1
|
Bazarbayeva A, Manzhuova L, Svyatova G, Berezina G, Sarsekbayeva F, Kamalova D. Association of B-Lineage Lymphoblastic Leukaemia Gene Polymorphisms with Poor Prognostic Features. Asian Pac J Cancer Prev 2024; 25:4339-4349. [PMID: 39733427 PMCID: PMC12008353 DOI: 10.31557/apjcp.2024.25.12.4339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Indexed: 12/31/2024] Open
Abstract
OBJECTIVE Of this study was to analyse the correlation of gene polymorphisms with clinical and laboratory data of paediatric patients with B-lineage acute lymphoblastic leukaemia with prognostically unfavourable features. METHODS A study of 200 children with B-lineage acute lymphoblastic leukaemia (B-ALL) treated with polychemotherapy programmes was conducted. Analysis by sex revealed a statistically insignificant predominance of the group of boys over girls (54%). The mean age of the subjects was 9.3±0.2 years. Genotyping of polymorphic loci was performed using TaqMan method of single site-specific amplification and genotyping. The data of patients with initial prognostically unfavourable clinical and laboratory data in the form of initial leukocytosis from 50 to 99 thousand - 10 (5%), over 100 thousand - 16 (8%), initial CNS lesion in the form of neuroleukaemia - 5 (2.5%), initial splenomegaly more than 6 cm - 12 (6%); patients with poor response to therapy, having absolute number of blast cells in peripheral blood over 1,000 on day 8 of treatment according to the protocol (response to prednisolone prophase) - 13 (7%), with unsatisfactory response to treatment on Day 15 - 40 patients (20%) and on Day 33 - 4 children (2%); also patients who developed relapse of the disease - 17 (9%). RESULTS According to the findings, of all 24 gene variants, 13 variants (54%), namely, HLA - rs6457327, TNF - rs1800630 and rs2229094, GATA3 - rs3824662, TP53 - rs1042522, CASP9 - rs4661636, CASP8 - rs10505477, CEBPE - rs2239633; PIP4K2A - rs7088318, CASC8 - rs10505477, IRF4 - rs87207, CYP1A1 - rs4646903 and rs7089424 of ARID5B gene were found to be associated with B-ALL and unfavourable prognostic features. CONCLUSIONS The findings of this study revealed significant associations of polymorphic genetic variants, which may serve as a basis for the development of effective methods for predicting the risk of relapse development and the timeliness of intensification of B-ALL treatment. Prompt genetic counselling of children with identified unfavourable genotypes of the investigated gene polymorphisms will make it possible to predict the development of relapse, resistance and/or poor response to B-ALL treatment, and to propose an individual strategy for monitoring children's health in the short and long term.
Collapse
Affiliation(s)
- Aigul Bazarbayeva
- Department of Science and Postgraduate Education, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Republic of Kazakhstan.
| | - Lyazat Manzhuova
- Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Republic of Kazakhstan.
| | - Gulnara Svyatova
- Republican Medical and Genetic Counselling Centre, Scientific Center for Obstetrics, Gynecology and Perinatology, Almaty, Republic of Kazakhstan.
| | - Galina Berezina
- Department of Strategic Development and Science, Scientific Center for Obstetrics, Gynecology and Perinatology, Almaty, Republic of Kazakhstan.
| | - Farida Sarsekbayeva
- Department of Biostatistics, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Republic of Kazakhstan.
| | - Diana Kamalova
- Department of Science and Postgraduate Education, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Republic of Kazakhstan.
| |
Collapse
|
2
|
Farasani A. Association of genetic predisposition studies in CYP1A1 polymorphism studies in acute myeloid leukemia. Saudi J Biol Sci 2024; 31:103917. [PMID: 38283773 PMCID: PMC10821586 DOI: 10.1016/j.sjbs.2023.103917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/30/2024] Open
Abstract
Cytochrome P450 Family 1 Subfamily A Member 1 (CYP1A1) gene is one of the sub-members of CYP450 family member and it encodes with the families of drug metabolizing enzyme families along with the cancers and leukemias. Among leukemias, AML is considered to be one of the important leukemia which attack the older adults. The aim of this study is to explore the role of A4889G polymorphism in CYP1A1 gene in acute myeloid leukemia (AML) in the Saudi population. This study was designed as an experimental case-control study in which 100 AML cases and 100 controls were selected. This in vivo study was carried out using genomic DNA extraction, polymerase chain reaction and agarose gel electrophoresis and then BsrDI restriction enzyme to digest the A4889G polymorphism of the PCR products. In this study, 200 subjects were digested and based on the appearance of the bands, genotypes were categorized. The attained data was used to calculate the clinical details as well as genotype analysis. The study results confirmed AG genotype (OR = 3.23, CI = 1.60-6.55, p = 0.0008), AG + GG (OR = 3.47, CI = 1.76-6.86, p = 0.0002) and GG + AA (OR = 12.47, CI = 6.18-15.17, p < 0.0001) and G vs A (OR = 3.15, CI = 1.71-5.81, p = 0.0001) were associated in AML cases. In conclusion, we confirm that A4889G polymorphism is associated with AML in the Saudi population.
Collapse
Affiliation(s)
- Abdullah Farasani
- Biomedical Research Unit, Medical Center, Jazan University, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
3
|
Crippa V, Malighetti F, Villa M, Graudenzi A, Piazza R, Mologni L, Ramazzotti D. Characterization of cancer subtypes associated with clinical outcomes by multi-omics integrative clustering. Comput Biol Med 2023; 162:107064. [PMID: 37267828 DOI: 10.1016/j.compbiomed.2023.107064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/03/2023] [Accepted: 05/27/2023] [Indexed: 06/04/2023]
Abstract
Cancer patients show heterogeneous phenotypes and very different outcomes and responses even to common treatments, such as standard chemotherapy. This state-of-affairs has motivated the need for the comprehensive characterization of cancer phenotypes and fueled the generation of large omics datasets, comprising multiple omics data reported for the same patients, which might now allow us to start deciphering cancer heterogeneity and implement personalized therapeutic strategies. In this work, we performed the analysis of four cancer types obtained from the latest efforts by The Cancer Genome Atlas, for which seven distinct omics data were available for each patient, in addition to curated clinical outcomes. We performed a uniform pipeline for raw data preprocessing and adopted the Cancer Integration via MultIkernel LeaRning (CIMLR) integrative clustering method to extract cancer subtypes. We then systematically review the discovered clusters for the considered cancer types, highlighting novel associations between the different omics and prognosis.
Collapse
Affiliation(s)
- Valentina Crippa
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.
| | - Federica Malighetti
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.
| | - Matteo Villa
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.
| | - Alex Graudenzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milano, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.
| | - Daniele Ramazzotti
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.
| |
Collapse
|
4
|
Shapoo NS, Masood A, Bhat JR, Bhatia AS, Shah IA, Ganai BA. CYP2D6 rs35742686 and rs3892097 Gene Polymorphisms and Childhood Acute Lymphoblastic Leukemia: Relation to Disease Susceptibility in Kashmiri Children. J Pediatr Genet 2022; 11:213-220. [DOI: 10.1055/s-0041-1723975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/17/2020] [Indexed: 10/22/2022]
Abstract
Abstract
CYP2D6 is one of the most widely investigated CYPs in relation to gene polymorphism. This study analyzed the relationship between CYP2D6 rs35742686 and rs3892097 single-nucleotide polymorphisms (SNPs) and potential risk factors in the development of acute lymphoblastic leukemia (ALL) in Kashmiri children. We recruited 300 cases and 600 controls for genotyping and risk factors assessment. Genotypes of rs35742686 and rs3892097 were analyzed by polymerase chain reaction-restriction fragment length polymorphism method. CYP2D6 expression analysis was done by quantitative reverse transcription polymerase chain reaction in ALL cases. Conditional logistic regression models were used to calculate odds ratios (OR) and 95% confidence intervals (CI). High risk of ALL was observed in cases who carried the mutant genotypes of rs35742686 (OR = 18.15; 95% CI = 4.13–79.66, p < 0.0001) or rs3892097 (OR = 24.06; 95% CI = 10.23–56.53, p < 0.0001). Significant interaction was observed between rs35742686 and rs3892097 SNPs (P interaction = 0.001). The risk associated with the variant genotypes of rs35742686 and rs3892097 was retained in the cases whose fathers were smokers or had maternal X-ray exposure (p < 0.001). Relative messenger ribonucleic acid expression across genotypes was significantly decreased in cases carrying rs357426863 (*3/*3) (n-fold = 0.37 ± 0.156, p < 0.0079) and rs3892097 SNPs (*4/*4) (n-fold = 0.02 ± 0.0075, p < 0.0001) suggesting these two events are independent in ALL cases. The study concluded that rs35742686 and rs3892097 SNPs are significantly associated with ALL risk in Kashmiri children.
Collapse
Affiliation(s)
- Nidha Sadiq Shapoo
- Department of Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Akbar Masood
- Department of Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Javid R. Bhat
- Department of Clinical Haematology, Sher-e-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - A. S. Bhatia
- Department of Biochemistry, Government Medical College, Jammu, Jammu and Kashmir, India
| | - Idrees A. Shah
- Department of Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Bashir A. Ganai
- Centre of Research for Development, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
5
|
Hu T, Zhou G, Li W. Association Between the Individual and Combined Effects of the GSTM1 and GSTT1 Polymorphisms and Risk of Leukemia: A Meta-Analysis. Front Genet 2022; 13:898937. [PMID: 35938012 PMCID: PMC9355274 DOI: 10.3389/fgene.2022.898937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Fourteen meta-analyses reported the individual effects of the GSTM1 and GSTT1 polymorphisms on leukemia risk. However, over 40 studies were not included in previously published meta-analyses. Moreover, one key aspect was that previous meta-analyses did not conduct the false-positive test on the aforementioned issues. Furthermore, previous meta-analyses did not observe the combined effects of GSTM1 present/null and GSTT1 present/null polymorphism with leukemia risk. Therefore, we conducted the current study to further analyze these associations. Objectives: This study aimed to investigate the association between the individual and combined effects of the GSTM1 present/null and GSTT1 present/null polymorphisms and the risk of leukemia. Methods: A meta-analysis was performed applying Meta-analyses of Observational Studies in Epidemiology (MOOSE) guidelines. Moreover, false-positive report probability (FPRP) and Bayesian false discovery probability (BFDP) were applied to investigate the false-positive results. Results: The individual GSTM1 and GSTT1 null genotypes and combined effects of the two genes were associated with a significantly increased leukemia risk in overall and several subgroup analyses, such as Asians, Caucasians, and so on. Then, further analysis was conducted using FPRP and BFDP. Significant associations were considered as "positive" results on the GSTM1 null genotype with leukemia risk in overall populations (FPRP < 0.001 and BFDP = 0.006), Asians (FPRP < 0.001 and BFDP < 0.001), and East Asian population (FPRP < 0.001 and BFDP = 0.002). For the GSTT1 null genotype, significant associations were regarded "positive" results in overall populations, acute myeloid leukemia (AML), Asians, and East Asian population. For the combined effects of the GSTM1 and GSTT1 polymorphisms, significant associations were also considered "positive" results in the overall analysis of Asians, Indians, and East Asian population. Conclusion: This study strongly indicates that the individual GSTM1 and GSTT1 null genotypes and combined effects of the two genes are associated with increased leukemia risk in Asians, especially in the East Asian population; the GSTT1 null genotype is associated with increased AML risk; the combined effects of the two genes are associated with increased leukemia risk in Indians.
Collapse
Affiliation(s)
- Ting Hu
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Guozhong Zhou
- Department of Cardiology, Pingxiang People’s Hospital, Pingxiang, China
| | - Wenjin Li
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| |
Collapse
|
6
|
Association Between SHMT1 rs1979277 Polymorphism and Risk of Acute Lymphoblastic Leukemia: A Systematic Review and Meta-analysis. J Pediatr Hematol Oncol 2022; 44:e616-e622. [PMID: 33974584 DOI: 10.1097/mph.0000000000002173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/06/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aim of this study was to explore the potential association the cytosolic serine hydroxy methyltransferase (SHMT1) rs1979277 polymorphism and the risk of acute lymphoblastic leukemia (ALL). MATERIALS AND METHODS Comprehensive search of Web of Science, PubMed, Ovid, Cochrane Library, Embase, China National Knowledge Infrastructure (CNKI), and China Biomedical Literature Database electronic database, was performed to identify relevant studies published throughout April 30, 2019. The heterogeneity in the study was judged by the I2 and P-values, and then the random ratio or fixed effect was used to calculate the pooled odds ratios (OR) based on the presence or absence of heterogeneity. Sensitivity analysis is used to estimate the impact of individual studies on aggregate estimates. The publication bias of the study was tested using a funnel plot and an Egger regression. RESULTS Nine studies with a total of 6492 participants (2971 patients; 3521 controls) were included in this meta-analysis. We found that SHMT1 rs1979277 polymorphism was not significantly associated with the risk of ALL in the dominant model: CC versus CT+TT (OR=0.84, 95% confidence interval [CI]: 0.46-1.54, P=0.57), recessive model: CC+CT versus TT (OR=0.81, 95% CI: 0.44-1.49, P=0.50) and allele model: C versus T (OR=0.84, 95% CI: 0.52-1.35, P=0.48). In subgroup analysis by ethnicity, no significant association were found in dominant, recessive and allele models in both Caucasian and Asian populations. CONCLUSION Our study indicated that the SHMT1 rs1979277 polymorphism was not associated with the risk of susceptibility to ALL.
Collapse
|
7
|
Baba SM, Pandith AA, Shah ZA, Geelani SA, Bhat J, Gul A, Guru SA, El-Serehy HA, Koul AM, Mansoor S. GSTT1 null and rs156697 Polymorphism in GSTO2 Influence the Risk and Therapeutic Outcome of B-Acute Lymphoblastic Leukemia Patients. Front Oncol 2021; 11:714421. [PMID: 34722260 PMCID: PMC8552530 DOI: 10.3389/fonc.2021.714421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Glutathione S-transferase (GST) gene deletion or polymorphic sequence variations lead to decreased enzyme activity that influences susceptibility and response to chemotherapy in acute lymphoblastic leukemia (ALL). This case-control study investigated the association of GST gene polymorphisms with the etiology and therapeutic outcome of B-ALL among Kashmiri population. METHODS A total of 300 individuals including 150 newly diagnosed B-ALL patients and an equal number of age and gender matched controls were genotyped for five GST gene polymorphisms by polymerase chain reaction-restriction fragment length polymorphism technique (PCR-RFLP) and multiplex PCR techniques. RESULTS Higher frequency of GSTT1 null, GSTO2-AG, and GSTO2-GG genotypes was observed in ALL cases compared to controls that associated significantly with ALL risk (GSTT1 null: OR = 2.93, p = 0.0001; GSTO2-AG: OR = 2.58, p = 0.01; GSTO2-GG: OR = 3.13, p = 0.01). GSTM1, GSTP1, and GSTO1 SNPs showed no significant association (p > 0.05). Combined genotype analysis revealed significant association of GSTT1 null/GSTM1 null (OR = 4.11, p = 0.011) and GSTT1 null/GSTP1-AG (OR = 4.93, p = 0.0003) with B-ALL susceptibility. Haplotype analysis of rs4925 and rs156697 revealed that carriers of CG haplotype had increased risk of B-ALL (p = 0.04). Kaplan-Meier plots revealed significantly inferior 3-year disease-free survival for GSTO2-GG carriers (p = 0.002). Multivariate analysis confirmed GSTO2-GG as an independent poor prognostic factor for DFS (HR = 4.5, p = 0.034). Among combined genotypes, only GSTT1 null/GSTP1-AG associated significantly with poorer DFS rates (p = 0.032). CONCLUSION This study demonstrated that GSTT1 null individually or in combination with GSTM1null and GSTP1-AG genotypes associated with increased B-ALL risk. Also, rs156697 variant genotypes (AG and GG) associated with B-ALL, whereas the GG genotype of rs156697 influenced the treatment outcome.
Collapse
Affiliation(s)
- Shahid M. Baba
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | | | - Zafar A. Shah
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | | | - Javid R. Bhat
- Advanced Centre for Human Genetics, SKIMS, Srinagar, India
| | - Ayaz Gul
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, India
| | - Sameer A. Guru
- Department of Developmental and System Biology, Lurie Children’s Hospital Northwest University, Chicago, IL, United States
| | - Hamed A. El-Serehy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abid M. Koul
- Advanced Centre for Human Genetics, SKIMS, Srinagar, India
| | - Sheikh Mansoor
- Advanced Centre for Human Genetics, SKIMS, Srinagar, India
| |
Collapse
|
8
|
Kampouraki E, Lourou M, Zervou MI, Ampazoglou ED, Yachnakis E, Katzilakis N, Goulielmos GN, Stiakaki E. Role of CXCL12, TP53 and CYP1A1 gene polymorphisms in susceptibility to pediatric acute lymphoblastic leukemia. Oncol Lett 2021; 22:659. [PMID: 34386081 DOI: 10.3892/ol.2021.12920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/26/2021] [Indexed: 01/02/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common type of childhood leukemia and represents one third of all pediatric malignancies. Epidemiological studies have shown that various genetic factors play a crucial role in leukemogenesis. Recent genetic association studies on cancer risk have focused on the effects of single-nucleotide polymorphisms (SNPs) in genes that regulate inflammation and tumor suppression, such as chemokines, TP53 and cytochrome P450s (CYPs). Genetic polymorphisms in the 3' untranslated region of the C-X-C motif chemokine ligand 12 (CXCL12; rs1801157) and TP53 (rs1042522) genes have been suggested to influence the risk of ALL in children, while other studies have indicated an association between the CYP1 subfamily A member 1 (CYP1A1)*2C (rs1048943) allele and leukemia risk. The aim of the present study was to investigate the possible association of rs1801157 (CXCL12), rs1042522 (TP53) and rs1048943 (CYP1A1*2C) SNPs with an increased susceptibility of developing ALL. These SNPs were analyzed in 86 children or adolescent patients with ALL and 125 control subjects by PCR-restriction fragment length polymorphism and allelic-specific chain reaction techniques. A higher frequency of CYP1A1*2C heterozygotes and TP53 rare homozygotes, which include the proline (Pro)/Pro genotype, was observed among children with ALL and control subjects, whereas no significant differences were observed for the CXCL12 SNP. Furthermore, the analysis of various allelic combinations of the aforementioned gene polymorphisms demonstrated a markedly increased risk of developing ALL in children. In conclusion, the present study demonstrated that there was a strong association between CYP1A1*2C heterozygotes, as well as the TP53 Pro/Pro genotype, and an increased susceptibility for pediatric ALL in Caucasians.
Collapse
Affiliation(s)
- Eleni Kampouraki
- Department of Pediatric Hematology-Oncology, University Hospital of Heraklion and Laboratory of Blood Diseases and Childhood Cancer Biology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Marilena Lourou
- Department of Pediatric Hematology-Oncology, University Hospital of Heraklion and Laboratory of Blood Diseases and Childhood Cancer Biology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Maria I Zervou
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, Medical School of Crete, University of Crete, 71003 Heraklion, Greece
| | - Evangelia-Dimitra Ampazoglou
- Department of Pediatric Hematology-Oncology, University Hospital of Heraklion and Laboratory of Blood Diseases and Childhood Cancer Biology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Emmanuel Yachnakis
- Laboratory of Bio-Medical Data Analyses, Digital Applications and Interdisciplinary Approaches, University of Crete, 71003 Heraklion, Greece
| | - Nikolaos Katzilakis
- Department of Pediatric Hematology-Oncology, University Hospital of Heraklion and Laboratory of Blood Diseases and Childhood Cancer Biology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - George N Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, Medical School of Crete, University of Crete, 71003 Heraklion, Greece
| | - Eftichia Stiakaki
- Department of Pediatric Hematology-Oncology, University Hospital of Heraklion and Laboratory of Blood Diseases and Childhood Cancer Biology, Medical School, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
9
|
Greaves M, Cazzaniga V, Ford A. Can we prevent childhood Leukaemia? Leukemia 2021; 35:1258-1264. [PMID: 33833382 PMCID: PMC8102184 DOI: 10.1038/s41375-021-01211-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/03/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| | - Valeria Cazzaniga
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Anthony Ford
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| |
Collapse
|
10
|
Nam SW, Lee KS, Yang JW, Ko Y, Eisenhut M, Lee KH, Shin JI, Kronbichler A. Understanding the genetics of systemic lupus erythematosus using Bayesian statistics and gene network analysis. Clin Exp Pediatr 2021; 64:208-222. [PMID: 32683804 PMCID: PMC8103040 DOI: 10.3345/cep.2020.00633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
The publication of genetic epidemiology meta-analyses has increased rapidly, but it has been suggested that many of the statistically significant results are false positive. In addition, most such meta-analyses have been redundant, duplicate, and erroneous, leading to research waste. In addition, since most claimed candidate gene associations were false-positives, correctly interpreting the published results is important. In this review, we emphasize the importance of interpreting the results of genetic epidemiology meta-analyses using Bayesian statistics and gene network analysis, which could be applied in other diseases.
Collapse
Affiliation(s)
- Seoung Wan Nam
- Department of Rheumatology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kwang Seob Lee
- Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Younhee Ko
- Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Korea
| | - Michael Eisenhut
- Department of Pediatrics, Luton & Dunstable University Hospital NHS Foundation Trust, Luton, UK
| | - Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea.,Division of Pediatric Nephrology, Severance Children's Hospital, Seoul, Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea.,Division of Pediatric Nephrology, Severance Children's Hospital, Seoul, Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea
| | - Andreas Kronbichler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Klco JM, Mullighan CG. Advances in germline predisposition to acute leukaemias and myeloid neoplasms. Nat Rev Cancer 2021; 21:122-137. [PMID: 33328584 PMCID: PMC8404376 DOI: 10.1038/s41568-020-00315-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Although much work has focused on the elucidation of somatic alterations that drive the development of acute leukaemias and other haematopoietic diseases, it has become increasingly recognized that germline mutations are common in many of these neoplasms. In this Review, we highlight the different genetic pathways impacted by germline mutations that can ultimately lead to the development of familial and sporadic haematological malignancies, including acute lymphoblastic leukaemia, acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS). Many of the genes disrupted by somatic mutations in these diseases (for example, TP53, RUNX1, IKZF1 and ETV6) are the same as those that harbour germline mutations in children and adolescents who develop these malignancies. Moreover, the presumption that familial leukaemias only present in childhood is no longer true, in large part due to the numerous studies demonstrating germline DDX41 mutations in adults with MDS and AML. Lastly, we highlight how different cooperating events can influence the ultimate phenotype in these different familial leukaemia syndromes.
Collapse
Affiliation(s)
- Jeffery M Klco
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Charles G Mullighan
- Department of Pathology and the Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
12
|
Plasma of Acute Lymphoblastic Leukemia Patients React to the Culture of a Mycovirus Containing Aspergillus flavus. J Pediatr Hematol Oncol 2020; 42:350-358. [PMID: 32576782 DOI: 10.1097/mph.0000000000001845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children and is also seen in adults. Currently, no plasma-based test for the detection of ALL is available. We have cultured the home of a patient with ALL and isolated a mycovirus containing Aspergillus flavus. This culture was subjected to electron microscopy, purification, and mass spectrometry. Using enzyme-linked immunosorbent assay technique, plasma of patients with ALL and long-term survivors of this disease were tested for antibodies, utilizing supernatant of the culture of this organism. The results were compared with 3 groups of controls, including healthy individuals, patients with sickle cell disease, and solid tumors. Using electron microscopy, the isolated A. flavus contained mycovirus particles. In chemical analysis, this organism did not produce any aflatoxin. Using an enzyme-linked immunosorbent assay technique, the supernatant of the culture of the mycovirus containing A. flavus could differentiate ALL patients from each group of controls (P<0.001). These studies provide a new technique for the detection of ALL and may add information for future research regarding leukemogenesis.
Collapse
|
13
|
Wang Y, Hu F, Li JY, Nie RC, Chen SL, Cai YY, Shu LL, Deng DJ, Xu JB, Liang Y. Systematic Construction and Validation of a Metabolic Risk Model for Prognostic Prediction in Acute Myelogenous Leukemia. Front Oncol 2020; 10:540. [PMID: 32373530 PMCID: PMC7186449 DOI: 10.3389/fonc.2020.00540] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/25/2020] [Indexed: 01/11/2023] Open
Abstract
Background: Acute myelogenous leukemia (AML) is a heterogeneous disease with recurrent gene mutations and variations in disease-associated gene expression, which may be useful for prognostic prediction. Methods: RNA matrix and clinical data of AML were downloaded from GEO, TCGA, and TARGET databases. Prognostic metabolic genes were identified by LASSO analysis to establish a metabolic model. Prognostic accuracy of the model was quantified by time-dependent receiver operating characteristic curves and the area under the curve (AUC). Survival analysis was performed by log-rank tests. Enriched pathways in different metabolic risk statuses were evaluated by gene set enrichment analyses (GSEA). Results: We identified nine genes to construct a prognostic model of shorter survival in the high-risk vs. low-risk group. The prognostic model showed good predictive efficacy, with AUCs for 5-year overall survival of 0.78 (0.73-0.83), 0.76 (0.62-0.89), and 0.66 (0.57-0.75) in the training, adult external, and pediatric external cohorts, respectively. Multivariable analysis demonstrated that the metabolic signature had independent prognostic value with hazard ratios of 2.75 (2.06-3.66), 1.89 (1.09-3.29), and 1.96 (1.00-3.84) in the training, adult external, and pediatric external cohorts, respectively. Combining metabolic signatures and classic prognostic factors improved 5-year overall survival prediction compared to the prediction by classic prognostic factors (p < 0.05). GSEA revealed that most pathways were metabolism-related, indicating potential mechanisms. Conclusion: We identified dysregulated metabolic features in AML and constructed a prognostic model to predict the survival of patients with AML.
Collapse
Affiliation(s)
- Yun Wang
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fang Hu
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jin-Yuan Li
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Run-Cong Nie
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Si-Liang Chen
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan-Yu Cai
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ling-Ling Shu
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - De-Jun Deng
- Department of Oncology and Hematology, Shenzhen Luohu District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Jing-Bo Xu
- Department of Hematology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yang Liang
- Sate key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
14
|
Jiang M, Guo X, Yuan L, Gao J, Huo L, Li Q. Killer cell immunoglobulin-like receptor gene cluster predisposes to susceptibility to B-cell acute lymphoblastic leukemia in Chinese children. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:536-542. [PMID: 32269692 PMCID: PMC7136999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/07/2020] [Indexed: 06/11/2023]
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is the most common malignancy in children. Killer cell immunoglobulin-like receptors (KIRs) are mainly expressed on natural killer (NK) cells and regulate killing of cancer cells. To investigate the possible association of KIR genes with B-ALL in Chinese children, we used polymerase chain reaction with sequence-specific primers (PCR-SSP) to determine the KIR genotypes of 137 B-ALL patients and 288 healthy children of Chinese Han origin. Herein we report no significant difference in the carrying frequency of individual KIR genes and haplotypes between patients and controls; however, individuals carrying C4Tx genotypes were more frequent in the B-ALL group compared with healthy controls (11.7% vs. 5.9%, P=0.038). In addition, the centromeric KIR gene cluster, KIR2DS2-2DL2-2DS3-2DL5, was significantly increased in the B-ALL group compared with healthy controls (13.9% vs. 7.3%, P=0.030). These data suggest that the C4Tx genotype and centromeric KIR gene cluster (KIR2DS2-2DL2-2DS3-2DL5) might predispose to susceptibility to B-ALL in Chinese children.
Collapse
Affiliation(s)
- Mingyan Jiang
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan UniversityChengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengdu, Sichuan Province, China
| | - Xia Guo
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan UniversityChengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengdu, Sichuan Province, China
| | - Lixing Yuan
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan UniversityChengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengdu, Sichuan Province, China
| | - Ju Gao
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan UniversityChengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengdu, Sichuan Province, China
| | - Li Huo
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan UniversityChengdu, Sichuan Province, China
- Department of Emergency, West China Second University Hospital, Sichuan UniversityChengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengdu, Sichuan Province, China
| | - Qiang Li
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan UniversityChengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengdu, Sichuan Province, China
| |
Collapse
|
15
|
Mosaad YM, Khashaba M, Darwish A, Darwish M, Elwassefy M, Abdelmabood S, Fawzy IM, Youssef LF, Elbasiouny RA. ARID5B rs10821936 and rs10994982 gene polymorphisms and acute lymphoblastic leukemia: relation to disease susceptibility and outcome. Pediatr Hematol Oncol 2019; 36:365-375. [PMID: 31424309 DOI: 10.1080/08880018.2019.1649333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
ARID5B rs10821936 and rs10994982 single nucleotide polymorphism (SNP) have been associated with the risk of acute lymphoblastic leukemia (ALL) in different ethnic populations. We investigated the association between the ARID5B rs10821936 C > T, rs10994982 A > G, and susceptibility to ALL in a cohort of Egyptian individuals and investigated their role in relation to disease outcome. Real-time PCR typing was done for ARID5B rs10821936 and rs10994982 SNPs for 128 pediatric ALL (pALL), 45 adult ALL (aALL), and 436 healthy controls. Significant risk associations were found between the C allele (p < 0.001, OR = 2.02), CC genotype (p < 0.001, OR = 2.72), CT genotype (p = 0.011, OR = 1.45) of ARID5B rs10821936 and pediatric ALL especially T-ALL and adult ALL (p < 0.05). The CA haplotype (C allele of rs10821936 + A allele of rs10994982) was associated with the risk of ALL either pediatric ALL or adult ALL (p < 0.001). In the studied Egyptian population, it can be concluded that the C allele, CC, and CT genotypes of ARID5B rs10821936 and the CA haplotype may be a susceptibility risk factor for pediatric and adult ALL. However, the SNPs of ARID5B rs10821936 and rs10994982 were not found to be strongly associated with ALL outcomes.
Collapse
Affiliation(s)
- Youssef M Mosaad
- Clinical Immunology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University , Mansoura , Egypt
| | - Mohamed Khashaba
- Neonatology Unit, Pediatric Department, Mansoura University Children's Hospital , Mansoura , Egypt
| | - Ahmad Darwish
- Hematology/Oncology Unit, Pediatric Department, Mansoura University Children's Hospital , Mansoura , Egypt
| | - Mohamed Darwish
- Clinical Immunology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University , Mansoura , Egypt
| | - Mona Elwassefy
- Clinical Immunology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University , Mansoura , Egypt
| | - Souzy Abdelmabood
- Hematology/Oncology Unit, Pediatric Department, Mansoura University Children's Hospital , Mansoura , Egypt
| | - Iman M Fawzy
- Laboratory Medicine Department, Mansoura Fever Hospital, Ministry of Health , Mansoura , Egypt
| | - Laila F Youssef
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University , Mansoura , Egypt
| | - Rehab A Elbasiouny
- Pediatric Department, Mansoura University Children's Hospital , Mansoura , Egypt
| |
Collapse
|
16
|
Gervasini G, Mota-Zamorano S. Clinical Implications of Methotrexate Pharmacogenetics in Childhood Acute Lymphoblastic Leukaemia. Curr Drug Metab 2019; 20:313-330. [DOI: 10.2174/1389200220666190130161758] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/11/2019] [Accepted: 01/19/2019] [Indexed: 12/14/2022]
Abstract
Background:In the past two decades, a great body of research has been published regarding the effects of genetic polymorphisms on methotrexate (MTX)-induced toxicity and efficacy. Of particular interest is the role of this compound in childhood acute lymphoblastic leukaemia (ALL), where it is a pivotal drug in the different treatment protocols, both at low and high doses. MTX acts on a variety of target enzymes in the folates cycle, as well as being transported out and into of the cell by several transmembrane proteins.Methods:We undertook a structured search of bibliographic databases for peer-reviewed research literature using a focused review question.Results:This review has intended to summarize the current knowledge concerning the clinical impact of polymorphisms in enzymes and transporters involved in MTX disposition and mechanism of action on paediatric patients with ALL.Conclusion:In this work, we describe why, in spite of the significant research efforts, pharmacogenetics findings in this setting have not yet found their way into routine clinical practice.
Collapse
Affiliation(s)
- Guillermo Gervasini
- Department of Medical & Surgical Therapeutics, Medical School, University of Extremadura, Av. Elvas s/n 06006, Badajoz, Spain
| | - Sonia Mota-Zamorano
- Department of Medical & Surgical Therapeutics, Medical School, University of Extremadura, Av. Elvas s/n 06006, Badajoz, Spain
| |
Collapse
|
17
|
Abstract
Treatment outcomes for acute lymphoblastic leukemia (ALL), especially pediatric ALL, have greatly improved due to the risk-adapted therapy. Combination of drug development, clinical practice, as well as basic genetic researches has brought the survival rate of ALL from less than 10% to more than 90% today, not only increasing the treatment efficacy but also limiting adverse drug reactions (ADRs). In this review, we summarized the landscape identification of ALL genetic alterations, which provided the opportunity to increase the survival rate and especially minimize the relapse risk of ALL, and highlighted the importance of the development of new technologies of genomic investigation for translational medicine.
Collapse
|
18
|
Genetic Mutations in B-Acute Lymphoblastic Leukemia Among African American and European American Children. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:e501-e508. [PMID: 30181103 DOI: 10.1016/j.clml.2018.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND The survival of patients with B-acute lymphoblastic leukemia (B-ALL) is significantly lower in African American (AA) children compared with European American children (EA). Here, we present a whole exome sequencing (WES) study showing race-specific genetic variations that may play a role on the disparate outcomes among AA and EA children with B-ALL. PATIENTS AND METHODS Five AA and 15 EA patients ranging in age from 1 to 18 years were enrolled. The median blast percentage was 94.8% (range, 64.5%-99.9%). Frozen bone marrow aspirate was used to extract DNA, and WES was performed, focusing on race and B-ALL-specific germline mutations. RESULTS Most genetic variants (n = 339) were shared between AA and EA children. Some genetic aberrations were only uniquely identified in AA (n = 58) and others in EA (n = 52) In AA, the genetic aberrations clustered in canonical pathways related to telomerase signaling and cancer signaling. In EA, the unique genetic aberration clustered in pathways related to stem cell pluripotency and hereditary cancer. CONCLUSIONS Our study revealed aberrant genetic aberrations in signaling networks that may contribute to race-specific aspects of leukemogenesis. Our results suggest the value of WES as a tool for development of individual gene signatures and gene scores for AA and EA children afflicted by B-ALL. These findings may ultimately impact disease management and contribute to the elimination of disparate outcomes in AA children with B-ALL.
Collapse
|
19
|
Abstract
In this Review, I present evidence supporting a multifactorial causation of childhood acute lymphoblastic leukaemia (ALL), a major subtype of paediatric cancer. ALL evolves in two discrete steps. First, in utero initiation by fusion gene formation or hyperdiploidy generates a covert, pre-leukaemic clone. Second, in a small fraction of these cases, the postnatal acquisition of secondary genetic changes (primarily V(D)J recombination-activating protein (RAG) and activation-induced cytidine deaminase (AID)-driven copy number alterations in the case of ETS translocation variant 6 (ETV6)-runt-related transcription factor 1 (RUNX1)+ ALL) drives conversion to overt leukaemia. Epidemiological and modelling studies endorse a dual role for common infections. Microbial exposures earlier in life are protective but, in their absence, later infections trigger the critical secondary mutations. Risk is further modified by inherited genetics, chance and, probably, diet. Childhood ALL can be viewed as a paradoxical consequence of progress in modern societies, where behavioural changes have restrained early microbial exposure. This engenders an evolutionary mismatch between historical adaptations of the immune system and contemporary lifestyles. Childhood ALL may be a preventable cancer.
Collapse
Affiliation(s)
- Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
20
|
Abstract
Folate has been studied in relation to many diseases, especially cancer. Although it has been postulated to exert a dual effect on development of cancer, its role remains to be clearly defined. Its effect on cancer is the result of gene-nutrient interaction between the genes in folate metabolic pathway and dietary folate availability; mutations in genes of folate metabolism have been shown to alter individual susceptibility to certain childhood cancers as well as response to cancer chemotherapy. Although mandatory fortification of food items with folate has been initiated in some countries, many countries are yet to adopt this due to concerns about undesired adverse effects of high folate levels on health, especially cancer. However, initial reports suggest that folate fortification has led to reduction in incidence of certain childhood cancers such as neuroblastoma, wilms tumour and leukaemias. Despite studies showing folate depletion during antifolate chemotherapy and higher toxicity of chemotherapy in folate-depleted individuals, folate supplementation during cancer chemotherapy is not routinely recommended. Studies investigating the precise effect of folate supplementation during chemotherapy on both short- and long-term outcomes of cancer are needed to arrive at a consensus guideline.
Collapse
Affiliation(s)
- Nirmalya Roy Moulik
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, King George's Medical University, Lucknow, India
| | - Archana Kumar
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, King George's Medical University, Lucknow, India
| | - Suraksha Agrawal
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
21
|
Lv H, Hu SY, Du ZZ, Zhai Z, Cao L, Sun YN, Lu J, Li J, He HL, Chai YH, Wang Y. Gene polymorphisms in the folate metabolic pathway and risk of pediatric acute lymphoblastic leukemia: a case-control study in a Chinese population. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1724-1731. [PMID: 31938276 PMCID: PMC6958122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 12/29/2017] [Indexed: 06/10/2023]
Abstract
Polymorphisms in folate pathway genes may influence susceptibility to pediatric acute lymphoblastic leukemia (ALL). This case-control study was undertaken to analyze the association of genetic polymorphisms (677C>T and 1298A>C) of methylenetetrahydrofolate reductase (MTHFR) and reduced folate carrier (RFC1) (80G>A) with the risk of pediatric ALL in China. A total of 176 pediatric ALL patients and 170 matched healthy subjects (as controls) were included and DNA was extracted from the peripheral blood. SNaPshot single nucleotide polymorphism typing was used to determine the genotypes of MTHFR 677C>T, MTHFR 1298A>C, and RFC1 80G>A. All statistical analyses were conducted with SAS software (version 9.2; SAS Institute). There were no significant differences in the genotype and allele frequencies of MTHFR 677C>T, MTHFR 1298A>C, or RFC1 80G>A between patients and controls. No significant correlation was found between the combined genotypes of these polymorphisms and the risk of developing ALL in this study. Furthermore, no significant differences were observed for 677C>T and 1298A>C frequencies between the control and case groups. There was no association between MTHFR 677C>T, MTHFR 1298A>C, or RFC1 80G>A gene polymorphisms and risk of pediatric ALL in the Han Chinese population.
Collapse
Affiliation(s)
- Hui Lv
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Shao-Yan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Zhi-Zuo Du
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Zong Zhai
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Lan Cao
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Yi-Na Sun
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Jie Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Hai-Long He
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Yi-Huan Chai
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| | - Yi Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University Suzhou 215123, Jiangsu Province, China
| |
Collapse
|
22
|
Genetic susceptibility in childhood acute lymphoblastic leukemia. Med Oncol 2017; 34:179. [PMID: 28905228 DOI: 10.1007/s12032-017-1038-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/05/2017] [Indexed: 12/27/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood malignancy and a leading cause of death due to disease in children. The genetic basis of ALL susceptibility has been supported by its association with certain congenital disorders and, more recently, by several genome-wide association studies (GWAS). These GWAS identified common variants in ARID5B, IKZF1, CEBPE, CDKN2A, PIP4K2A, LHPP and ELK3 influencing ALL risk. However, the risk variants of these SNPs were not validated in all populations, suggesting that some of the loci could be population specific. On the other hand, the currently identified risk SNPs in these genes only account for 19% of the additive heritable risk. This estimation indicates that additional susceptibility variants could be discovered. In this review, we will provide an overview of the most important findings carried out in genetic susceptibility of childhood ALL in all GWAS and subsequent studies and we will also point to future directions that could be explored in the near future.
Collapse
|
23
|
Al-Absi B, Razif MFM, Noor SM, Saif-Ali R, Aqlan M, Salem SD, Ahmed RH, Muniandy S. Contributions of IKZF1, DDC, CDKN2A, CEBPE, and LMO1 Gene Polymorphisms to Acute Lymphoblastic Leukemia in a Yemeni Population. Genet Test Mol Biomarkers 2017; 21:592-599. [PMID: 28768142 DOI: 10.1089/gtmb.2017.0084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Genome-wide and candidate gene association studies have previously revealed links between a predisposition to acute lymphoblastic leukemia (ALL) and genetic polymorphisms in the following genes: IKZF1 (7p12.2; ID: 10320), DDC (7p12.2; ID: 1644), CDKN2A (9p21.3; ID: 1029), CEBPE (14q11.2; ID: 1053), and LMO1 (11p15; ID: 4004). In this study, we aimed to conduct an investigation into the possible association between polymorphisms in these genes and ALL within a sample of Yemeni children of Arab-Asian descent. METHODS Seven single-nucleotide polymorphisms (SNPs) in IKZF1, three SNPs in DDC, two SNPs in CDKN2A, two SNPs in CEBPE, and three SNPs in LMO1 were genotyped in 289 Yemeni children (136 cases and 153 controls), using the nanofluidic Dynamic Array (Fluidigm 192.24 Dynamic Array). Logistic regression analyses were used to estimate ALL risk, and the strength of association was expressed as odds ratios with 95% confidence intervals. RESULTS We found that the IKZF1 SNP rs10235796 C allele (p = 0.002), the IKZF1 rs6964969 A>G polymorphism (p = 0.048, GG vs. AA), the CDKN2A rs3731246 G>C polymorphism (p = 0.047, GC+CC vs. GG), and the CDKN2A SNP rs3731246 C allele (p = 0.007) were significantly associated with ALL in Yemenis of Arab-Asian descent. In addition, a borderline association was found between IKZF1 rs4132601 T>G variant and ALL risk. No associations were found between the IKZF1 SNPs (rs11978267; rs7789635), DDC SNPs (rs3779084; rs880028; rs7809758), CDKN2A SNP (rs3731217), the CEBPE SNPs (rs2239633; rs12434881) and LMO1 SNPs (rs442264; rs3794012; rs4237770) with ALL in Yemeni children. CONCLUSION The IKZF1 SNPs, rs10235796 and rs6964969, and the CDKN2A SNP rs3731246 (previously unreported) could serve as risk markers for ALL susceptibility in Yemeni children.
Collapse
Affiliation(s)
- Boshra Al-Absi
- 1 Department of Molecular Medicine, University of Malaya , Kuala Lumpur, Malaysia
| | - Muhammad F M Razif
- 1 Department of Molecular Medicine, University of Malaya , Kuala Lumpur, Malaysia
| | - Suzita M Noor
- 2 Department of Biomedical Science, Faculty of Medicine, University of Malaya , Kuala Lumpur, Malaysia
| | - Riyadh Saif-Ali
- 3 Department of Biochemistry and Molecular Biology, Faculty of Medicine, Sana'a University , Sana'a, Yemen
| | - Mohammed Aqlan
- 4 Department of Pediatrics, Al-Kuwait University Hospital , Sana'a, Yemen
| | - Sameer D Salem
- 3 Department of Biochemistry and Molecular Biology, Faculty of Medicine, Sana'a University , Sana'a, Yemen
| | - Radwan H Ahmed
- 1 Department of Molecular Medicine, University of Malaya , Kuala Lumpur, Malaysia
| | - Sekaran Muniandy
- 5 Department of Biochemistry, Faculty of Medicine, MAHSA University , Kuala Lumpur, Malaysia
| |
Collapse
|
24
|
Burdennyy AM, Loginov VI, Zavarykina TM, Braga EA, Kubatiev AA. The role of molecular genetic alterations in genes involved in folate and homocysteine metabolism in multifactorial diseases pathogenesis. RUSS J GENET+ 2017. [DOI: 10.1134/s1022795417040044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
25
|
Al-Absi B, Noor SM, Saif-Ali R, Salem SD, Ahmed RH, Razif MF, Muniandy S. Association of ARID5B gene variants with acute lymphoblastic leukemia in Yemeni children. Tumour Biol 2017; 39:1010428317697573. [PMID: 28381164 DOI: 10.1177/1010428317697573] [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: 12/11/2022] Open
Abstract
Studies have shown an association between ARID5B gene polymorphisms and childhood acute lymphoblastic leukemia. However, the association between ARID5B variants and acute lymphoblastic leukemia among the Arab population still needs to be studied. The aim of this study was to investigate the association between ARID5B variants with acute lymphoblastic leukemia in Yemeni children. A total of 14 ARID5B gene single nucleotide polymorphisms (SNPs) were genotyped in 289 Yemeni children, of whom 136 had acute lymphoblastic leukemia and 153 were controls, using the nanofluidic Dynamic Array (Fluidigm 192.24 Dynamic Array). Using logistic regression adjusted for age and gender, the risks of acute lymphoblastic leukemia were presented as odds ratios and 95% confidence intervals. We found that nine SNPs were associated with acute lymphoblastic leukemia under additive genetic models: rs7073837, rs10740055, rs7089424, rs10821936, rs4506592, rs10994982, rs7896246, rs10821938, and rs7923074. Furthermore, the recessive models revealed that six SNPs were risk factors for acute lymphoblastic leukemia: rs10740055, rs7089424, rs10994982, rs7896246, rs10821938, and rs7923074. The gender-specific impact of these SNPs under the recessive genetic model revealed that SNPs rs10740055, rs10994982, and rs6479779 in females, and rs10821938 and rs7923074 in males were significantly associated with acute lymphoblastic leukemia risk. Under the dominant model, SNPs rs7073837, rs10821936, rs7896246, and rs6479778 in males only showed striking association with acute lymphoblastic leukemia. The additive model revealed that SNPs with significant association with acute lymphoblastic leukemia were rs10821936 (both males and females); rs7073837, rs10740055, rs10994982, and rs4948487 (females only); and rs7089424, rs7896246, rs10821938, and rs7923074 (males only). In addition, the ARID5B haplotype block (CGAACACAA) showed a higher risk for acute lymphoblastic leukemia. The haplotype (CCCGACTGC) was associated with protection against acute lymphoblastic leukemia. In conclusion, our study has shown that ARID5B variants are associated with acute lymphoblastic leukemia in Yemeni children with several gender biases of ARID5B single nucleotide polymorphisms reported.
Collapse
Affiliation(s)
- Boshra Al-Absi
- 1 Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Suzita M Noor
- 2 Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Riyadh Saif-Ali
- 3 Department of Biochemistry and Molecular Biology, Faculty of Medicine, Sana'a University, Sana'a, Yemen
| | - Sameer D Salem
- 3 Department of Biochemistry and Molecular Biology, Faculty of Medicine, Sana'a University, Sana'a, Yemen
| | - Radwan H Ahmed
- 1 Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Muhammad Fm Razif
- 1 Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sekaran Muniandy
- 1 Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
26
|
[Acute lymphoblastic leukemia: a genomic perspective]. BOLETIN MEDICO DEL HOSPITAL INFANTIL DE MEXICO 2017; 74:13-26. [PMID: 29364809 DOI: 10.1016/j.bmhimx.2016.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/14/2016] [Accepted: 07/07/2016] [Indexed: 11/22/2022] Open
Abstract
In parallel to the human genome sequencing project, several technological platforms have been developed that let us gain insight into the genome structure of human entities, as well as evaluate their usefulness in the clinical approach of the patient. Thus, in acute lymphoblastic leukemia (ALL), the most common pediatric malignancy, genomic tools promise to be useful to detect patients at high risk of relapse, either at diagnosis or during treatment (minimal residual disease), and they also increase the possibility to identify cases at risk of adverse reactions to chemotherapy. Therefore, the physician could offer patient-tailored therapeutic schemes. A clear example of the useful genomic tools is the identification of single nucleotide polymorphisms (SNPs) in the thiopurine methyl transferase (TPMT) gene, where the presence of two null alleles (homozygous or compound heterozygous) indicates the need to reduce the dose of mercaptopurine by up to 90% to avoid toxic effects which could lead to the death of the patient. In this review, we provide an overview of the genomic perspective of ALL, describing some strategies that contribute to the identification of biomarkers with potential clinical application.
Collapse
|
27
|
Rougée LRA, Riches Z, Berman JM, Collier AC. The Ontogeny and Population Variability of Human Hepatic NADPH Dehydrogenase Quinone Oxido-Reductase 1 (NQO1). Drug Metab Dispos 2016; 44:967-74. [PMID: 26856346 PMCID: PMC4931887 DOI: 10.1124/dmd.115.068650] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/05/2016] [Indexed: 01/16/2023] Open
Abstract
The NADPH dehydrogenase quinone oxido-reductase 1 (NQO1) enzyme is an antioxidant and metabolic enzyme that performs two electron reduction of quinones and other chemicals. Based on the physiologic role(s) of NQO1, we hypothesized that expression and activity of this enzyme would vary with age and other demographic variables. Cytosols from 117 archived human livers were investigated for changes in NQO1 with age, sex, obesity, and ethnicity. Protein expression but not activity of NQO1 was weakly negatively correlated with age (Spearman r = -0.2, P = 0.03). No sex differences were observed for either protein expression or activity and for ethnicity; Caucasians had greater NQO1 activity than Asians (P < 0.05). Overweight children had statistically significantly higher NQO1 activity as compared with ideal weight children (P < 0.05) although this difference was not observed in adults. These findings establish that NQO1 is approximately as active in children as adults. However, modeled NQO1 clearance (both allometric and physiologically based pharmacokinetics) predicted maturation at 23 to 26 years. This is almost certainly an overestimate, with error in the model resulting from a small sample size and inability to scale for age-related changes in hepatic cellularity and/or cytosolic protein content, and indicates a delay in reaching maximum clearance through the NQO1 pathway that is affected by physiologic development as much, or more than, biochemical development. Obesity may increase hepatic NQO1 activity in children, which is likely a protective mechanism in oxidative stress, but may also have significant implications for drug and chemical disposition in obese children.
Collapse
Affiliation(s)
- Luc R A Rougée
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| | - Zoe Riches
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| | - Jacob M Berman
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| | - Abby C Collier
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| |
Collapse
|
28
|
Gharbi H, Ben Hassine I, Soltani I, Safra I, Ouerhani S, Bel Haj Othmen H, Teber M, Farah A, Amouri H, Toumi NH, Abdennebi S, Abbes S, Menif S. Association of genetic variation in IKZF1, ARID5B, CDKN2A, and CEBPE with the risk of acute lymphoblastic leukemia in Tunisian children and their contribution to racial differences in leukemia incidence. Pediatr Hematol Oncol 2016; 33:157-67. [PMID: 27184773 DOI: 10.3109/08880018.2016.1161685] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent genome-wide association studies (GWAS) focusing on pediatric acute lymphoblastic leukemia (ALL), the most common malignancy in children younger than 15 years old, have found evidence that single-nucleotide polymorphisms (SNPs) in IKZF1 (7p12.2), ARID5B (10q21.2), CDKN2A (9p21.3), and CEBPE (14q11.2) are strongly associated to the risk of developing pediatric ALL. These studies have been conducted in European and Thai populations, and it is unclear whether these observations generalize to other populations with a lower incidence of pediatric ALL. In order to explore the impact of these variants on pediatric ALL risk in the Tunisian population, we genotyped 58 cases of pediatric ALL and 150 controls for SNPs rs4132601 (7p12.2), rs7089424 (10q21.2), rs3731217 (9p21.3), and rs2239633 (14q11.2). Our results, which are consistent with findings in European populations, show that 3 SNPs, i.e., rs4132601 (P = .00116, odds ratio [OR] = 2.78, 95% confidence interval [CI] = [1.42, 5.87]), rs7089424 (P = .0022, OR = 0.49, 95% CI = [0.31, 0.79]), and rs2239633 (P = .0010, OR = 0.47, 95% CI = [0.29, 0.75]) are significantly associated with a higher risk of developing pediatric ALL (P < .05). Furthermore, we show differences in allele frequencies in SNPs between Tunisian and Caucasian and/or Thai populations (e.g., CEBPE, rs2239633; population attributable risk [PAR] ∼15-fold the PAR of Thai population). These differences, combined with differences in linkage disequilibrium structure between populations and differences in size between populations, may contribute to racial differences in pediatric ALL incidence.
Collapse
Affiliation(s)
- Hanene Gharbi
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Islem Ben Hassine
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Ismail Soltani
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Ines Safra
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Slah Ouerhani
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Hind Bel Haj Othmen
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Mouheb Teber
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Ahlem Farah
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Hassiba Amouri
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | | | | | - Salem Abbes
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Samia Menif
- a Laboratory of Molecular and Cellular Hematology , Pasteur Institute of Tunis, University of Tunis El Manar , Tunis , Tunisia
| |
Collapse
|
29
|
Ghosh R, Rossner P, Honkova K, Dostal M, Sram RJ, Hertz-Picciotto I. Air pollution and childhood bronchitis: Interaction with xenobiotic, immune regulatory and DNA repair genes. ENVIRONMENT INTERNATIONAL 2016; 87:94-100. [PMID: 26655675 DOI: 10.1016/j.envint.2015.10.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 10/05/2015] [Accepted: 10/05/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Gene-environment interactions have been investigated for diseases such as asthma, chronic obstructive pulmonary disease, cancer etc. but acute disease like bronchitis has rarely been studied. We investigated interactions between air pollution (polycyclic aromatic hydrocarbons (PAH) and particulate matter <2.5 μm (PM2.5)) and single nucleotide polymorphisms (SNP) in EPHX1, IL10, STAT4 and XPC genes in relation to bronchitis in children aged 0-2 years. METHODS A stratified random sample of 1133 Czech children, born between 1994 and 1998 in two districts, were followed since birth, of which 626 were genotyped. Pediatrician-diagnosed bronchitis episodes were obtained from the medical records. Central-site monitors measured air pollution exposure. We used multivariable logistic regression and estimated coefficients using generalized estimating equations. Interaction was assessed between pollutants and genes and associations in genotype-specific strata were presented. False discovery rate was used to adjust for multiple comparisons. RESULTS There were 803 episodes of bronchitis with an incidence rate of 56 per 1000 child-months. We found significant gene-environment interaction between PAH and four SNPs (EPHX1, (rs2854461), STAT4 (rs16833215), XPC (rs2228001 and rs2733532)), which became non-significant after adjusting for multiple comparisons. PM2.5 interactions with two XPC SNPs (rs2228001 and rs2733532) remained significant after accounting for multiple comparisons and those with CC alleles had a more than doubling of odds, OR=2.65 (95% CI: 1.91, 3.69) and 2.72 (95% CI: 1.95, 3.78), respectively, per 25 μg/m(3) increase in exposure. CONCLUSION The findings suggest that the DNA repair gene XPC may play an important role in the air pollution-induced pathogenesis of the inflammatory disease bronchitis.
Collapse
Affiliation(s)
- Rakesh Ghosh
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States.
| | - Pavel Rossner
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14220 Prague, Czech Republic
| | - Katerina Honkova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14220 Prague, Czech Republic
| | - Miroslav Dostal
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14220 Prague, Czech Republic
| | - Radim J Sram
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14220 Prague, Czech Republic
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California, Davis, United States
| |
Collapse
|
30
|
Carbonari D, Chiarella P, Mansi A, Pigini D, Iavicoli S, Tranfo G. Biomarkers of susceptibility following benzene exposure: influence of genetic polymorphisms on benzene metabolism and health effects. Biomark Med 2016; 10:145-63. [PMID: 26764284 DOI: 10.2217/bmm.15.106] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Benzene is a ubiquitous occupational and environmental pollutant. Improved industrial hygiene allowed airborne concentrations close to the environmental context (1-1000 µg/m(3)). Conversely, new limits for benzene levels in urban air were set (5 µg/m(3)). The biomonitoring of exposure to such low benzene concentrations are performed measuring specific and sensitive biomarkers such as S-phenylmercapturic acid, trans, trans-muconic acid and urinary benzene: many studies referred high variability in the levels of these biomarkers, suggesting the involvement of polymorphic metabolic genes in the individual susceptibility to benzene toxicity. We reviewed the influence of metabolic polymorphisms on the biomarkers levels of benzene exposure and effect, in order to understand the real impact of benzene exposure on subjects with increased susceptibility.
Collapse
Affiliation(s)
- Damiano Carbonari
- INAIL Reaserch, Department of Occupational & Environmental Medicine, Epidemiology & Hygiene, Via Fontana Candida 1 - 00040 Monte Porzio Catone (RM), Italy
| | - Pieranna Chiarella
- INAIL Reaserch, Department of Occupational & Environmental Medicine, Epidemiology & Hygiene, Via Fontana Candida 1 - 00040 Monte Porzio Catone (RM), Italy
| | - Antonella Mansi
- INAIL Reaserch, Department of Occupational & Environmental Medicine, Epidemiology & Hygiene, Via Fontana Candida 1 - 00040 Monte Porzio Catone (RM), Italy
| | - Daniela Pigini
- INAIL Reaserch, Department of Occupational & Environmental Medicine, Epidemiology & Hygiene, Via Fontana Candida 1 - 00040 Monte Porzio Catone (RM), Italy
| | - Sergio Iavicoli
- INAIL Reaserch, Department of Occupational & Environmental Medicine, Epidemiology & Hygiene, Via Fontana Candida 1 - 00040 Monte Porzio Catone (RM), Italy
| | - Giovanna Tranfo
- INAIL Reaserch, Department of Occupational & Environmental Medicine, Epidemiology & Hygiene, Via Fontana Candida 1 - 00040 Monte Porzio Catone (RM), Italy
| |
Collapse
|
31
|
Huang X, Gao Y, He J, Cai J, Ta N, Jiang H, Zhu J, Zheng J. The association between RFC1 G80A polymorphism and cancer susceptibility: Evidence from 33 studies. J Cancer 2016; 7:144-152. [PMID: 26819637 PMCID: PMC4716846 DOI: 10.7150/jca.13303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/18/2015] [Indexed: 02/07/2023] Open
Abstract
Aberrant folate metabolism is closely related to tumorigenesis. Genetic variations in the Reduced folate carrier 1 (RFC1) may alter the progress of folate metabolism, and thereby cause the initiation and progress of the cancer. Considerable studies have performed to investigate the association between RFC1 G80A (rs1051266) polymorphism and cancer susceptibility, but the conclusions were conflicting. Therefore, we conducted a meta-analysis to reevaluate the association of RFC1 G80A polymorphism with cancer risk. PubMed and EMBASE were searched for eligible studies. The association of RFC1 G80A polymorphism and cancer risk was evaluated by the pooled odds ratios (ORs) and corresponding 95% confidence intervals (CIs). The significant association was found between RFC1 G80A polymorphism and hematological malignance susceptibility (A vs. G: OR=1.11, 95%CI=1.003-1.23, P=0.045; GA vs. GG: OR=1.18, 95%CI=1.06-1.31, P=0.002; AA+GA vs. GG: OR=1.18, 95%CI=1.07-1.29, P=0.001). Stratified analysis by ethnicity indicated that the association became more prominent among Caucasians (GA vs. GG: OR=1.28, 95%CI=1.12-1.45, P<0.001; AA+GA vs. GG: OR=1.21, 95%CI=1.08-1.36, P=0.001). In term of the cancer type, this polymorphism significantly increased the risk of acute lymphoblast leukemia (GA vs. GG: OR=1.13, 95%CI=1.001-1.28, P=0.048; AA+GA vs. GG: OR=1.28, 95%CI=1.13-1.46, P<0.001) and acute myeloid leukemia (GA vs. GG: OR=2.57, 95%CI=1.37-4.85, P=0.003). No significant association between RFC1 G80A polymorphism and overall solid cancer risk was observed, but a protective association with digestive cancer risk was found (GA vs. GG: OR=0.89, 95%CI= 0.81-0.99, P=0.030). The comprehensive meta-analysis encouraged the notion that RFC1 G80A polymorphism may play an important role in hematopoietic system malignance. These findings need further validation in the large multicenter investigations.
Collapse
Affiliation(s)
- Xiaoyi Huang
- 1. Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yisha Gao
- 1. Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jing He
- 2. Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jiao Cai
- 3. Department of Pathophysiology, Second Military Medical University, Shanghai 200433, China
| | - Na Ta
- 1. Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Hui Jiang
- 1. Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jinhong Zhu
- 4. Molecular Epidemiology Laboratory and Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Jianming Zheng
- 1. Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
32
|
Song J, Kim D, Chun C, Jin E. miR-370 and miR-373 regulate the pathogenesis of osteoarthritis by modulating one-carbon metabolism via SHMT-2 and MECP-2, respectively. Aging Cell 2015; 14:826-37. [PMID: 26103880 PMCID: PMC4568970 DOI: 10.1111/acel.12363] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2015] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to determine the mechanism underlying the association between one-carbon metabolism and DNA methylation during chronic degenerative joint disorder, osteoarthritis (OA). Articular chondrocytes were isolated from human OA cartilage and normal cartilage biopsied, and the degree of cartilage degradation was determined by safranin O staining. We found that the expression levels of SHMT-2 and MECP-2 were increased in OA chondrocytes, and 3′UTR reporter assays showed that SHMT-2 and MECP-2 are the direct targets of miR-370 and miR-373, respectively, in human articular chondrocytes. Our experiments showed that miR-370 and miR-373 levels were significantly lower in OA chondrocytes compared to normal chondrocytes. Overexpression of miR-370 or miR-373, or knockdown of SHMT-2 or MECP-2 reduced both MMP-13 expression and apoptotic cell death in cultured OA chondrocytes. In vivo, we found that introduction of miR-370 or miR-373 into the cartilage of mice that had undergone destabilization of the medial meniscus (DMM) surgery significantly reduced the cartilage destruction in this model, whereas introduction of SHMT-2 or MECP-2 increased the severity of cartilage destruction. Together, these results show that miR-370 and miR-373 contribute to the pathogenesis of OA and act as negative regulators of SHMT-2 and MECP-2, respectively.
Collapse
Affiliation(s)
- Jinsoo Song
- Department of Biological Sciences College of Natural Sciences Wonkwang University Iksan Chunbuk 570‐749Korea
| | - Dongkyun Kim
- Department of Biological Sciences College of Natural Sciences Wonkwang University Iksan Chunbuk 570‐749Korea
| | - Churl‐Hong Chun
- Department of Orthopedic Surgery Wonkwang University School of Medicine Iksan Chunbuk 570‐749Korea
| | - Eun‐Jung Jin
- Department of Biological Sciences College of Natural Sciences Wonkwang University Iksan Chunbuk 570‐749Korea
| |
Collapse
|
33
|
Carvalho DC, Wanderley AV, Amador MAT, Fernandes MR, Cavalcante GC, Pantoja KBCC, Mello FAR, de Assumpção PP, Khayat AS, Ribeiro-Dos-Santos Â, Santos S, Dos Santos NPC. Amerindian genetic ancestry and INDEL polymorphisms associated with susceptibility of childhood B-cell Leukemia in an admixed population from the Brazilian Amazon. Leuk Res 2015; 39:S0145-2126(15)30361-1. [PMID: 26321572 DOI: 10.1016/j.leukres.2015.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 07/14/2015] [Accepted: 08/15/2015] [Indexed: 12/29/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant tumor common in children. Studies of genetic susceptibility to cancer using biallelic insertion/deletion (INDEL) type polymorphisms associated with cancer development pathways may help to clarify etymology of ALL. In this study, we investigate the role of eight functional INDEL polymorphisms and influence of genetic ancestry to B-cell ALL susceptibility in children of Brazilian Amazon population, which has a high degree of inter-ethnic admixture. Ancestry analysis was estimated using a panel of 48 autosomal ancestry informative markers. 130 B-cell ALL patients and 125 healthy controls were included in this study. The odds ratios and 95% confidence intervals were adjusted for confounders. The results indicated an association between the investigated INDEL polymorphisms in CASP8 (rs3834129), CYP19A1 (rs11575899) e XRCC1 (rs3213239) genes in the development of B-cell ALL. The carriers of Insertion/Insertion (Ins/Ins) genotype of the polymorphism in CASP8 gene presented reduced chances of developing B-cell ALL (P=0.001; OR=0.353; 95% CI=0.192-0.651). The Deletion/Deletion (Del/Del) genotype of the polymorphism in CYP19A1 gene was associated to a lower chance of developing B-cell ALL (P=3.35×10-6; OR=0.121; 95% CI=0.050-0.295), while Del/Del genotype of the polymorphism in XRCC1 gene was associated to a higher chance of developing B-cell ALL (P=2.01×10-4; OR=6.559; 95% CI=2.433-17.681). We also found that Amerindian ancestry correlates with the risk of B-cell ALL. For each increase of 10% in the Amerindian ancestry results in 1.4-fold chances of developing B-cell ALL (OR=1.406; 95% IC=1.123-1.761), while each increase of 10% in the European ancestry presents a protection effect in the development of B-cell ALL (OR=0.666; 95% IC=0.536-0.827). The results suggest that genetic factors influence leukemogenesis and might be explored in the stratification of B-cell ALL risk in admixed populations.
Collapse
Affiliation(s)
- Darlen C Carvalho
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| | - Alayde V Wanderley
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Hospital Ophir Loyola, Departamento de Pediatria, Belém, Pará, PA, Brazil.
| | - Marcos A T Amador
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| | - Marianne R Fernandes
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| | - Giovanna C Cavalcante
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| | - Karla B C C Pantoja
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| | - Fernando A R Mello
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil.
| | - Paulo P de Assumpção
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA, Brazil.
| | - André S Khayat
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil.
| | - Ândrea Ribeiro-Dos-Santos
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| | - Sidney Santos
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| | - Ney P C Dos Santos
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Pará, PA, Brazil; Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Belém, Pará, PA, Brazil.
| |
Collapse
|
34
|
De Braekeleer M, De Braekeleer E, Douet-Guilbert N. Geographic/ethnic variability of chromosomal and molecular abnormalities in leukemia. Expert Rev Anticancer Ther 2015. [DOI: 10.1586/14737140.2015.1068123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
Inherited genetic variation in childhood acute lymphoblastic leukemia. Blood 2015; 125:3988-95. [PMID: 25999454 DOI: 10.1182/blood-2014-12-580001] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/30/2015] [Indexed: 02/06/2023] Open
Abstract
Although somatically acquired genomic alterations have long been recognized as the hallmarks of acute lymphoblastic leukemia (ALL), the last decade has shown that inherited genetic variations (germline) are important determinants of interpatient variability in ALL susceptibility, drug response, and toxicities of ALL therapy. In particular, unbiased genome-wide association studies have identified germline variants strongly associated with the predisposition to ALL in children, providing novel insight into the mechanisms of leukemogenesis and evidence for complex interactions between inherited and acquired genetic variations in ALL. Similar genome-wide approaches have also discovered novel germline genetic risk factors that independently influence ALL prognosis and those that strongly modify host susceptibility to adverse effects of antileukemic agents (eg, vincristine, asparaginase, glucocorticoids). There are examples of germline genomic associations that warrant routine clinical use in the treatment of childhood ALL (eg, TPMT and mercaptopurine dosing), but most have not reached this level of actionability. Future studies are needed to integrate both somatic and germline variants to predict risk of relapse and host toxicities, with the eventual goal of implementing genetics-driven precision-medicine approaches in ALL treatment.
Collapse
|
36
|
Hsu LI, Chokkalingam AP, Briggs FBS, Walsh K, Crouse V, Fu C, Metayer C, Wiemels JL, Barcellos LF, Buffler PA. Association of genetic variation in IKZF1, ARID5B, and CEBPE and surrogates for early-life infections with the risk of acute lymphoblastic leukemia in Hispanic children. Cancer Causes Control 2015; 26:609-19. [PMID: 25761407 PMCID: PMC4504234 DOI: 10.1007/s10552-015-0550-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 03/02/2015] [Indexed: 12/01/2022]
Abstract
BACKGROUND Genome-wide association studies focusing on European-ancestry populations have identified ALL risk loci on IKZF1, ARID5B, and CEBPE. To capture the impacts of these genes on ALL risk in the California Hispanic population, we comprehensively assessed the variation within the genes and further assessed the joint effects between the genetic variation and surrogates for early-life infections (the presence of older siblings, daycare attendance, and ear infections). METHODS Genotypic data for 323 Hispanic ALL cases and 454 controls from the California Childhood Leukemia Study were generated using Illumina OmniExpress v1 platform. Logistic regression assuming a log-additive model estimated odds ratios (OR) associated with each SNP, adjusted for age, sex, and the first five principal components. In addition, we examined potential interactions between six ALL risk alleles and surrogates for early-life infections using logistic regression models that included an interaction term. RESULTS Significant associations between genotypes at IKZF1, ARID5B, and CEBPE and ALL risk were identified: rs7780012, OR 0.50, 95% confidence interval (CI) 0.35-0.71 (p = 0.004); rs7089424, OR 2.12, 95% CI 1.70-2.65 (p = 1.16 × 10(-9)); rs4982731, OR 1.69, 95% CI 1.37-2.08 (p = 2.35 × 10(-6)), respectively. Evidence for multiplicative interactions between genetic variants and surrogates for early-life infections with ALL risk was not observed. CONCLUSIONS Consistent with findings in non-Hispanic White population, our study showed that variants within IKZF1, ARID5B, and CEBPE were associated with increased ALL risk, and the effects for ARID5B and CEBPE were most prominent in the high-hyperdiploid ALL subtype in the California Hispanic population. Results implicate the ARID5B, CEBPE, and IKZF1 genes in the pathogenesis of childhood ALL.
Collapse
Affiliation(s)
- Ling-I Hsu
- School of Public Health, University of California, Berkeley, Berkeley, CA, 94720, USA,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Weich N, Nuñez MC, Galimberti G, Elena G, Acevedo S, Larripa I, Fundia AF. Polymorphic variants of GSTM1, GSTT1, and GSTP1 genes in childhood acute leukemias: A preliminary study in Argentina. ACTA ACUST UNITED AC 2015; 20:511-6. [PMID: 25799091 DOI: 10.1179/1607845415y.0000000007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND AIM Despite recent major advances in leukemia research, the etiopathogenesis of childhood leukemias remains far elusive. Individual predisposing factors, including polymorphisms in detoxification enzymes, have been implicated in the molecular pathogenesis and heterogeneity of the disease. Genetic polymorphisms of glutathione S-transferases (GSTs) that alter enzyme activity could be an additional factor that increases the risk of acute leukemia, but data are lacking in Argentina. We assessed the association of GST polymorphisms and the susceptibility to childhood leukemia in Argentina by conducting an exploratory case-control study and correlated patients' genotype to clinical and biological features. METHODS Deletion polymorphisms in GSTM1 and GSTT1 genes and the single nucleotide polymorphism in GSTP1 c.313A>G (rs1695; p.105Ile>Val) were genotyped by PCR-RFLP in 36 patients and 133 healthy individuals. RESULTS GSTM1-null genotype was associated with a lower risk of developing acute leukemia (P = 0.013; OR: 0.31; CI: 0.12-0.80), while GSTP1-GG variants displayed an increased risk (P = 0.01; OR: 3.9; CI: 1.85-8.2). However, no differences were found for GSTT1 gene. Conclusion These preliminary results, to be validated in a larger population from Argentina, suggest that the development of pediatric leukemia may be differentially influenced by polymorphic variants in GST genes.
Collapse
|
38
|
Goričar K, Erčulj N, Faganel Kotnik B, Debeljak M, Hovnik T, Jazbec J, Dolžan V. The association of folate pathway and DNA repair polymorphisms with susceptibility to childhood acute lymphoblastic leukemia. Gene 2015; 562:203-9. [PMID: 25746326 DOI: 10.1016/j.gene.2015.02.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 01/10/2015] [Accepted: 02/26/2015] [Indexed: 01/21/2023]
Abstract
Genetic factors may play an important role in susceptibility to childhood acute lymphoblastic leukemia (ALL). The aim of our study was to evaluate the associations of genetic polymorphisms in folate pathway and DNA repair genes with susceptibility to ALL. In total, 121 children with ALL and 184 unrelated healthy controls of Slovenian origin were genotyped for 14 polymorphisms in seven genes of folate pathway, base excision repair and homologous recombination repair (TYMS, MTHFR, OGG1, XRCC1, NBN, RAD51, and XRCC3). In addition, the exon 6 of NBN was screened for the presence of mutations using denaturing high performance liquid chromatography. Twelve polymorphisms were in Hardy-Weinberg equilibrium in controls and their genotype frequencies were in agreement with those reported in other Caucasian populations. Among the investigated polymorphisms and mutations, NBN Glu185Gln significantly decreased susceptibility to B-cell ALL (p=0.037), while TYMS 3R allele decreased susceptibility to T-cell ALL (p=0.011). Moreover, significantly decreased susceptibility to ALL was observed for MTHFR TA (p=0.030) and RAD51 GTT haplotypes (p=0.016). Susceptibility to ALL increased with the increasing number of risk alleles (ptrend=0.007). We also observed significant influence of hOGG-RAD51 and NBN-RAD51 interactions on susceptibility to ALL. Our results suggest that combination of several polymorphisms in DNA repair and folate pathways may significantly affect susceptibility to childhood ALL.
Collapse
Affiliation(s)
- Katja Goričar
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| | - Nina Erčulj
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| | - Barbara Faganel Kotnik
- University Medical Center, University Children's Hospital, Oncology and Haematology Unit, Bohoričeva 20, Ljubljana, Slovenia.
| | - Maruša Debeljak
- University Medical Center, University Children's Hospital, Center for Medical Genetics, Vrazov trg 1, Ljubljana, Slovenia.
| | - Tinka Hovnik
- University Medical Center, University Children's Hospital, Center for Medical Genetics, Vrazov trg 1, Ljubljana, Slovenia.
| | - Janez Jazbec
- University Medical Center, University Children's Hospital, Oncology and Haematology Unit, Bohoričeva 20, Ljubljana, Slovenia.
| | - Vita Dolžan
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry, Pharmacogenetics Laboratory, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| |
Collapse
|
39
|
Milne E, Greenop KR, Scott RJ, Haber M, Norris MD, Attia J, Jamieson SE, Miller M, Bower C, Bailey HD, Dawson S, McCowage GB, de Klerk NH, van Bockxmeer FM, Armstrong BK. Folate pathway gene polymorphisms, maternal folic acid use, and risk of childhood acute lymphoblastic leukemia. Cancer Epidemiol Biomarkers Prev 2015; 24:48-56. [PMID: 25395472 DOI: 10.1158/1055-9965.epi-14-0680] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Several studies suggest that maternal folic acid supplementation before or during pregnancy protects against childhood acute lymphoblastic leukemia (ALL). We investigated associations between ALL risk and folate pathway gene polymorphisms, and their modification by maternal folic acid supplements, in a population-based case-control study (2003-2007). METHODS All Australian pediatric oncology centers provided cases; controls were recruited by national random digit dialing. Data from 392 cases and 535 controls were included. Seven folate pathway gene polymorphisms (MTHFR 677C>T, MTHFR 1298A>C, MTRR 66A>G, MTR 2756 A>G, MTR 5049 C>A, CBS 844 Ins68, and CBS 2199 T>C) were genotyped in children and their parents. Information on prepregnancy maternal folic acid supplement use was collected. ORs were estimated with unconditional logistic regression adjusted for frequency-matched variables and potential confounders. Case-parent trios were also analyzed. RESULTS There was some evidence of a reduced risk of ALL among children who had, or whose father had, the MTRR 66GG genotype: ORs 0.60 [95% confidence interval (CI) 0.39-0.91] and 0.64 (95% CI, 0.40-1.03), respectively. The ORs for paternal MTHFR 677CT and TT genotypes were 1.41 (95% CI, 1.02-1.93) and 1.81 (95% CI, 1.06-3.07). ORs varied little by maternal folic acid supplementation. CONCLUSIONS Some folate pathway gene polymorphisms in the child or a parent may influence ALL risk. While biologically plausible, underlying mechanisms for these associations need further elucidation. IMPACT Folate pathway polymorphisms may be related to risk of childhood ALL, but larger studies are needed for conclusive results.
Collapse
Affiliation(s)
- Elizabeth Milne
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.
| | - Kathryn R Greenop
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Rodney J Scott
- Hunter Medical Research Institute, John Hunter Hospital, New Lambton, New South Wales, Australia. School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, New South Wales, Australia. Hunter Area Pathology Service, HNEHealth, Newcastle, New South Wales, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - John Attia
- Hunter Medical Research Institute, John Hunter Hospital, New Lambton, New South Wales, Australia. School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia
| | - Sarra E Jamieson
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Margaret Miller
- School of Exercise and Health Sciences, Edith Cowan University, Mount Lawley, Western Australia, Australia
| | - Carol Bower
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Helen D Bailey
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia. Section of Environment and Radiation, International Agency for Research on Cancer, Lyon, France
| | - Somer Dawson
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | | | - Nicholas H de Klerk
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Frank M van Bockxmeer
- Department of Clinical Biochemistry, Royal Perth Hospital and the School of Surgery, University of Western Australia, Perth, Western Australia, Australia
| | - Bruce K Armstrong
- Sax Institute, Haymarket, New South Wales, Australia. Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Ma L, Liu H, Ruan L, Yang X, Yang H, Feng Y. Multidrug resistance gene 1 C1236T polymorphism and susceptibility to leukemia: A meta-analysis. Biomed Rep 2014; 3:83-87. [PMID: 25469253 DOI: 10.3892/br.2014.387] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/09/2014] [Indexed: 12/26/2022] Open
Abstract
Several studies have investigated the association between multidrug resistance gene (MDR1) C1236T polymorphism and leukemia risk, however, these published studies have yielded conflicting results. Thus, the present study carried out a meta-analysis to provide a more precise estimate of the effect of this polymorphism on the susceptibility to leukemia. The published case-control studies regarding the association between MDR1 C1236T polymorphism and leukemia risk were included following a computerized search of PubMed, Elsevier, The Cochrane Library, China National Knowledge Infrastructure and Wanfang Database. Either fixed- or random-effects models were applied to calculate the combined odds ratios (ORs) and 95% confidence intervals (CIs) by RevMan 5.2 software. Seven studies, including 846 cases and 1,523 controls, were included in the present meta-analysis. The results indicated that there was no significant association between the MDR1 C1236T polymorphism and leukemia risk in overall comparisons in all four genetic models (CT vs. CC: OR, 1.31, 95% CI, 0.89-1.91, P=0.17; TT vs. CC: OR, 2.16, 95% CI, 0.99-4.70, P=0.05; TT vs. CC+CT: OR, 1.72, 95% CI, 0.91-3.25, P=0.09; and CT+TT vs. CC: OR, 1.57, 95% CI, 0.96-2.56, P=0.07). In the subgroup analysis according to specific ethnicity, age and the type of leukemia, a significant association was found in adult leukemia (CT+TT vs. CC: OR, 2.77, 95% CI, 1.05-7.31, P=0.04) and chronic myeloid leukemia (CT vs. CC: OR, 1.71, 95% CI, 1.05-2.80, P=0.03). No significant publication bias was detected by funnel plot. Therefore, the meta-analysis indicated that the MDR1 C1236T polymorphism may contribute to the susceptibility to adult leukemia and chronic myeloid leukemia. Furthe well-designed studies based on larger sample sizes are required to validate these findings.
Collapse
Affiliation(s)
- Limin Ma
- Department of Hematology, The First Affiliated Hospital, Luoyang, Henan 471003, P.R. China
| | - Hongchao Liu
- Medical College, Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Linhai Ruan
- Department of Hematology, The First Affiliated Hospital, Luoyang, Henan 471003, P.R. China
| | - Xuewen Yang
- Department of Hematology, The First Affiliated Hospital, Luoyang, Henan 471003, P.R. China
| | - Haiping Yang
- Department of Hematology, The First Affiliated Hospital, Luoyang, Henan 471003, P.R. China
| | - Yanming Feng
- Medical College, Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
41
|
Laurier D, Grosche B, Auvinen A, Clavel J, Cobaleda C, Dehos A, Hornhardt S, Jacob S, Kaatsch P, Kosti O, Kuehni C, Lightfoot T, Spycher B, Van Nieuwenhuyse A, Wakeford R, Ziegelberger G. Childhood leukaemia risks: from unexplained findings near nuclear installations to recommendations for future research. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2014; 34:R53-R68. [PMID: 24938793 DOI: 10.1088/0952-4746/34/3/r53] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Recent findings related to childhood leukaemia incidence near nuclear installations have raised questions which can be answered neither by current knowledge on radiation risk nor by other established risk factors. In 2012, a workshop was organised on this topic with two objectives: (a) review of results and discussion of methodological limitations of studies near nuclear installations; (b) identification of directions for future research into the causes and pathogenesis of childhood leukaemia. The workshop gathered 42 participants from different disciplines, extending widely outside of the radiation protection field. Regarding the proximity of nuclear installations, the need for continuous surveillance of childhood leukaemia incidence was highlighted, including a better characterisation of the local population. The creation of collaborative working groups was recommended for consistency in methodologies and the possibility of combining data for future analyses. Regarding the causes of childhood leukaemia, major fields of research were discussed (environmental risk factors, genetics, infections, immunity, stem cells, experimental research). The need for multidisciplinary collaboration in developing research activities was underlined, including the prevalence of potential predisposition markers and investigating further the infectious aetiology hypothesis. Animal studies and genetic/epigenetic approaches appear of great interest. Routes for future research were pointed out.
Collapse
Affiliation(s)
- D Laurier
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), BP 17, F-92262 Fontenay-aux-Roses Cedex, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Smolkova B, Dusinska M, Hemminki K. NBN and XRCC3 genetic variants in childhood acute lymphoblastic leukaemia. Cancer Epidemiol 2014; 38:563-8. [PMID: 25176580 DOI: 10.1016/j.canep.2014.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 08/03/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022]
Abstract
Nibrin and DNA repair protein XRCC3 are involved in DNA double-strand break repair. We genotyped seven tagging SNPs in these genes (rs1805794, rs709816; rs1063054; rs7141928, rs1799794, rs861530, rs861539) with the aim to analyse their association with acute lymphoblastic leukaemia (ALL), a disease, that is characterised by elevated genetic instability. Study consisted of 460 paediatric ALL cases and 552 healthy controls. For selection of DNA sequence variants we employed SNP-tagging approach, incorporating the HAPMAP CEU reference panel data. We did not find association of analysed and tagged SNPs and derived haplotypes with the ALL risk thus did not confirm the hypothesis that analysed DNA recombination repair variants account for increased susceptibility to ALL.
Collapse
Affiliation(s)
- Bozena Smolkova
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| | - Maria Dusinska
- Health Effects Laboratory MILK, NILU-Norwegian Institute for Air Research, Kjeller, Norway.
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
43
|
Glutathione-S-transferase polymorphism and acute lymphoblastic leukemia (ALL) in north Indian children: a case-control study and meta-analysis. J Hum Genet 2014; 59:529-35. [PMID: 25102096 DOI: 10.1038/jhg.2014.66] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/03/2014] [Accepted: 07/04/2014] [Indexed: 02/02/2023]
Abstract
Various studies on association of glutathione S-transferase (GST) polymorphisms and childhood acute lymphoblastic leukemia (ALL) have yielded conflicting results. We examined this association among north Indian children and conducted an updated meta-analysis to overcome sample size-related limitations. GSTM1, GSTP1 and GSTT1 genotypes in 100 children with ALL and 300 healthy controls were compared. GSTT1 null mutation (odds ratio (OR) 2.54, 95% confidence interval (CI) 1.50-4.32) and GSTP1 homozygous mutation (OR 3.13, 95%CI 1.48-6.59) were found to increase the risk of childhood ALL, while GSTM1 did not alter the risk. Meta-analysis included 22, 10 and 20 studies examining the association of childhood ALL with GSTM1, GSTP1 and GSTT1 genotypes, respectively. Only GSTM1 genotype (OR 1.29, 95%CI 1.10-1.62) was associated with increased risk in the overall analysis. However, both GSTM1 (OR 1.54, 95%CI 1.12-2.10) and GSTT1 (OR 1.63, 95%CI 1.32-1.99) null genotypes were associated with increased risk in Asian subjects. The risk of developing childhood ALL was not associated with GSTP1 genotype.
Collapse
|
44
|
Offenmüller S, Ravindranath Y, Goyette G, Kanakapalli D, Miller KS, Brecht IB, Zolk O. Focused screening of a panel of cancer-related genetic polymorphisms reveals new susceptibility loci for pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 2014; 61:1411-5. [PMID: 24604828 DOI: 10.1002/pbc.25011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 02/03/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND A genetic predisposition to acute lymphoblastic leukemia (ALL) in childhood is well established. Currently known risk loci, however, explain only one third of the estimated total risk related to common genetic variations. PROCEDURE We genotyped 1,421 polymorphisms in 407 candidate genes from the SNP500Cancer database (National Cancer Institute) using the Illumina Cancer SNP Panel. We investigated 78 cases (aged 0-19 years at diagnosis, and mixed ethnic background) of childhood B-precursor ALL and compared genotype data with those of 1,417 HapMap controls. To account for the ethnic diversity of the study population, structured association by genetically matching cases and controls using identity-by-state similarity was used. Case-control association analyses were performed using Cochran-Mantel-Haenszel tests, adjusted for the population substructure. RESULTS Common variations rs6966 (3' UTR of PPP1R13L, chr 19q13.32, P = 4.55 × 10(-9)) and rs414580 (intron 2 of MSR1, chr 8p22, P = 6.09 × 10(-8)) were significantly associated with ALL. These SNPs remained significant after adjustment for multiple testing. The SNP rs6966 tags a haplotype block which includes SNPs in PPP1R13L and ERCC2 genes, which are related to DNA repair and cell survival. rs6966 and rs414580 conferred allelic odds ratios of 3.74 (95% confidence interval [CI] 2.31-6.04) and 3.93 (95% CI 2.31-6.69), respectively. CONCLUSIONS These findings reveal two independent novel susceptibility loci for childhood ALL.
Collapse
Affiliation(s)
- Sonja Offenmüller
- Pediatric Oncology and Hematology, University Children's Hospital, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Roy Moulik N, Parveen F, Kumar A, Awasthi S, Agrawal S. MTHFR gene polymorphism in acute lymphoblastic leukemia among North Indian children: a case-control study and meta-analysis updated from 2011. J Hum Genet 2014; 59:397-404. [PMID: 24919644 DOI: 10.1038/jhg.2014.44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 03/13/2014] [Accepted: 04/27/2014] [Indexed: 11/09/2022]
Abstract
Studies on the association of methylenetetrahydrofolate reductase (MTHFR) genotype in childhood acute lymphoblastic leukemia (ALL) have yielded conflicting results. The present study examines this association in north Indian children with ALL and includes an updated meta-analysis. MTHFR (677 and 1298) genotype of children with ALL and healthy adult controls were done by the PCR-restriction fragment length polymorphism (PCR-RFLP) method and were compared using various models of inheritance. A total of 150 patients and 300 controls were included. The 677T allele was found protective (odds ratio (OR) 0.21, 95% confidence interval (CI) 0.04-0.94), whereas 1298C allele led to an increase in risk (OR 4.44, 95% CI 2.19-8.99) of childhood ALL. Meta-analysis included 31 and 27 studies examining the association of 677 and 1298 genotypes, respectively. The 677 C -> T polymorphism was protective (OR 0.90, 95% CI 0.82-0.99). Protection was more pronounced in folate-sufficient populations as compared with those not covered by folate fortification guidelines. The 1298A->C polymorphism was associated with a marginal increase in risk (OR 1.19, 95% CI 1.01-1.40).
Collapse
Affiliation(s)
- Nirmalya Roy Moulik
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, King George's Medical University, Lucknow, India
| | - Farah Parveen
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Archana Kumar
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, King George's Medical University, Lucknow, India
| | - Shally Awasthi
- Department of Pediatrics, King George's Medical University, Lucknow, India
| | - Suraksha Agrawal
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
46
|
Li C, Zhou Y. Association between NQO1 C609T polymorphism and acute lymphoblastic leukemia risk: evidence from an updated meta-analysis based on 17 case-control studies. J Cancer Res Clin Oncol 2014; 140:873-81. [PMID: 24488035 DOI: 10.1007/s00432-014-1595-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Quinone oxidoreductase (NQO1) C609T polymorphisms have been implicated in acute lymphoblastic leukemia (ALL) risk, but previously published studies were inconsistent and recent meta-analyses were not adequate. The aim of this study was to determine more precise estimations for the relationship between the NQO1 C609T polymorphism and the risk of ALL. METHODS Electronic searches for all publications were conducted on association between this variant and ALL in several databases updated in May 2013. The quality of studies was evaluated using the Newcastle-Ottawa Scale. Crude odds ratios (ORs) with 95 % confidence intervals (CIs) were used to assess the strength of the association. Seventeen studies were identified, including 2,264 ALL patients and 3,798 controls. RESULTS Overall, significantly elevated ALL risk was associated with NQO1 C609T variant genotypes when all of the studies were pooled into the meta-analysis (TT vs. CC: OR 1.46, 95 % CI 1.18-1.79; dominant model: OR 1.45, 95 % CI 1.19-1.77). In the subgroup analysis by ethnicity, significantly increased risks were found for non-Asians (T/T vs. C/C: OR 1.74, 95 % CI 1.29-2.36; dominant model: T/T + C/T vs. C/C: OR 1.7, 95 % CI 1.27-2.29). When stratified by adult or children studies, statistically significantly elevated risks were found among adult studies (codominant model: C/T vs. C/C: OR 1.38, 95 % CI 1.02-1.87; dominant model: T/T + C/T vs. C/C: OR 1.52, 95 % CI 1.18-1.97) and children studies (recessive model: T/T vs. C/T + C/C: OR 1.34, 95 % CI 1.05-1.7). CONCLUSIONS Our results indicate that the C609T polymorphism of the NQO1 gene is an important genetic risk factor in ALL.
Collapse
Affiliation(s)
- Cuiping Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | | |
Collapse
|
47
|
Li C, Liu Y, Wei S, Zhou Y. A meta-analysis of the association between NQO1 C609T variation and acute myeloid leukemia risk. Pediatr Blood Cancer 2014; 61:771-7. [PMID: 24474393 DOI: 10.1002/pbc.24924] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/11/2013] [Indexed: 11/11/2022]
Abstract
Quinone oxidoreductase (NQO1) C609T polymorphisms have been implicated in acute myeloblastic leukemia (AML) risk, but previously published studies are inconsistent and recent meta-analyses have not been adequate. To derive a more precise estimation of the relationship, a meta-analysis was performed. Medline, PubMed, Embase, and Web of Science were searched. The quality of studies was evaluated by using the Newcastle-Ottawa Scale (NOS). Crude ORs with 95% CIs were used to assess the strength of association between the NQO1 C609T polymorphisms and AML risk. A total of 14 studies including 2,245 cases and 3,310 controls were involved in this meta-analysis. Overall, significantly elevated AML risk was associated with NQO1 C609T variant genotypes when all studies were pooled into the meta-analysis (TT vs. CC: OR = 1.44, 95% CI = 1.15-1.81; dominant model: OR = 1.35, 95% CI = 1.09-1.68). In the subgroup analysis by ethnicity, significantly increased risks were found for Asians (OR = 1.47, 95% CI = 1.13-1.93, P = 0.005, I(2) = 48.4%, P = 0.071 for heterogeneity). When stratified by studies of adults or children, statistically significantly elevated risks were found among adults (OR = 1.37, 95% CI = 1.06-1.76, P = 0.017, I(2) = 42.2%, P = 0.097 for heterogeneity). The accumulated evidence indicates that NQO1 C609T seems to confer a risk factor for AML among Asians and adults. Significant between-study heterogeneity was observed, thus more studies based on larger case-control population are required to further evaluate the role of NQO1 C609T polymorphism in AML.
Collapse
Affiliation(s)
- Cuiping Li
- School of Public Health, Guangxi Medical University, Nanning, People's Republic of China
| | | | | | | |
Collapse
|
48
|
Andrade FG, Furtado-Silva JM, Gonçalves BADA, Thuler LCS, Barbosa TC, Emerenciano M, Siqueira A, Pombo-de-Oliveira MS. RAS mutations in early age leukaemia modulated by NQO1 rs1800566 (C609T) are associated with second-hand smoking exposures. BMC Cancer 2014; 14:133. [PMID: 24571676 PMCID: PMC3946262 DOI: 10.1186/1471-2407-14-133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 02/17/2014] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Deregulation of the MAPK genes signalling caused by somatic mutations have been implied in leukaemia pathogenesis, including RAS mutation (RASmut) in acute myeloid leukaemia (AML), which has been associated with intra-uterine chemical exposures. A case-case study was conducted in order to explore maternal and child exposures to tobacco smoking associations with early age leukaemia (EAL). METHODS Covariables of reference were MLL rearrangements (MLL-r), RASmut and NQO1 rs1800566 (C609T). Samples from 150 acute lymphoblastic leukaemia (ALL) and 85 AML were included. Maternal exposures were assessed using a structured questionnaire with demographic, personal habits and residence history information. Restriction fragment length polymorphism and denaturing high performance liquid chromatography were used to screen FLT3, KRAS, and NRAS mutations; direct sequencing was performed to validate the results. NQO1 polymorphism was detected by real-time allelic discrimination technique. RESULTS Overall, RASmut were detected in 28.7% of EAL cases; BRAFmut was found only in one AML patient. Higher rate of KRASmut was found in ALL (30.3%) compared to AML (20.8%) with MLL-r; RASmut showed an association with second-hand tobacco smoking exposures (OR, 3.06, 95% CI, 1.03-9.07). A considerable increased risk for EAL with the combination of RASmut and NQO1 609CT (OR, 4.24, 95% CI, 1.24-14.50) was observed. CONCLUSIONS Our data demonstrated the increased risk association between maternal smoking and EAL with MLL-r. Additionally, suggests that children second-hand tobacco exposures are associated with increased risk of EAL with RASmut modulated by NQO1 rs1800566 (C609T).
Collapse
Affiliation(s)
- Francianne Gomes Andrade
- Paediatric Haematology-Oncology Program, Research Centre, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, 37, Rio de Janeiro/RJ 20231-050, Brasil
| | - Juliana Montibeller Furtado-Silva
- Paediatric Haematology-Oncology Program, Research Centre, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, 37, Rio de Janeiro/RJ 20231-050, Brasil
- Great Ormond Street Hospital, London, United Kingdom
| | - Bruno Alves de Aguiar Gonçalves
- Paediatric Haematology-Oncology Program, Research Centre, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, 37, Rio de Janeiro/RJ 20231-050, Brasil
| | | | - Thayana Conceição Barbosa
- Paediatric Haematology-Oncology Program, Research Centre, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, 37, Rio de Janeiro/RJ 20231-050, Brasil
| | - Mariana Emerenciano
- Paediatric Haematology-Oncology Program, Research Centre, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, 37, Rio de Janeiro/RJ 20231-050, Brasil
| | | | - Maria S Pombo-de-Oliveira
- Paediatric Haematology-Oncology Program, Research Centre, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, 37, Rio de Janeiro/RJ 20231-050, Brasil
| |
Collapse
|
49
|
Emerenciano M, Barbosa TC, Lopes BA, Blunck CB, Faro A, Andrade C, Meyer C, Marschalek R, Pombo-de-Oliveira MS. ARID5B polymorphism confers an increased risk to acquire specific MLL rearrangements in early childhood leukemia. BMC Cancer 2014; 14:127. [PMID: 24564228 PMCID: PMC3948138 DOI: 10.1186/1471-2407-14-127] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 02/12/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute leukemia in early age (EAL) is characterized by acquired genetic alterations such as MLL rearrangements (MLL-r). The aim of this case-controlled study was to investigate whether single nucleotide polymorphisms (SNPs) of IKZF1, ARID5B, and CEBPE could be related to the onset of EAL cases (<24 months-old at diagnosis). METHODS The SNPs (IKZF1 rs11978267, ARID5B rs10821936 and rs10994982, CEBPE rs2239633) were genotyped in 265 cases [169 acute lymphoblastic leukemia (ALL) and 96 acute myeloid leukaemia (AML)] and 505 controls by Taqman allelic discrimination assay. Logistic regression was used to evaluate the association between SNPs of cases and controls, adjusted on skin color and/or age. The risk was determined by calculating odds ratios (ORs) with 95% confidence interval (CI). RESULTS Children with the IKZF1 SNP had an increased risk of developing MLL-germline ALL in white children. The heterozygous/mutant genotype in ARID5B rs10994982 significantly increased the risk for MLL-germline leukemia in white and non-white children (OR 2.60, 95% CI: 1.09-6.18 and OR 3.55, 95% CI: 1.57-8.68, respectively). The heterozygous genotype in ARID5B rs10821936 increased the risk for MLL-r leukemia in both white and non-white (OR 2.06, 95% CI: 1.12-3.79 and OR 2.36, 95% CI: 1.09-5.10, respectively). Furthermore, ARID5B rs10821936 conferred increased risk for MLL-MLLT3 positive cases (OR 7.10, 95% CI:1.54-32.68). Our data do not show evidence that CEBPE rs2239633 confers increased genetic susceptibility to EAL. CONCLUSIONS IKZF1 and CEBPE variants seem to play a minor role in genetic susceptibility to EAL, while ARID5B rs10821936 increased the risk of MLL-MLLT3. This result shows that genetic susceptibility could be associated with the differences regarding MLL breakpoints and partner genes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maria S Pombo-de-Oliveira
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rua André Cavalcanti 37, Rio de Janeiro/RJ 20231-050, Brasil.
| | | |
Collapse
|
50
|
The role of KIR genes and their cognate HLA class I ligands in childhood acute lymphoblastic leukemia. Blood 2014; 123:2497-503. [PMID: 24518758 DOI: 10.1182/blood-2013-11-540625] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Killer cell immunoglobulin-like receptors (KIRs), via interaction with their cognate HLA class I ligands, play a crucial role in the development and activity of natural killer cells. Following recent reports of KIR gene associations in childhood acute lymphoblastic leukemia (ALL), we present a more in-depth investigation of KIR genes and their cognate HLA ligands on childhood ALL risk. Genotyping of 16 KIR genes, along with HLA class I groups C1/C2 and Bw4 supertype ligands, was carried out in 212 childhood ALL cases and 231 healthy controls. Frequencies of KIR genes, KIR haplotypes, and combinations of KIR-HLA ligands were tested for disease association using logistic regression analyses. KIR A/A genotype frequency was significantly increased in cases (33.5%) compared with controls (24.2%) (odds ratio [OR] = 1.57; 95% confidence interval [CI], 1.04-2.39). Stratifying analysis by ethnicity, a significant difference in KIR genotype frequency was demonstrated in Hispanic cases (34.2%) compared with controls (21.9%) (OR = 1.86; 95% CI, 1.05-3.31). Homozygosity for the HLA-Bw4 allele was strongly associated with increased ALL risk exclusively in non-Hispanic white children (OR = 3.93; 95% CI, 1.44-12.64). Our findings suggest a role for KIR genes and their HLA ligands in childhood ALL etiology that may vary among ethnic groups.
Collapse
|