358473 PMCID: PMC8931500 DOI: 10.1016/j.ymgme.2019.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 01/03/2023]
Abstract
Inborn errors of metabolism have traditionally been viewed as the quintessential single gene disorders; defects in one gene leads to loss of activity of one enzyme causing a metabolic imbalance and clinical disease. However, reality has never been quite that simple, and the classic "one gene-one enzyme" paradigm has been upended in many ways. Multiple gene defects can lead to the same biochemical phenotype, often with different clinical symptoms. Additionally, different mutations in the same gene can cause variable phenotypes, often most dramatic when a disease can be identified by pre-symptomatic screening. Moreover, response to therapy is not homogeneous across diseases and specific mutations. Perhaps the biggest deviation from traditional monogenic inheritance is in the setting of synergistic heterozygosity, a multigenic inheritance pattern in which mutations in multiple genes in a metabolic pathway lead to sufficient disruption of flux through the pathway, mimicking a monogenic disorder caused by homozygous defects in one gene in that pathway. In addition, widespread adoption of whole exome and whole genome sequencing in medical genetics has led to the realization that individual patients with apparently hybrid phenotypes can have mutations in more than one gene, leading to a mixed genetic disorder. Each of these situations point to a need for as much precision as possible in diagnosing metabolic disease, and it is likely to become increasingly critical to drive therapy. This article examines examples in traditional monogenic disorders that illustrates these points and define inborn errors of metabolism as complex genetic traits on the leading edge of precision medicine.
Collapse
Affiliation(s)
- Jerry Vockley
- University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America.
| | - Steven F Dobrowolski
- University of Pittsburgh School of Medicine, Department of Pathology, Pittsburgh, PA. UPMC Children's Hospital of Pittsburgh. 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America
| | - Georgianne L Arnold
- University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, United States of America
| | | | - Terry G J Derks
- Section of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, PO box 30 001, 9700, RB, Groningen, the Netherlands
| | - David A Weinstein
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, United States of America; GSD Program, Connecticut Children's Medical Center, Hartford, CT 06106, United States of America
| |
Collapse
|
17
|
Identification of Genetic Risk Factors for Neonatal Hyperbilirubinemia in Fujian Province, Southeastern China: A Case-Control Study. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7803175. [PMID: 30298137 PMCID: PMC6157199 DOI: 10.1155/2018/7803175] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 08/09/2018] [Indexed: 12/18/2022]
Abstract
To date, the genetic risk factors for neonatal hyperbilirubinemia remain unknown in Southeastern China. This case-control study aimed to identify the genetic risk factors for neonatal hyperbilirubinemia in Fujian, Southeastern China. A total of 286 hyperbilirubinemic newborns were enrolled as a case group, and 250 randomly selected newborns without jaundice or with a bilirubin level that was lower than the threshold required for phototherapy served as controls. The serum levels of total bilirubin, unconjugated bilirubin, and direct bilirubin were measured, and the common genetic loci in UGT1A1, OATP1B1, and HO-1 genes were genotyped. Higher incidence of ABO incompatibility and G6PD deficiency was detected in the case group compared to the control group (P < 0.01). There were significant differences in the frequencies of rs4148323 and rs1805173 genotypes between the case and control groups (P < 0.05). At the rs4148323 locus, the frequencies of GA heterozygotes and AA mutant homozygotes were higher in the case group than in the control group (P < 0.05), and at the rs1805173 locus, the frequencies of LS, MS, and SS genotypes were higher in the case group than in the control group (P < 0.05). A higher frequency of rs4148323 A allele and rs1805173 S allele was detected in the case group compared to the control group (P = 0). Additionally, multivariate logistic regression analysis identified that the mutant genotype of rs4148323 in the UGT1A1 gene, ABO incompatibility, G6PD deficiency, and SS genotype at rs1805173 locus of the HO-1 gene were genetic risk factors of neonatal hyperbilirubinemia. Our data demonstrate that G211 mutation in the UGT1A1 gene, ABO incompatibility, G6PD deficiency, and the SS genotype of the repeats in the promoter region of the HO-1 gene are risk factors for neonatal hyperbilirubinemia in Fujian, Southeastern China.
Collapse
|
18
|
Wagner KH, Shiels RG, Lang CA, Seyed Khoei N, Bulmer AC. Diagnostic criteria and contributors to Gilbert's syndrome. Crit Rev Clin Lab Sci 2018; 55:129-139. [PMID: 29390925 DOI: 10.1080/10408363.2018.1428526] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Hyperbilirubinemia is a well-known condition in the clinical setting; however, the causes of elevated serum bilirubin are diverse, as are the clinical ramifications of this condition. For example, diagnoses of individuals vary depending on whether they exhibit an unconjugated or conjugated hyperbilirubinemia. Diagnoses can include conditions of disordered bilirubin metabolism (Gilbert's, Crigler-Najjar, Rotor, or Dubin-Johnson syndromes) or an acquired disease, including alcoholic/non-alcoholic fatty liver disease, hepatotropic hepatitis, cirrhosis, or hepato-biliary malignancy. Assessment of bilirubin concentrations is typically conducted as part of routine liver function testing. Mildly elevated total bilirubin with normal serum activities of liver transaminases, biliary damage markers, and red blood cell counts, however, may indicate the presence of Gilbert's syndrome (GS), a benign condition that is present in ∼5-10% of the population. In this case, mildly elevated unconjugated bilirubin in GS is strongly associated with "reduced" prevalence of chronic diseases, particularly cardiovascular diseases (CVD) and type 2 diabetes mellitus (and associated risk factors), as well as CVD-related and all-cause mortality. These reports challenge the dogma that bilirubin is simply a potentially neurotoxic by-product of heme catabolism and emphasize the importance of understanding its potential beneficial physiologic and detrimental pathophysiologic effects, in order to appropriately consider bilirubin test results within the clinical laboratory setting. With this information, we hope to improve the understanding of disorders of bilirubin metabolism, emphasize the diagnostic importance of these conditions, and outline the potential impact GS may have on resistance to disease.
Collapse
Affiliation(s)
- Karl-Heinz Wagner
- a Department of Nutritional Sciences and Research Platform Active Ageing , University of Vienna , Vienna , Austria
| | - Ryan G Shiels
- b School of Medical Science and Menzies Health Institute Queensland , Griffith University , Gold Coast , Australia
| | - Claudia Anna Lang
- a Department of Nutritional Sciences and Research Platform Active Ageing , University of Vienna , Vienna , Austria
| | - Nazlisadat Seyed Khoei
- a Department of Nutritional Sciences and Research Platform Active Ageing , University of Vienna , Vienna , Austria
| | - Andrew C Bulmer
- b School of Medical Science and Menzies Health Institute Queensland , Griffith University , Gold Coast , Australia
| |
Collapse
|
19
|
Bockor L, Bortolussi G, Vodret S, Iaconcig A, Jašprová J, Zelenka J, Vitek L, Tiribelli C, Muro AF. Modulation of bilirubin neurotoxicity by the Abcb1 transporter in the Ugt1-/- lethal mouse model of neonatal hyperbilirubinemia. Hum Mol Genet 2017; 26:145-157. [PMID: 28025333 DOI: 10.1093/hmg/ddw375] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/27/2016] [Indexed: 01/20/2023] Open
Abstract
Moderate neonatal jaundice is the most common clinical condition during newborn life. However, a combination of factors may result in acute hyperbilirubinemia, placing infants at risk of developing bilirubin encephalopathy and death by kernicterus. While most risk factors are known, the mechanisms acting to reduce susceptibility to bilirubin neurotoxicity remain unclear. The presence of modifier genes modulating the risk of developing bilirubin-induced brain damage is increasingly being recognised. The Abcb1 and Abcc1 members of the ABC family of transporters have been suggested to have an active role in exporting unconjugated bilirubin from the central nervous system into plasma. However, their role in reducing the risk of developing neurological damage and death during neonatal development is still unknown.To this end, we mated Abcb1a/b-/- and Abcc1-/- strains with Ugt1-/- mice, which develop severe neonatal hyperbilirubinemia. While about 60% of Ugt1-/- mice survived after temporary phototherapy, all Abcb1a/b-/-/Ugt1-/- mice died before postnatal day 21, showing higher cerebellar levels of unconjugated bilirubin. Interestingly, Abcc1 role appeared to be less important.In the cerebellum of Ugt1-/- mice, hyperbilirubinemia induced the expression of Car and Pxr nuclear receptors, known regulators of genes involved in the genotoxic response.We demonstrated a critical role of Abcb1 in protecting the cerebellum from bilirubin toxicity during neonatal development, the most clinically relevant phase for human babies, providing further understanding of the mechanisms regulating bilirubin neurotoxicity in vivo. Pharmacological treatments aimed to increase Abcb1 and Abcc1 expression, could represent a therapeutic option to reduce the risk of bilirubin neurotoxicity.
Collapse
Affiliation(s)
- Luka Bockor
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Simone Vodret
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Iaconcig
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Jana Jašprová
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technicka 3, 166 28 Prague, Czech Republic
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic.,Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Claudio Tiribelli
- Centro Studi Fegato, Fondazione Italiana Fegato, AREA Science Park, Campus Basovizza Trieste, Italy and
| | - Andrés F Muro
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| |
Collapse
|
20
|
Chung EK, Gable EK, Golden WC, Hudson JA, Hackman NM, Andrews JP, Jackson DS, Beavers JB, Mirchandani DR, Kellams A, Krevitsky ME, Monroe K, Madlon-Kay DJ, Stratbucker W, Campbell D, Collins J, Rauch D. Current Scope of Practice for Newborn Care in Non-Intensive Hospital Settings. Hosp Pediatr 2017; 7:471-482. [PMID: 28694290 DOI: 10.1542/hpeds.2016-0206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Esther K Chung
- Department of Pediatrics, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania and Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware;
| | - E Kaye Gable
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina and Cone Health, Greensboro, North Carolina
| | - W Christopher Golden
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer A Hudson
- Department of Pediatrics, Greenville Health System, Greenville, South Carolina
| | - Nicole M Hackman
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jennifer P Andrews
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - DeeAnne S Jackson
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jessica B Beavers
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Dipti R Mirchandani
- Department of Pediatrics, Hofstra Northwell School of Medicine at Hofstra University, Hempstead, New York and Cohen Children's Medical Center of New York, New Hyde Park, New York
| | - Ann Kellams
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Meredith E Krevitsky
- Department of Pediatrics, Hofstra Northwell School of Medicine at Hofstra University, Hempstead, New York and Cohen Children's Medical Center of New York, New Hyde Park, New York
| | - Kimberly Monroe
- Department of Pediatrics and Communicable Diseases, C.S. Mott Children's Hospital and University of Michigan, Ann Arbor, Michigan
| | - Diane J Madlon-Kay
- Department of Family Medicine and Community Health, University of Minnesota Medical School, Minneapolis, Minnesota
| | - William Stratbucker
- Department of Pediatrics, Michigan State University and Helen DeVos Children's Hospital, Grand Rapids, Michigan
| | - Deborah Campbell
- Department of Pediatrics, Albert Einstein College of Medicine, New York, New York and Children's Hospital at Montefiore, Bronx, New York
| | - Jolene Collins
- Department of Pediatrics, University of Southern California Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California; and
| | - Daniel Rauch
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, Elmhurst, New York
| |
Collapse
|
21
|
Golden WC. The African-American neonate at risk for extreme hyperbilirubinemia: a better management strategy is needed. J Perinatol 2017; 37:321-322. [PMID: 28400613 DOI: 10.1038/jp.2017.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- W C Golden
- Division of Neonatology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
22
|
Schutzman DL, Baudhuin LM, Gatien E, Ajayi S, Wong RJ. Effect of genetic variants of bilirubin metabolism on the degree of hyperbilirubinemia in African-American newborns. J Perinatol 2017; 37:432-435. [PMID: 27977017 DOI: 10.1038/jp.2016.232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/23/2016] [Accepted: 11/04/2016] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The objective of our study was to measure the effect of genetic variants of these two enzymes, UGT1A1 and SLCO1B1, in the bilirubin metabolic pathway on the degree of hyperbilirubinemia in a cohort of African-American (AA) infants from our well-baby nursery. In addition, a second objective was to document the types and frequencies of genetic variations of these enzymes in our cohort. STUDY DESIGN A prospective study of 180 AA infants from the Well Baby Nursery of an inner city community hospital, all of whose mothers were type O pos. Sixty infants were ABO-incompatible direct antiglobulin test (DAT) pos, 60 were ABO-incompatible DAT neg and 60 were type O+. Blood for carboxyhemoglobin (COHb) and variants of the enzymes uridine diphosphoglucuronosyltransferase 1A1 and hepatic solute carrier organic anion transporter 1B1 (SLCO1B1) was drawn at the time of the infants' initial bilirubin, and the infants' precise percentile on the Bhutani nomogram was calculated. RESULTS Variants in the two enzymes studied were quite common. In total, 21.1% were positive for a Gilbert phenotype, whereas an additional 42.4% were heterozygous for the *28 or *37 variant of UGT1A1. In total, 67.2% were homozygous for the *60 variant of the phenobarbital responsive enhancer module. In total, 41.1% were homozygous for the *1b variant of SLCO1B1, whereas an additional 12.7% were positive for the *4 variant of this gene. In total, 20.6% of infants had variations in both genes. Using logistic regression when COHbc was assessed with each of the different variants, only COHbc (P<0.0001 to 0.0004) was significantly associated with the level of hyperbilirubinemia as defined by the Bhutani nomogram. CONCLUSION Although we have found quite a large number of genetic variants of the UGT1A1 and SLCO1B1 genes in the AA population, it does not appear that they have a significant impact on the incidence of hyperbilirubinemia among this group of infants.
Collapse
Affiliation(s)
- D L Schutzman
- Department of Pediatrics and Adolescent Medicine, Einstein Medical Center, Philadelphia, PA, USA
| | - L M Baudhuin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - E Gatien
- Department of Pediatrics, Michigan State University, Lansing, MI, USA
| | - S Ajayi
- Division of Neonatology, Saint Christopher's Hospital for Children, Philadelphia, PA, USA
| | - R J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
23
|
Cervantes-Alvarez E, Wang Y, Collin de l'Hortet A, Guzman-Lepe J, Zhu J, Takeishi K. Current strategies to generate mature human induced pluripotent stem cells derived cholangiocytes and future applications. Organogenesis 2017; 13:1-15. [PMID: 28055309 DOI: 10.1080/15476278.2016.1278133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stem cell research has significantly evolved over the last few years, allowing the differentiation of pluripotent cells into almost any kind of lineage possible. Studies that focus on the liver have considerably taken a leap into this novel technology, and hepatocyte-like cells are being generated that are close to resembling actual hepatocytes both genotypically and phenotypically. The potential of this extends from disease models to bioengineering, and even also innovative therapies for end-stage liver disease. Nonetheless, too few attention has been given to the non-parenchymal cells which are also fundamental for normal liver function. This includes cholangiocytes, the cells of the biliary epithelium, without whose role in bile modification and metabolism would impair hepatocyte survival. Such can be observed in diseases that target them, so called cholangiopathies, for which there is much yet to study so as to improve therapeutical options. Protocols that describe the induction of human induced pluripotent stem cells into cholangiocytes are scarce, although progress is being achieved in this area as well. In order to give the current view on this emerging research field, and in hopes to motivate further advances, we present here a review on the known differentiation strategies with sight into future applications.
Collapse
Affiliation(s)
- Eduardo Cervantes-Alvarez
- a Department of Pathology , University of Pittsburgh , Pittsburgh , PA , USA.,b PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México , Mexico City , México
| | - Yang Wang
- a Department of Pathology , University of Pittsburgh , Pittsburgh , PA , USA.,c Department of Hepatobiliary Surgery , Peking University People's Hospital , Beijing , China
| | | | - Jorge Guzman-Lepe
- a Department of Pathology , University of Pittsburgh , Pittsburgh , PA , USA
| | - Jiye Zhu
- c Department of Hepatobiliary Surgery , Peking University People's Hospital , Beijing , China
| | - Kazuki Takeishi
- a Department of Pathology , University of Pittsburgh , Pittsburgh , PA , USA
| |
Collapse
|
24
|
Tomerak RH, Helal NF, Shaker OG, Yousef MA. Association between the Specific UGT1A1 Promoter Sequence Variant (c-3279T>G) and Unconjugated Neonatal Hyperbilirubinemia. J Trop Pediatr 2016; 62:457-463. [PMID: 27318112 DOI: 10.1093/tropej/fmw031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We investigated the association between c-3279T>G and unconjugated neonatal hyperbilirubinemia. In all, 141 neonates were recruited; 63 had hyperbilirubinemia necessitating treatment, and 78 with bilirubin < 7 mg/dl served as the control group. The frequency of occurrence of c-3279T > G allele was significantly higher in the hyperbilirubinemic (49.2%) than in the control group (25.6%). The homozygous (p = 0.001, OR = 17.7 and CI = 3.9-79.3) rather than the heterozygous state (p = 0.3, OR = 0.7 and CI = 0.3-1.6) was associated with hyperbilirubinemia. Among the hyperbilirubinemic group, comparison between the three genotypes, homozygous mutation, heterozygous mutation and the normal allele, revealed that the former was associated with significantly higher mean peak total serum bilirubin [mean ± standard deviation (SD): 33.7 ± 8.2, 26.9 ± 2.8 and 21± 2.7, respectively, p-value = 0.0001], higher bilirubin/albumin ratio (p = 0.000) and a longer duration of hospital stay (p = 0.001). Homozygous c-3279T > G mutation represents an important risk factor for the development of neonatal hyperbilirubinemia.
Collapse
|
25
|
Riordan SM, Bittel DC, Le Pichon JB, Gazzin S, Tiribelli C, Watchko JF, Wennberg RP, Shapiro SM. A Hypothesis for Using Pathway Genetic Load Analysis for Understanding Complex Outcomes in Bilirubin Encephalopathy. Front Neurosci 2016; 10:376. [PMID: 27587993 PMCID: PMC4988977 DOI: 10.3389/fnins.2016.00376] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/02/2016] [Indexed: 01/18/2023] Open
Abstract
Genetic-based susceptibility to bilirubin neurotoxicity and chronic bilirubin encephalopathy (kernicterus) is still poorly understood. Neonatal jaundice affects 60–80% of newborns, and considerable effort goes into preventing this relatively benign condition from escalating into the development of kernicterus making the incidence of this potentially devastating condition very rare in more developed countries. The current understanding of the genetic background of kernicterus is largely comprised of mutations related to alterations of bilirubin production, elimination, or both. Less is known about mutations that may predispose or protect against CNS bilirubin neurotoxicity. The lack of a monogenetic source for this risk of bilirubin neurotoxicity suggests that disease progression is dependent upon an overall decrease in the functionality of one or more essential genetically controlled metabolic pathways. In other words, a “load” is placed on key pathways in the form of multiple genetic variants that combine to create a vulnerable phenotype. The idea of epistatic interactions creating a pathway genetic load (PGL) that affects the response to a specific insult has been previously reported as a PGL score. We hypothesize that the PGL score can be used to investigate whether increased susceptibility to bilirubin-induced CNS damage in neonates is due to a mutational load being placed on key genetic pathways important to the central nervous system's response to bilirubin neurotoxicity. We propose a modification of the PGL score method that replaces the use of a canonical pathway with custom gene lists organized into three tiers with descending levels of evidence combined with the utilization of single nucleotide polymorphism (SNP) causality prediction methods. The PGL score has the potential to explain the genetic background of complex bilirubin induced neurological disorders (BIND) such as kernicterus and could be the key to understanding ranges of outcome severity in complex diseases. We anticipate that this method could be useful for improving the care of jaundiced newborns through its use as an at-risk screen. Importantly, this method would also be useful in uncovering basic knowledge about this and other polygenetic diseases whose genetic source is difficult to discern through traditional means such as a genome-wide association study.
Collapse
Affiliation(s)
- Sean M Riordan
- Division of Child Neurology, Department of Pediatrics, Children's Mercy HospitalKansas City, MO, USA; Department of Neurology, University of Kansas Medical CenterKansas City, KS, USA
| | - Douglas C Bittel
- Ward Family Heart Center, Children's Mercy HospitalKansas City, MO, USA; Department of Pediatrics, University of Missouri-Kansas City School of MedicineKansas City, MO, USA
| | - Jean-Baptiste Le Pichon
- Division of Child Neurology, Department of Pediatrics, Children's Mercy HospitalKansas City, MO, USA; Department of Neurology, University of Kansas Medical CenterKansas City, KS, USA; Department of Pediatrics, University of Missouri-Kansas City School of MedicineKansas City, MO, USA; Department of Pediatrics, University of Kansas Medical CenterKansas City, KS, USA
| | - Silvia Gazzin
- Italian Liver Foundation, Centro Studi Fegato (CSF) Trieste, Italy
| | - Claudio Tiribelli
- Italian Liver Foundation, Centro Studi Fegato (CSF)Trieste, Italy; Department of Medical Sciences, University of TriesteTrieste, Italy
| | - Jon F Watchko
- Division of Newborn Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | | | - Steven M Shapiro
- Division of Child Neurology, Department of Pediatrics, Children's Mercy HospitalKansas City, MO, USA; Department of Neurology, University of Kansas Medical CenterKansas City, KS, USA; Department of Pediatrics, University of Missouri-Kansas City School of MedicineKansas City, MO, USA; Department of Pediatrics, University of Kansas Medical CenterKansas City, KS, USA
| |
Collapse
|
26
|
Yang H, Wang Q, Zheng L, Zheng XB, Lin M, Zhan XF, Yang LY. Clinical Significance of UGT1A1 Genetic Analysis in Chinese Neonates with Severe Hyperbilirubinemia. Pediatr Neonatol 2016; 57:310-7. [PMID: 26727668 DOI: 10.1016/j.pedneo.2015.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/12/2015] [Accepted: 08/14/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Neonatal hyperbilirubinemia is common in Asia, and the importance of genetically determined conditions has been recently recognized. The aim of this study was to assess the clinical utility of genetic testing in Chinese neonates with severe hyperbilirubinemia. METHODS Fifty-eight term infants with bilirubin level ≥ 20 mg/dL (342 μmol/L), and 65 controls were enrolled in the study. Variation status of UGT1A1, G6PD, and thalassemia genes in our study cohort was determined by direct sequencing or genotype assays. RESULTS Among these case infants, seven were confirmed with G6PD deficiency, four were heterozygous for α- or β-thalassemia, and forty-four were detected with at least one heterozygous UGT1A1 functional variant, including nine homozygous for UGT1A1 variation. As well as the predominant c.211G>A (Gly71Arg) variant, three UGT1A1 coding variants [c.1091C>T (Pro364Leu), c.1352C>T (pro451leu), and c.1456C>T (Tyr486Asp)] were observed in our case neonates. The results of multivariate logistic regressions, adjusted for covariates, revealed odds ratios for neonates who carried heterozygous, homozygous variation at nucleotide 211 of UGT1A1, and G6PD deficiency of 3.47 (1.26-9.55), 12.46 (1.09-142.7) ,and 12.87 (1.32-135.87) compared with those having the wild genotype and normal G6PD activity, respectively. CONCLUSION Besides G6PD-deficiency screening, UGT1A1 genetic analysis, and especially the UGT1A1*6(c.211G>A, p.Arg71Gly) polymorphism detection, may be taken into consideration for early diagnosis and treatment of severe hyperbilirubinemic newborns in southern China.
Collapse
Affiliation(s)
- Hui Yang
- Laboratory Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China; Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, PR China
| | - Qian Wang
- Laboratory Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.
| | - Lei Zheng
- Laboratory Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China
| | - Xiang-Bin Zheng
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, PR China
| | - Min Lin
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, PR China
| | - Xiao-Fen Zhan
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, PR China
| | - Li-Ye Yang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, PR China.
| |
Collapse
|
27
|
Cheung TP, Van Rostenberghe H, Ismail R, Nawawi NN, Abdullah NA, Ramli N, Ibrahim NR, Hj. Abd. Majid N, Mohd Yusoff N, Nishio H, Yusoff S. High resolution melting analysis of the NR1I3 genetic variants: Is there an association with neonatal hyperbilirubinemia? Gene 2015; 573:198-204. [DOI: 10.1016/j.gene.2015.07.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/28/2015] [Accepted: 07/13/2015] [Indexed: 01/11/2023]
|
28
|
Yu Z, Zhu K, Wang L, Liu Y, Sun J. Association of Neonatal Hyperbilirubinemia with UGT1A1 Gene Polymorphisms: A Meta-Analysis. Med Sci Monit 2015; 21:3104-14. [PMID: 26467199 PMCID: PMC4612146 DOI: 10.12659/msm.894043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background The results of studies on association between the polymorphisms in the coding region and the promoter of uridine diphosphateglucuronosyl transferase 1A1 (UGT1A1) and neonatal hyperbilirubinemia are controversial. This study aimed to determine whether the UGT1A1 gene polymorphisms of Gly71Arg and TATA promoter were significant risk factors associated with neonatal hyperbilirubinemia. Material/Methods The PubMed, Cochrane Library, and Embase databases were searched for papers that describe the association between UGT1A1 polymorphisms and neonatal hyperbilirubinemia. Summary odds ratios and 95% confidence intervals (CI) were estimated based on a fixed-effects model or random-effects model, depending on the absence or presence of significant heterogeneity. Results A total of 32 eligible studies and 6520 participants were identified. Among them, 24 studies focused on the association of neonatal hyperbilirubinemia with UGT1A1 Gly71Arg polymorphisms, and a significant difference was found for the comparison of AA vs. AG+GG (OR=3.47, 95% CI=2.29–5.28, P<0.0001). We included 19 studies on the association of neonatal hyperbilirubinemia with UGT1A1 TATA promoter polymorphism, which also found a statistically significant difference between 7/7 and 6/7 + 6/6 (OR=2.24, 95% CI=1.29–3.92, P=0.004). Conclusions This meta-analysis demonstrated that UGT1A1 polymorphisms (Gly71Arg and TATA promoter) significantly increase the risk of neonatal hyperbilirubinemia.
Collapse
Affiliation(s)
- Zibi Yu
- Songjiang Branch of The Affiliated Shuguang Hospital of Shanghai University of TCM, Shanghai, China (mainland)
| | - Kaichang Zhu
- Department of Urology, Fengxian District Central Hospital, Shanghai, China (mainland)
| | - Li Wang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Dalian City, Dalian, Liaoning, China (mainland)
| | - Ying Liu
- Department of Neonatology, Obstetrics and Gynecology Hospital of Dalian City, Dalian, Liaoning, China (mainland)
| | - Jianmei Sun
- Department of Neonatology, Affiliated Tongji Hospital of Tongji University, Shanghai, China (mainland)
| |
Collapse
|
29
|
Katayama Y, Yokota T, Zhao H, Wong RJ, Stevenson DK, Taniguchi-Ikeda M, Nakamura H, Iijima K, Morioka I. Association of HMOX1 gene promoter polymorphisms with hyperbilirubinemia in the early neonatal period. Pediatr Int 2015; 57:645-9. [PMID: 25625535 DOI: 10.1111/ped.12591] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/13/2015] [Accepted: 01/19/2015] [Indexed: 11/27/2022]
Abstract
BACKGROUND Heme oxygenase (HO) is the rate-limiting enzyme in the heme degradation pathway that produces bilirubin. The promoter region of human heme oxygenase-1 (HMOX1) contains a polymorphic (GT)n repeat that can regulate gene expression. Here, we investigated the association of (GT)n repeat length in the HMOX1 promoter region with neonatal hyperbilirubinemia in a population of Japanese term neonates. METHODS Using polymerase chain reaction and fragment analysis, we determined the number of (GT)n repeats in 149 Japanese neonates. To omit the effects of the G71R mutation in uridine diphosphoglucuronosyltransferase on hyperbilirubinemia, we excluded 41 neonates with the G71R mutation. As a result, 25 neonates with hyperbilirubinemia and 83 non-hyperbilirubinemic controls were included in this prospective case-control study. Allele and genotype frequencies of (GT)n repeats in the HMOX1 gene were compared between hyperbilirubinemic and non-hyperbilirubinemic control neonates. RESULTS The prevalence of short alleles (< 22 (GT)n repeats) was significantly higher in hyperbilirubinemic than in control neonates (18% vs 7%, P = 0.015). Hyperbilirubinemia was more frequent in homozygous or heterozygous short allele carriers than control neonates (28% vs 11%, respectively, P = 0.03). Possession of short alleles was significantly associated with the development of neonatal hyperbilirubinemia (OR, 3.1; 95%CI: 1.03-9.53). CONCLUSIONS Infants carrying short alleles (< 22 (GT)n repeats) in the HMOX1 gene promoter region appear to be at a higher risk for developing neonatal hyperbilirubinemia.
Collapse
Affiliation(s)
- Yoshinori Katayama
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan.,Department of Pediatrics, Takatsuki General Hospital, Takatsuki, Japan
| | - Tomoyuki Yokota
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hui Zhao
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Ronald J Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | - Hajime Nakamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ichiro Morioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
30
|
Petersen JP, Henriksen TB, Hollegaard MV, Vandborg PK, Hougaard DM, Thorlacius-Ussing O, Ebbesen F. Extreme neonatal hyperbilirubinemia and a specific genotype: a population-based case-control study. Pediatrics 2014; 134:510-5. [PMID: 25092941 DOI: 10.1542/peds.2014-0035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES Extreme hyperbilirubinemia (plasma bilirubin ≥ 24.5 mg/dL) is an important risk factor for severe bilirubin encephalopathy. Several risk factors for hyperbilirubinemia are known, but in a large number of patients, a causal factor is never established. UGT1A1 is the rate-limiting enzyme in bilirubin's metabolism. The genotype of Gilbert syndrome, the UGT1A1*28 allele, causes markedly reduced activity of this enzyme, but its association with neonatal hyperbilirubinemia is uncertain and its relationship with extreme hyperbilirubinemia has not been studied. We examined whether the UGT1A1*28 allele is associated with extreme hyperbilirubinemia. METHODS The UGT1A1*28 allele was assessed in a case-control study of 231 white infants who had extreme hyperbilirubinemia in Denmark from 2000 to 2007 and 432 white controls. Cases were identified in the Danish Extreme Hyperbilirubinemia Database that covers the entire population. Genotypes were obtained through the Danish Neonatal Screening Biobank. Subgroup analysis was done for AB0 incompatible cases. RESULTS No association was found between the UGT1A1*28 allele and extreme hyperbilirubinemia. With the common genotype as reference, the odds ratio of extreme hyperbilirubinemia was 0.87 (range, 0.68-1.13) for UGT1A1*28 heterozygotes and 0.77 (range, 0.46-1.27) for homozygotes. Also, no association was found for AB0 incompatible cases. CONCLUSIONS The UGT1A1*28 allele was not associated with risk for extreme hyperbilirubinemia in this study.
Collapse
Affiliation(s)
- Jesper Padkær Petersen
- Pediatric Department, and Pediatric Department, Aarhus University Hospital, Aarhus, Denmark; and
| | | | | | | | | | | | | |
Collapse
|
31
|
Zhou Y, Wang SN, Li H, Zha W, Wang X, Liu Y, Sun J, Peng Q, Li S, Chen Y, Jin L. Association of UGT1A1 variants and hyperbilirubinemia in breast-fed full-term Chinese infants. PLoS One 2014; 9:e104251. [PMID: 25102181 PMCID: PMC4125195 DOI: 10.1371/journal.pone.0104251] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 06/20/2014] [Indexed: 01/10/2023] Open
Abstract
A retrospective case control study of breast-fed full-term infants was carried out to determine whether variants in Uridine Diphosphate Glucuronosyl Transferase 1A1 (UGT1A1) and Heme Oxygenase-1 (HMOX1) were associated with neonatal hyperbilirubinemia. Eight genetic variants of UGT1A1 and 3 genetic variants of HMOX1 were genotyped in 170 hyperbilirubinemic newborns and 779 controls. Five significant associations with breast-fed hyperbilirubinemia were detected after adjusting for gender, birth season, birth weight, delivery mode, gestational age and False Discovery Rate (FDR) correction: the dominant effect of rs887829 (c-364t) (Odds Ratio (OR): 0.55; 95% Confidence Interval (CI): 0.34-0.89; p = 0.014), the additive effect of (TA)n repeat (OR: 0.59; 95%CI: 0.38-0.91; p = 0.017), the dominant effect of rs4148323 (Gly71Arg, G211A) (OR: 2.02; 95%CI: 1.44-2.85; p = 5.0×10-5), the recessive effect of rs6717546 (g+914a) (OR: 0.30; 95%CI: 0.11-0.83; p = 0.021) and rs6719561 (t+2558c) (OR: 0.38; 95%CI: 0.20-0.75; p = 0.005). Neonates carrying the minor allele of rs887829 (TA)n repeat had significantly lower peak bilirubin than wild types, while the minor allele carriers of rs4148323 had significantly higher peak bilirubin than wild types. No association was found in HMOX1. Our findings added to the understanding of the significance of UGT1A1 in association with neonatal hyperbilirubinemia in East Asian population. Additional studies were required to investigate the mechanisms of the protective effects.
Collapse
Affiliation(s)
- Youyou Zhou
- Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - San-nan Wang
- Department of Neonatology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Hong Li
- Center for Reproduction and Genetics and Suzhou Maternal-Child Medical Center, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Weifeng Zha
- Department of Neonatology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Xuli Wang
- Department of Neonatology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Yuanyuan Liu
- Department of Neonatology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Jian Sun
- Center for Reproduction and Genetics and Suzhou Maternal-Child Medical Center, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Qianqian Peng
- Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shilin Li
- Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Ying Chen
- Center for Reproduction and Genetics and Suzhou Maternal-Child Medical Center, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Li Jin
- Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Ho HY, Cheng ML, Chiu DTY. Glucose-6-phosphate dehydrogenase--beyond the realm of red cell biology. Free Radic Res 2014; 48:1028-48. [PMID: 24720642 DOI: 10.3109/10715762.2014.913788] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is critical to the maintenance of NADPH pool and redox homeostasis. Conventionally, G6PD deficiency has been associated with hemolytic disorders. Most biochemical variants were identified and characterized at molecular level. Recently, a number of studies have shone light on the roles of G6PD in aspects of physiology other than erythrocytic pathophysiology. G6PD deficiency alters the redox homeostasis, and affects dysfunctional cell growth and signaling, anomalous embryonic development, and altered susceptibility to infection. The present article gives a brief review of basic science and clinical findings about G6PD, and covers the latest development in the field. Moreover, how G6PD status alters the susceptibility of the affected individuals to certain degenerative diseases is also discussed.
Collapse
Affiliation(s)
- H-Y Ho
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University , Kwei-san, Tao-yuan , Taiwan
| | | | | |
Collapse
|
33
|
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; 172:1627-32. [PMID: 23877636 DOI: 10.1007/s00431-013-2091-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/01/2013] [Indexed: 11/29/2022]
Abstract
Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in bilirubin metabolism, and its genetic variant may modulate hyperbilirubinemia risk in neonates. The aim of the present study was to assess the association between heme oxygenase-1 gene variants and hyperbilirubinemia risk in Indian newborns. In a prospective case-control study, we analyzed (GT)n repeats and g.-413A>T variant of HO-1 gene and UGT1A1 gene variants in 100 case newborns with total serum bilirubin (TSB) levels exceeding 95th percentile and 100 control newborns with TSB levels below 75th percentile on the hour-specific bilirubin nomogram of the American Academy of Pediatrics. Study population consisted of term (37-41 weeks) and late preterm (34-36 weeks) newborns during the first 2 weeks of age. In our analysis, the (GT)n allele was highly polymorphic, ranging in number from 15 to 40. The incidence of short (GT)n allele (≤ 20) was significantly higher in neonates with hyperbilirubinemia than in controls. Although g.-413A>T variant was widely prevalent in the study population, no difference was noted in its prevalence between cases and controls. Short (GT)n repeats of HO-1 gene, c.211G>A variant of UGT1A1 gene, and excessive weight loss were independent risk factors for neonatal hyperbilirubinemia. In the presence of two or more risk factors, the odds of developing neonatal hyperbilirubinemia were high. Shorter (GT)n genotype in the promoter region of HO-1 gene is significantly associated with hyperbilirubinemia risk in Indian newborns. This genotype may interact with other genetic and clinical risk factors to further potentiate hyperbilirubinemia risk in newborns.
Collapse
Affiliation(s)
- Pankaj Kumar Tiwari
- Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221 005, India
| | | | | | | | | |
Collapse
|
34
|
Abstract
In the neonate, the liver is relatively immature and undergoes several changes in its functional capacity during the early postnatal period. The essential liver functions can be classified into three categories: metabolism, detoxification, and bile synthesis. In general, the immature liver function has limited consequences on the healthy term neonate. However, preterm neonates are particularly susceptible to the effects of the immature liver function placing them at risk of hypoglycemia, hyperbilirubinemia, cholestasis, bleeding, and impaired drug metabolism. An appreciation of the dynamic changes in liver function during the neonatal period is essential for successful management of neonates who require medical and surgical interventions. This review will focus on the neonatal liver function as well as the changes that the liver undergoes as it matures.
Collapse
Affiliation(s)
- James Grijalva
- Department of Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, Massachusetts 02115
| | - Khashayar Vakili
- Department of Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, Massachusetts 02115.
| |
Collapse
|
35
|
Žaja O, Tiljak MK, Štefanović M, Tumbri J, Jurčić Z. Correlation of UGT1A1 TATA-box polymorphism and jaundice in breastfed newborns-early presentation of Gilbert's syndrome. J Matern Fetal Neonatal Med 2013; 27:844-50. [PMID: 23981182 DOI: 10.3109/14767058.2013.837879] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The etiology of jaundice in otherwise healthy breastfed newborns that can present as early-onset exaggerated physiologic jaundice, or late breast milk jaundice (BMJ), is not yet entirely understood. This study tested the hypothesis that molecular marker for Gilbert's syndrome (GS), UGT1A1 TATA-box polymorphism, is associated with this disorders. METHODS We have investigated the UGT1A1 polymorphism frequency and its relation to severity of hyperbilirubinemia and jaundice duration among 220 exclusively breastfed term newborns; 57 of them with non-physiologic hyperbilirubinemia (NH), and 163 with BMJ, and in 187 healthy controls. RESULTS Significant differences in TA7/7 genotype frequency were established. The highest frequency was observed among the newborns with BMJ (42.0%), intermediate in the NH group (24.6%), while the controls had the lowest TA7/7 frequency (12.8%). Linear increase in TA7/7 frequency was observed depending on the duration of jaundice, peaking at 42.4% in newborns with the longest jaundice duration. Positive correlation between the serum bilirubin levels and the TATA-box length was established in all groups. CONCLUSION This study provides evidence that UGT1A1 TATA-box polymorphism is an important risk factor for developing jaundice in term breastfed newborns, presented as either early non-physiologic hyperbilirubinemia or breast milk jaundice. These results further support the original Odell's idea of neonatal jaundice as an early presentation of GS.
Collapse
Affiliation(s)
- Orjena Žaja
- Department of Pediatric Gastroenterology and Hepatology, Clinical Hospital Centre Sestre Milosrdnice , Zagreb , Croatia
| | | | | | | | | |
Collapse
|
36
|
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. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2013. [DOI: 10.1016/j.jpedp.2013.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
37
|
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 (Rio J) 2013; 89:434-43. [PMID: 23850112 DOI: 10.1016/j.jped.2013.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 01/30/2013] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To determine whether three variants (388 G>A, 521 T>C, and 463 C>A) of the solute carrier organic anion transporter family member 1B1 (SLCO1B1) are associated with neonatal hyperbilirubinemia. DATA SOURCE The China National Knowledge Infrastructure and MEDLINE databases were searched. The systematic review with meta-analysis included genetic studies which assessed the association between neonatal hyperbilirubinemia and 388 G>A, 521 T>C, and 463 C>A variants of SLCO1B1 between January of 1980 and December of 2012. Data selection and extraction were performed independently by two reviewers. SUMMARY OF THE FINDINGS Ten articles were included in the study. The results revealed that SLCO1B1 388 G>A is associated with an increased risk of neonatal hyperbilirubinemia (OR, 1.39; 95% CI, 1.07-1.82) in Chinese neonates, but not in white, Thai, Latin American, or Malaysian neonates. The SLCO1B1 521 T>C mutation showed a low risk of neonatal hyperbilirubinemia in Chinese neonates, while no significant associations were found in Brazilian, white, Asian, Thai, and Malaysian neonates. There were no significant differences in SLCO1B1 463 C>A between the hyperbilirubinemia and the control group. CONCLUSION This study demonstrated that the 388 G>A mutation of the SLCO1B1 gene is a risk factor for developing neonatal hyperbilirubinemia in Chinese neonates, but not in white, Thai, Brazilian, or Malaysian populations; the SLCO1B1 521 T>C mutation provides protection for neonatal hyperbilirubinemia in Chinese neonates, but not in white, Thai, Brazilian, or Malaysian populations.
Collapse
Affiliation(s)
- Jiebo Liu
- PhD. Department of Pediatrics, The Fifth People's Hospital of Shenzhen, Shenzhen, China.
| | | | | | | | | |
Collapse
|
38
|
Polymorphic variants of SLCO1B1 in neonatal hyperbilirubinemia in China. Ital J Pediatr 2013; 39:49. [PMID: 24090270 PMCID: PMC3750622 DOI: 10.1186/1824-7288-39-49] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/10/2013] [Indexed: 12/02/2022] Open
Abstract
Background To evaluate the association between the genetic polymorphism of the solute carrier organic anion transporter family member 1B1 (SLCO1B1, also known as organic anion transport polypeptide C) and hyperbilirubinemia in Chinese neonates. Methods 183 infants with hyperbilirubinemia and 192 control subjects from the Fifth People’s Hospital of Shenzhen were recruited. Polymerase chain reaction, restriction fragment length polymorphisms and agarose gel electrophoresis techniques were used to detect genetic variants of SLCO1B1. Results The study revealed that SLCO1B1 388 G > A occurred significantly more frequently in neonates with hyperbilirubinemia than in controls (RR = 1.50; 95% CI: 1.13–2.00). There were no significant differences in SLCO1B1 521 T > C between the hyperbilirubinemia and the control group (RR, 1.00; 95% CI, 0.72–1.40). No carriage of the C to A substitution at nucleotide 463 was detected. Conclusion The SLCO1B1 388 G > A variant is associated with neonatal hyperbilirubinemia in Chinese neonates.
Collapse
|
39
|
Wong F, Boo N, Othman A. Risk factors associated with unconjugated neonatal hyperbilirubinemia in Malaysian neonates. J Trop Pediatr 2013; 59:280-5. [PMID: 23640907 DOI: 10.1093/tropej/fmt023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To investigate the risk factors associated with neonatal hyperbilirubinemia in Malaysian neonates. METHODS A prospective study was conducted to investigate the effects of glucose-6-phosphate dehydrogenase (G6PD) mutation, variant uridine diphosphate glucuronosyltransferase UGT1A1 gene and hepatic organic anion transporter protein (OATP2) gene on a group of neonates. Hyperbilirubinemia was defined as a total serum bilirubin level of ≥250 µmol/l. RESULTS Of 318 neonates, 52 (16.4%) had hyperbilirubinemia. The incidence of G6PD mutation was 5.4% (15/280) among these infants. The incidence of G6PD mutation was significantly higher in the male neonates with hyperbilirubinemia (7.8%) when compared with the normal male neonates without hyperbilirubinemia (1.8%; p = 0.03). Logistic regression analysis showed that the significant risk factors for neonatal hyperbilirubinemia were Malay ethnicity [adjusted odds ratio (OR), 2.77; 95% confidence interval (CI): 1.31-5.86; p = 0.007] and G6PD mutation (adjusted OR, 3.29; 95% CI: 1.06-10.1820; p = 0.039). The gender, birth weight and gestation age of neonates, variant c.211G > A and variant of OATP2 gene were not significant. CONCLUSIONS Neonates with Malay ethnicity and G6PD mutation were at risk for hyperbilirubinemia.
Collapse
Affiliation(s)
- Feiliang Wong
- Unit NICU, Department of Pediatrics, UKMMC, 56000, Kuala Lumpur, Malaysia.
| | | | | |
Collapse
|
40
|
Christensen RD, Yaish HM, Nussenzveig RH, Reading NS, Agarwal AM, Eggert LD, Prchal JT. Acute kernicterus in a neonate with O/B blood group incompatibility and a mutation in SLC4A1. Pediatrics 2013; 132:e531-4. [PMID: 23878048 DOI: 10.1542/peds.2012-3799] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We cared for a term female newborn, who at 108 hours of age, with a total serum bilirubin of 15.4 mg/dL, was discharged from the hospital on home phototherapy. At a return appointment 44 hours later, her total serum bilirubin was 41.7 mg/dL and signs of acute kernicterus were present. Maternal/fetal blood group O/B incompatibility was identified, with a negative direct antiglobulin test, which was positive on retesting. She had abundant spherocytes on blood smear, and these persisted at follow-up, but neither parent had spherocytes identified. A heterozygous SLC4A1(E508K) mutation (gene encoding erythrocyte membrane protein band 3) was found, and in silico predicted to result in damaged erythrocyte cytoskeletal protein function. No mutations were identified in other red cell cytoskeleton genes (ANK1, SPTA1, SPTB, EPB41, EPB42) and the UGT1A1 promoter region was normal. Neurologic follow-up at 2 and 4 months showed developmental delays consistent with mild kernicterus.
Collapse
Affiliation(s)
- Robert D Christensen
- Women and Newborns Program, Intermountain Healthcare, Salt Lake City, Utah 84403, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Christensen RD, Lambert DK, Henry E, Eggert LD, Yaish HM, Reading NS, Prchal JT. Unexplained extreme hyperbilirubinemia among neonates in a multihospital healthcare system. Blood Cells Mol Dis 2012; 50:105-9. [PMID: 23123126 DOI: 10.1016/j.bcmd.2012.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 10/17/2012] [Indexed: 10/27/2022]
Abstract
We report a series of neonates who developed a total serum bilirubin (TSB) >20mg/dL during a recent ten-year period in a multihospital healthcare system. The incidence of a TSB >20mg/dL fell after instituting a pre-hospital discharge bilirubin screening program in 2003/2004 (91.3 cases/10,000 births before vs. 72.4/10,000 after), but the incidence has subsequently remained unchanged. No specific cause for the hyperbilirubinemia was identified in 66% of (n=32) cases with a TSB >30 mg/dL or in 76% of (n=112) cases with a TSB 25.0-29.9 mg/dL. We hypothesized that hemolysis was a common contributing mechanism, but our review of hospital records indicated that in most instances these infants were not evaluated sufficiently to test this hypothesis. Records review showed maternal and neonatal blood types and direct antiglobulin testing were performed in >95% cases, but rarely were other tests for hemolysis obtained. In the ten-year period reviewed there were zero instances where erythrocyte morphology from a blood film examination or Heinz body evaluation by a pediatric hematologist or pathologist were performed. In 3% of cases pyruvate kinase was tested, 3% were evaluated by hemoglobin electrophoresis, 3% had a haptoglobin measurement, and 16% were tested for G6PD deficiency. Thus, determining the cause for hyperbilirubinemia in neonates remains a problem at Intermountain Healthcare and, we submit, elsewhere. As a result, the majority of infants with a TSB >25mg/dL have no specific causation identified. We speculate that most of these cases involve hemolysis and that the etiology could be identified if searched for more systematically. With this in mind, we propose a "consistent approach" to evaluating the cause(s) of hyperbilirubinemia among neonates with a TSB >25mg/dL.
Collapse
Affiliation(s)
- Robert D Christensen
- Women and Newborn's Clinical Program, Intermountain Healthcare, Salt Lake City, UT, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Chen S, Yueh MF, Evans RM, Tukey RH. Pregnane-x-receptor controls hepatic glucuronidation during pregnancy and neonatal development in humanized UGT1 mice. Hepatology 2012; 56:658-67. [PMID: 22371261 PMCID: PMC3383890 DOI: 10.1002/hep.25671] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 02/13/2012] [Indexed: 02/01/2023]
Abstract
UNLABELLED In humanized UDP glucuronosyltransferase-1 (hUGT1) mice that express the entire UGT1 locus, the maternal hepatic UGT1A genes are dramatically induced 12-14 days after conception. Steroid induction of the UGT1A1 gene indicates that xenobiotic sensors, such as the pregnane X receptor (PXR) and constitutive androstane receptor (CAR), may underlie the induction process. In contrast, neonatal hUGT1 mice display severe hyperbilirubinemia, with limited expression of the UGT1A genes. This study identifies PXR as both a positive and negative regulator of the UGT1A1 gene. Pregnancy hormones, in particular the glucocorticoids, target PXR as a positive regulator of human glucuronidation. Employing reverse genetics, where PXR has been genetically deleted, hUGT1/Pxr(-/-) mice show limited induction of the liver UGT1A genes during pregnancy, whereas the exact opposite occurs in newborn mice. Neonatal hUGT1 mice show delayed expression of hepatic UGT1A1 and are severely hyperbilirubinemic. However, in hUGT1/Pxr(-/-) mice, hyperbilirubinemia is greatly reduced due to induction of hepatic UGT1A1. Thus, PXR serves to repress UGT1A1 gene expression during development. Transcriptional silencing of the UGT1A1 gene was relieved in neonatal hUGT1 hepatocytes through interruption of PXR by small interfering RNA. CONCLUSION PXR is a key regulator of pregnancy induced glucuronidation capacity in addition to modulating the severity of neonatal jaundice.
Collapse
Affiliation(s)
- Shujuan Chen
- Department of Pharmacology, Laboratory of Environmental Toxicology, University of CaliforniaSan Diego, La Jolla, CA
| | - Mei-Fei Yueh
- Department of Pharmacology, Laboratory of Environmental Toxicology, University of CaliforniaSan Diego, La Jolla, CA
| | - Ronald M Evans
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute of Biological StudiesLa Jolla, CA
| | - Robert H Tukey
- Department of Pharmacology, Laboratory of Environmental Toxicology, University of CaliforniaSan Diego, La Jolla, CA,Department of Chemistry & Biochemistry, Laboratory of Environmental Toxicology, University of CaliforniaSan Diego, La Jolla, CA
| |
Collapse
|
43
|
UGT1A1, SLCO1B1, and SLCO1B3 polymorphisms vs. neonatal hyperbilirubinemia: is there an association? Pediatr Res 2012; 72:169-73. [PMID: 22580719 DOI: 10.1038/pr.2012.60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Jaundice is a physiological phenomenon; however, severe hyperbilirubinemia occurs in only 5 to 6% of the healthy newborn population. It has been suggested that genetic variation could enhance the risk of hyperbilirubinemia when coexpressed with other icterogenic conditions. METHODS The study included newborns with a gestational age of greater than 35 wk and weights greater than 2,000 g with indications for phototherapy. The polymorphisms from UGT1A1 (rs8175347), SLCO1B1 (rs4149056 and rs2306283), and SLCO1B3 (rs17680137 and rs2117032) were analyzed by capillary electrophoresis and hydrolysis probes. RESULTS A total of 167 hyperbilirubinemic infants and 247 control subjects were enrolled. The gender, ABO incompatibility, birth weight, and gestational age differed between the groups, but the allelic and genotypic frequency of the polymorphisms from SLCO1B genes did not. In logistic regression, the ABO incompatibility, gestational age, and polymorphic T allele of rs2117032 remained in the model. The presence of this polymorphism seemed to provide protection from hyperbilirubinemia. The individuals who were homozygous for the G allele of rs2306283 and who were glucose 6-phosphate-dehydrogenase deficient were more frequent among the cases. CONCLUSION Although genetic variation accounts for a good part of this condition, the association between different polymorphisms and environmental factors has yet to be explained.
Collapse
|
44
|
Abstract
After completing this article, readers should be able to: 1. List the risk factors for severe hyperbilirubinemia. 2. Distinguish between physiologic jaundice and pathologic jaundice of the newborn. 3. Recognize the clinical manifestations of acute bilirubin encephalopathy and the permanent clinical sequelae of kernicterus.4. Describe the evaluation of hyperbilirubinemia from birth through 3 months of age. 5. Manage neonatal hyperbilirubinemia, including referral to the neonatal intensive care unit for exchange transfusion.
Collapse
Affiliation(s)
- Bryon J Lauer
- Drexel University College of Medicine, St. Christopher's Hospital for Children, Philadelphia, PA, USA
| | | |
Collapse
|
45
|
Long J, Zhang S, Fang X, Luo Y, Liu J. Association of neonatal hyperbilirubinemia with uridine diphosphate-glucuronosyltransferase 1A1 gene polymorphisms: meta-analysis. Pediatr Int 2011; 53:530-40. [PMID: 21342357 DOI: 10.1111/j.1442-200x.2011.03337.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Recent reports have suggested that genetic factors, including mutations in the coding region or promoter of uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1) may increase the risk of development of neonatal hyperbilirubinemia, but the relationship has not been evaluated on systematic review or meta-analysis. METHODS A meta-analysis of observational studies reporting effect estimates and 95% confidence intervals (95%CI) was conducted on the association between UGT1A1 polymorphisms and neonatal hyperbilirubinemia. RESULTS A total of 27 eligible studies were identified. In total, 17 studies focused on the association of neonatal hyperbilirubinemia with UGT1A1 Gly71Arg polymorphisms, which indicated that these polymorphisms were associated with an increased risk of neonatal hyperbilirubinemia (A/A+G/A vs G/G: odds ratio [OR], 2.70; P= 0.00; 95%CI: 2.22-3.29; I(2) = 0.0%; P(heterogeneity) = 0.55). Subgroup analyses by ethnicity validated this correlation in Asian, but not in Caucasian, populations (OR, 1.74; P= 0.10; 95%CI: 0.90-3.35; I(2) = 0.00%; P(heterogeneity) = 0.67). Furthermore, 18 studies focused on the association of neonatal hyperbilirubinemia with UGT1A1 TATA promoter polymorphisms. These studies concluded that TATA promoter variants were not associated with an increased risk of neonatal hyperbilirubinemia (7/7 + 6/7 vs 6/6: OR, 1.13; P= 0.23; 95%CI: 0.93-1.37; I(2) = 80.0%; P(heterogeneity) = 0.00). CONCLUSION UGT1A1 Gly71Arg polymorphisms are a risk factor for developing neonatal hyperbilirubinemia in Asian, but not Caucasian, subjects. UGT1A1 TATA promoter polymorphisms were not associated with an increased risk of neonatal hyperbilirubinemia in Asian subjects, but results from the Caucasian population were conflicting and require further epidemiological investigation.
Collapse
Affiliation(s)
- Jun Long
- Department of Pediatrics, Fifth People's Hospital of Shenzhen, Shenzhen, China
| | | | | | | | | |
Collapse
|
46
|
Long J, Zhang S, Fang X, Luo Y, Liu J. Neonatal hyperbilirubinemia and Gly71Arg mutation of UGT1A1 gene: a Chinese case-control study followed by systematic review of existing evidence. Acta Paediatr 2011; 100:966-71. [PMID: 21272068 DOI: 10.1111/j.1651-2227.2011.02176.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
AIM To determine whether the UDP-glucuronosyltransferase 1A1 gene (UGT1A1) Gly71Arg (211G>A) mutation is associated with neonatal hyperbilirubinemia. METHODS The study consisted of two parts. The case-control study included 112 hyperbilirubinemic infants and 105 control subjects from the Fifth People's Hospital of Shenzhen. Polymerase chain reaction, restriction fragment length polymorphisms and agarose gel electrophoresis techniques were used to detect the UGT1A1 211G>A mutation. Meta-analyses was performed to assess the association between neonatal hyperbilirubinemia and UGT1A1 211G>A. RESULTS Our case-control study revealed that the likelihood of developing neonatal hyperbilirubinemia was 2.65 times higher in the infants with the A allele in the UGT1A1 211G>A than in the infants with the G allele (95% CI, 1.60-4.39). Meta-analyses (including data from our study) revealed that UGT1A1 211G>A is associated with an increased risk of neonatal hyperbilirubinemia [odds ratio (OR), 2.37; 95% CI, 2.05-2.74]. In the subgroup analyses based on ethnicity, significantly elevated risks were found in Asian populations (OR, 2.45; 95% CI, 2.10-2.84), but no significant associations were present in Caucasian populations (OR, 1.54; 95% CI, 0.87-2.75). CONCLUSION The UGT1A1 211G>A mutation is associated with neonatal hyperbilirubinemia in Asians, but not in Caucasians.
Collapse
Affiliation(s)
- Jun Long
- Department of Pediatrics, The Fifth People's Hospital of Shenzhen, Shenzhen, China
| | | | | | | | | |
Collapse
|
47
|
McKay TR, Rahim AA, Buckley SM, Ward NJ, Chan JK, Howe SJ, Waddington SN. Perinatal gene transfer to the liver. Curr Pharm Des 2011; 17:2528-41. [PMID: 21774770 PMCID: PMC3182410 DOI: 10.2174/138161211797247541] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 06/28/2011] [Indexed: 01/08/2023]
Abstract
The liver acts as a host to many functions hence raising the possibility that any one may be compromised by a single gene defect. Inherited or de novo mutations in these genes may result in relatively mild diseases or be so devastating that death within the first weeks or months of life is inevitable. Some diseases can be managed using conventional medicines whereas others are, as yet, untreatable. In this review we consider the application of early intervention gene therapy in neonatal and fetal preclinical studies. We appraise the tools of this technology, including lentivirus, adenovirus and adeno-associated virus (AAV)-based vectors. We highlight the application of these for a range of diseases including hemophilia, urea cycle disorders such as ornithine transcarbamylase deficiency, organic acidemias, lysosomal storage diseases including mucopolysaccharidoses, glycogen storage diseases and bile metabolism. We conclude by assessing the advantages and disadvantages associated with fetal and neonatal liver gene transfer.
Collapse
Affiliation(s)
- Tristan R McKay
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Ahad A Rahim
- Institute for Women’s Health, University College London, London, UK
| | | | - Natalie J Ward
- Institute for Women’s Health, University College London, London, UK
| | - Jerry K.Y Chan
- Experimental Fetal Medicine Group, National University of Singapore, Singapore
| | - Steven J Howe
- Institute of Child Health, University College London, London, UK
| | | |
Collapse
|
48
|
de Azevedo LA, Santin AP, Wagner SC, da Fontoura Zaleski C, Bock H, Saraiva-Pereira ML, de Castro SM. Prevalence ofUGT1A1Gene Polymorphism in Patients with Hemolytic Anemia in Southern Brazil. Genet Test Mol Biomarkers 2011; 15:107-10. [DOI: 10.1089/gtmb.2010.0060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
| | - Ana Paula Santin
- Department of Analysis, Pharmacy School, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Sandrine Comparsi Wagner
- Department of Genetics, Biosciences Institute, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Hugo Bock
- Laboratory of Genetics Identification, Experimental Research Centre, HCPA, Porto Alegre, Brazil
| | - Maria Luiza Saraiva-Pereira
- Laboratory of Genetics Identification, Experimental Research Centre, HCPA, Porto Alegre, Brazil
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Simone Martins de Castro
- Department of Analysis, Pharmacy School, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|