1
|
Patwardhan MV, Kane TQ, Chiong E, Rahmat JN, Mahendran R. Loss of Glutathione-S-Transferase Theta 2 (GSTT2) Modulates the Tumor Microenvironment and Response to BCG Immunotherapy in a Murine Orthotopic Model of Bladder Cancer. Int J Mol Sci 2024; 25:13296. [PMID: 39769061 PMCID: PMC11676541 DOI: 10.3390/ijms252413296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Loss of the glutathione-S-transferases Theta 2 (Gstt2) expression is associated with an improved response to intravesical Mycobacterium bovis, Bacillus Calmette-Guérin (BCG) immunotherapy for non-muscle-invasive bladder cancer (NMIBC) patients who receive fewer BCG instillations. To delineate the cause, Gstt2 knockout (KO) and wildtype (WT) C57Bl/6J mice were implanted with tumors before treatment with BCG or saline. RNA was analyzed via single-cell RNA sequencing (scRNA-seq) and real-time polymerase chain reaction (RT-PCR). BCG induced PD-L1 expression in WT mice bladders, while pro-inflammatory TNF-α was upregulated in KO bladders. ScRNA-seq analysis showed that Gstt2 WT mice bladders had a higher proportion of matrix remodeling fibroblasts, M2 macrophages, and neuronal cells. In KO mice, distinct tumor cell types, activated fibroblasts, and M1 macrophages were enriched in the bladders. In WT bladders, the genes expressed supported tumorigenesis and immunosuppressive PD-L1 expression. In contrast, Gstt2 KO bladders expressed genes involved in inflammation, immune activation, and tumor suppression. An 11-gene signature (Hmga2, Peak 1, Kras, Slc2a1, Ankfn1, Ahnak, Cmss1, Fmo5, Gphn, Plec, Gstt2), derived from the scRNA-seq analysis predicted response in NMIBC patients (The Cancer Genome Atlas (TCGA) database). In conclusion, our results indicate that patients with WT Gstt2 may benefit from anti-PD-L1 checkpoint inhibition therapy.
Collapse
Affiliation(s)
- Mugdha V. Patwardhan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (M.V.P.); (E.C.); (J.N.R.)
| | - Toh Qin Kane
- Genomics and Data Analytics Core, Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Edmund Chiong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (M.V.P.); (E.C.); (J.N.R.)
- Department of Urology, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Juwita Norasmara Rahmat
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (M.V.P.); (E.C.); (J.N.R.)
| | - Ratha Mahendran
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (M.V.P.); (E.C.); (J.N.R.)
| |
Collapse
|
2
|
Boussios S, Sheriff M, Ovsepian SV. Molecular Biology of Cancer-Interplay of Malignant Cells with Emerging Therapies. Int J Mol Sci 2024; 25:13090. [PMID: 39684799 DOI: 10.3390/ijms252313090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer is currently one of the leading causes of death worldwide, and according to data from the World Health Organization reported in 2020, it ranks as the second leading cause of death globally, accounting for 10 million fatalities [...].
Collapse
Affiliation(s)
- Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury CT1 1QU, UK
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King's College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK
| | - Matin Sheriff
- Department of Urology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK
| | - Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Gillingham ME4 4AG, UK
- Faculty of Medicine, Tbilisi State University, Tbilisi 0179, Georgia
| |
Collapse
|
3
|
Rahmat JN, Tham SM, Ong TL, Lim YK, Patwardhan MV, Nee Mani LR, Kamaraj R, Chan YH, Chong TW, Chiong E, Esuvaranathan K, Mahendran R. Glutathione-S-Transferase Theta 2 (GSTT2) Modulates the Response to Bacillus Calmette-Guérin Immunotherapy in Bladder Cancer Patients. Int J Mol Sci 2024; 25:8947. [PMID: 39201633 PMCID: PMC11354831 DOI: 10.3390/ijms25168947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Glutathione-S-transferases (GST) enzymes detoxify xenobiotics and are implicated in response to anticancer therapy. This study evaluated the association of GST theta 1 (GSTT1), GSTT2, and GSTT2B with Mycobacterium bovis Bacillus Calmette-Guérin (BCG) response in non-muscle-invasive bladder cancer treatment. In vitro assessments of GSTT2 knockout (KO) effects were performed using cell lines and dendritic cells (DCs) from GSTT2KO mice. Deletion of GSTT2B, GSTT1, and single-nucleotide polymorphisms in the promoter region of GSTT2 was analysed in patients (n = 205) and healthy controls (n = 150). Silencing GSTT2 expression in MGH cells (GSTT2BFL/FL) resulted in increased BCG survival (p < 0.05) and decreased cellular reactive oxygen species. In our population, there are 24.2% with GSTT2BDel/Del and 24.5% with GSTT2BFL/FL. With ≤ 8 instillations of BCG therapy (n = 51), 12.5% of GSTT2BDel/Del and 53.8% of GSTT2BFL/FL patients had a recurrence (p = 0.041). With ≥9 instillations (n = 153), the disease recurred in 45.5% of GSTT2BDel/Del and 50% of GSTT2BFL/FL. GSTT2FL/FL patients had an increased likelihood of recurrence post-BCG therapy (HR 5.5 [1.87-16.69] p < 0.002). DCs from GSTT2KO mice produced three-fold more IL6 than wild-type DCs, indicating a robust inflammatory response. To summarise, GSTT2BDel/Del patients respond better to less BCG therapy and could be candidates for a reduced surveillance regimen.
Collapse
Affiliation(s)
- Juwita N. Rahmat
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
| | - Sin Mun Tham
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
| | - Ting Li Ong
- School of Engineering, Biomedical Engineering, Temasek Polytechnic, Singapore 529757, Singapore
| | - Yew Koon Lim
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
| | - Mugdha Vijay Patwardhan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
| | - Lata Raman Nee Mani
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
| | - Revathi Kamaraj
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
| | - Yiong Huak Chan
- Biostatistics Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
| | - Tsung Wen Chong
- Department of Urology, Singapore General Hospital, Singapore 169608, Singapore;
- Division of Surgery & Surgical Oncology, National Cancer Center Singapore, Singapore 168583, Singapore
| | - Edmund Chiong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
- Department of Urology, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Kesavan Esuvaranathan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
- Department of Urology, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Ratha Mahendran
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (J.N.R.); (S.M.T.); (Y.K.L.); (M.V.P.); (L.R.N.M.); (R.K.); (K.E.)
| |
Collapse
|
4
|
Chen WC, Brandenburg JT, Choudhury A, Hayat M, Sengupta D, Swiel Y, Babb de Villiers C, Ferndale L, Aldous C, Soo CC, Lee S, Curtis C, Newton R, Waterboer T, Sitas F, Bradshaw D, Abnet CC, Ramsay M, Parker MI, Singh E, Lewis CM, Mathew CG. Genome-wide association study of esophageal squamous cell cancer identifies shared and distinct risk variants in African and Chinese populations. Am J Hum Genet 2023; 110:1690-1703. [PMID: 37673066 PMCID: PMC10577073 DOI: 10.1016/j.ajhg.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) has a high disease burden in sub-Saharan Africa and has a very poor prognosis. Genome-wide association studies (GWASs) of ESCC in predominantly East Asian populations indicate a substantial genetic contribution to its etiology, but no genome-wide studies have been done in populations of African ancestry. Here, we report a GWAS in 1,686 African individuals with ESCC and 3,217 population-matched control individuals to investigate its genetic etiology. We identified a genome-wide-significant risk locus on chromosome 9 upstream of FAM120A (rs12379660, p = 4.58 × 10-8, odds ratio = 1.28, 95% confidence interval = 1.22-1.34), as well as a potential African-specific risk locus on chromosome 2 (rs142741123, p = 5.49 × 10-8) within MYO1B. FAM120A is a component of oxidative stress-induced survival signals, and the associated variants at the FAM120A locus co-localized with highly significant cis-eQTLs in FAM120AOS in both esophageal mucosa and esophageal muscularis tissue. A trans-ethnic meta-analysis was then performed with the African ESCC study and a Chinese ESCC study in a combined total of 3,699 ESCC-affected individuals and 5,918 control individuals, which identified three genome-wide-significant loci on chromosome 9 at FAM120A (rs12379660, pmeta = 9.36 × 10-10), chromosome 10 at PLCE1 (rs7099485, pmeta = 1.48 × 10-8), and chromosome 22 at CHEK2 (rs1033667, pmeta = 1.47 × 10-9). This indicates the existence of both shared and distinct genetic risk loci for ESCC in African and Asian populations. Our GWAS of ESCC conducted in a population of African ancestry indicates a substantial genetic contribution to ESCC risk in Africa.
Collapse
Affiliation(s)
- Wenlong Carl Chen
- National Cancer Registry, National Health Laboratory Service, Johannesburg 2131, South Africa; Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa; Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Jean-Tristan Brandenburg
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Mahtaab Hayat
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Dhriti Sengupta
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Yaniv Swiel
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; School of Electrical & Information Engineering, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Chantal Babb de Villiers
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Lucien Ferndale
- Department of Surgery, Grey's Hospital, Pietermaritzburg 3200, South Africa; College of Health Sciences, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Colleen Aldous
- College of Health Sciences, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4013, South Africa
| | - Cassandra C Soo
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Sang Lee
- Social, Genetic and Development Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, SE5 8AF London, UK; NIHR BioResource Centre Maudsley, South London and Maudsley NHS Foundation Trust, King's College London, SE5 8AF London, UK
| | - Charles Curtis
- Social, Genetic and Development Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, SE5 8AF London, UK; NIHR BioResource Centre Maudsley, South London and Maudsley NHS Foundation Trust, King's College London, SE5 8AF London, UK
| | - Rob Newton
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; University of York, YO10 5DD York, UK
| | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Freddy Sitas
- Burden of Disease Research Unit, South African Medical Research Council, Cape Town 7505, South Africa; Centre for Primary Health Care and Equity, School of Population, University of New South Wales, Sydney, NSW 2052, Australia; Menzies Centre of Health Policy, School of Public Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Debbie Bradshaw
- Burden of Disease Research Unit, South African Medical Research Council, Cape Town 7505, South Africa
| | - Christian C Abnet
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20892, USA
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - M Iqbal Parker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Elvira Singh
- National Cancer Registry, National Health Laboratory Service, Johannesburg 2131, South Africa; School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Cathryn M Lewis
- Social, Genetic and Development Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, SE5 8AF London, UK; Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, SE1 9RT London, UK
| | - Christopher G Mathew
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa; Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, SE1 9RT London, UK.
| |
Collapse
|
5
|
Raeisi Dehkordi S, Luebeck J, Bafna V. FaNDOM: Fast nested distance-based seeding of optical maps. PATTERNS (NEW YORK, N.Y.) 2021; 2:100248. [PMID: 34027500 PMCID: PMC8134938 DOI: 10.1016/j.patter.2021.100248] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/08/2021] [Accepted: 04/01/2021] [Indexed: 12/25/2022]
Abstract
Optical mapping (OM) provides single-molecule readouts of fluorescently labeled sequence motifs on long fragments of DNA, resolved to nucleotide-level coordinates. With the advent of microfluidic technologies for analysis of DNA molecules, it is possible to inexpensively generate long OM data ( > 150 kbp) at high coverage. In addition to scaffolding for de novo assembly, OM data can be aligned to a reference genome for identification of genomic structural variants. We introduce FaNDOM (Fast Nested Distance Seeding of Optical Maps)-an optical map alignment tool that greatly reduces the search space of the alignment process. On four benchmark human datasets, FaNDOM was significantly (4-14×) faster than competing tools while maintaining comparable sensitivity and specificity. We used FaNDOM to map variants in three cancer cell lines and identified many biologically interesting structural variants, including deletions, duplications, gene fusions and gene-disrupting rearrangements. FaNDOM is publicly available at https://github.com/jluebeck/FaNDOM.
Collapse
Affiliation(s)
- Siavash Raeisi Dehkordi
- Department of Computer Science & Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jens Luebeck
- Department of Computer Science & Engineering, University of California, San Diego, La Jolla, CA 92093, USA
- Bioinformatics & Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vineet Bafna
- Department of Computer Science & Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Protein structure analysis of the interactions between SARS-CoV-2 spike protein and the human ACE2 receptor: from conformational changes to novel neutralizing antibodies. Cell Mol Life Sci 2020; 78:1501-1522. [PMID: 32623480 PMCID: PMC7334636 DOI: 10.1007/s00018-020-03580-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/10/2020] [Accepted: 06/22/2020] [Indexed: 12/19/2022]
Abstract
The recent severe acute respiratory syndrome, known as Coronavirus Disease 2019 (COVID-19) has spread so much rapidly and severely to induce World Health Organization (WHO) to declare a state of emergency over the new coronavirus SARS-CoV-2 pandemic. While several countries have chosen the almost complete lock-down for slowing down SARS-CoV-2 spread, the scientific community is called to respond to the devastating outbreak by identifying new tools for diagnosis and treatment of the dangerous COVID-19. With this aim, we performed an in silico comparative modeling analysis, which allows gaining new insights into the main conformational changes occurring in the SARS-CoV-2 spike protein, at the level of the receptor-binding domain (RBD), along interactions with human cells angiotensin-converting enzyme 2 (ACE2) receptor, that favor human cell invasion. Furthermore, our analysis provides (1) an ideal pipeline to identify already characterized antibodies that might target SARS-CoV-2 spike RBD, aiming to prevent interactions with the human ACE2, and (2) instructions for building new possible neutralizing antibodies, according to chemical/physical space restraints and complementary determining regions (CDR) mutagenesis of the identified existing antibodies. The proposed antibodies show in silico high affinity for SARS-CoV-2 spike RBD and can be used as reference antibodies also for building new high-affinity antibodies against present and future coronaviruses able to invade human cells through interactions of their spike proteins with the human ACE2. More in general, our analysis provides indications for the set-up of the right biological molecular context for investigating spike RBD–ACE2 interactions for the development of new vaccines, diagnostic kits, and other treatments based on the targeting of SARS-CoV-2 spike protein.
Collapse
|
7
|
Genetic variants association with cancers in African-based populations: A systematic review. Cancer Epidemiol 2020; 67:101739. [PMID: 32554299 DOI: 10.1016/j.canep.2020.101739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/21/2020] [Accepted: 04/25/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer is the single leading cause of human deaths worldwide. The highest incidence and mortality are recorded from Africa. The last two decades have witnessed extensive research which has led to emerging prognosis and new gene therapy technologies. Cancer therapy in Africa is derived with little input from African population data. While a number of cancer studies on African populations have suggested varied susceptible variant, no comprehensive review of these studies has been undertaken to assess their coverage across Africa. METHODS This study aimed to undertake a review of all molecular genetic studies that interrogated the genetic variants of cancers in African-based populations. Our search methodology was modelled after the Cochrane systematic review protocol, which included MeSH terms and related keywords. RESULTS Ninety-seven articles studying 13 cancer types, were reviewed. 91 articles screened for polymorphisms using PCR-based techniques while three used SNP array, two used whole exome sequencing and one used pyrosequencing. North African (NA) countries undertook 51/97 (53 %) studies on 12/13 (92 %) cancer types while the Sub Saharan Africa (SSA) countries undertook 46/97 (47 %) studies on 7/13 (54 %) cancer types. Twelve out of these thirteen cancer type studies suggested susceptibility to their target polymorphism (p > 0.05). No study replicated or validated variants detected. CONCLUSION Research on genetic determinants in African-based population cancer offers translational benefits. We recommended large scale, multi-national genome association studies using high throughput techniques. SSA needs to receive more attention due to the shortage of this type of study and data in the region.
Collapse
|
8
|
Dai P, Shen D, Shen J, Tang Q, Xi M, Li Y, Li C. The roles of Nrf2 and autophagy in modulating inflammation mediated by TLR4 - NFκB in A549 cell exposed to layer house particulate matter 2.5 (PM 2.5). CHEMOSPHERE 2019; 235:1134-1145. [PMID: 31561304 DOI: 10.1016/j.chemosphere.2019.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/23/2019] [Accepted: 07/01/2019] [Indexed: 06/10/2023]
Abstract
Particulate matter (PM) from layer house has adverse effect on people and chicken respiratory health, which can further influence animal performance and reduce production efficiency. However, little study focus on the respiratory inflammation induced by PM2.5 from layer house and the underlying mechanism also unclear. In this study, human adenocarcinoma alveolar basal epithelial cells (A549 cell) was subjected to the PM2.5 from layer house to evaluate the inflammation reaction caused by PM2.5 and explore the role of Nrf2 and autophagy in regulating the inflammation. Results showed that the viability of A549 cell decreased in a time - and concentration - dependent manner after PM2.5 treatment. TNFα, IL6, and IL8 increased significantly treated with PM2.5 at 12 h. RNA sequencing indicated differentially expressed genes were enriched in immune system process, oxidative stress (OS), endoplasmic reticulum stress (ERS), and autophagy. Further studies showed TLR4 - NFκB p65 signal pathway involved in the inflammation reaction caused by PM2.5. The overexpression of Nrf2 decreased the level of TNFα, IL6, IL8 markedly as well as the level of NFκB p65 and NFκB pp65. OS and ERS were also limited under overactivation of Nrf2 in PM2.5 treated cells. Autophagy induced by PM2.5 promoted the inflammation through increasing the level of NFκB p65 and NFκB pp65. Autophagy deficient strengthened the expression of Nrf2. Collectively, our study revealed Nrf2 prevents inflammation caused by layer house PM2.5 stimulation, however, autophagy exerts a promotive role in TLR4 - NFκB p65 mediating inflammation in A549 cell.
Collapse
Affiliation(s)
- Pengyuan Dai
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Dan Shen
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Jiakun Shen
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Qian Tang
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Mengxue Xi
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Yansen Li
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China
| | - Chunmei Li
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, PR China.
| |
Collapse
|
9
|
Simba H, Kuivaniemi H, Lutje V, Tromp G, Sewram V. Systematic Review of Genetic Factors in the Etiology of Esophageal Squamous Cell Carcinoma in African Populations. Front Genet 2019; 10:642. [PMID: 31428123 PMCID: PMC6687768 DOI: 10.3389/fgene.2019.00642] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 06/18/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Esophageal squamous cell carcinoma (ESCC), one of the most aggressive cancers, is endemic in Sub-Saharan Africa, constituting a major health burden. It has the most divergence in cancer incidence globally, with high prevalence reported in East Asia, Southern Europe, and in East and Southern Africa. Its etiology is multifactorial, with lifestyle, environmental, and genetic risk factors. Very little is known about the role of genetic factors in ESCC development and progression among African populations. The study aimed to systematically assess the evidence on genetic variants associated with ESCC in African populations. Methods: We carried out a comprehensive search of all African published studies up to April 2019, using PubMed, Embase, Scopus, and African Index Medicus databases. Quality assessment and data extraction were carried out by two investigators. The strength of the associations was measured by odds ratios and 95% confidence intervals. Results: Twenty-three genetic studies on ESCC in African populations were included in the systematic review. They were carried out on Black and admixed South African populations, as well as on Malawian, Sudanese, and Kenyan populations. Most studies were candidate gene studies and included DNA sequence variants in 58 different genes. Only one study carried out whole-exome sequencing of 59 ESCC patients. Sample sizes varied from 18 to 880 cases and 88 to 939 controls. Altogether, over 100 variants in 37 genes were part of 17 case-control genetic association studies to identify susceptibility loci for ESCC. In these studies, 25 variants in 20 genes were reported to have a statistically significant association. In addition, eight studies investigated changes in cancer tissues and identified somatic alterations in 17 genes and evidence of loss of heterozygosity, copy number variation, and microsatellite instability. Two genes were assessed for both genetic association and somatic mutation. Conclusions: Comprehensive large-scale studies on the genetic basis of ESCC are still lacking in Africa. Sample sizes in existing studies are too small to draw definitive conclusions about ESCC etiology. Only a small number of African populations have been analyzed, and replication and validation studies are missing. The genetic etiology of ESCC in Africa is, therefore, still poorly defined.
Collapse
Affiliation(s)
- Hannah Simba
- African Cancer Institute, Division of Health Systems and Public Health, Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vittoria Lutje
- Cochrane Infectious Diseases Group, Liverpool, United Kingdom
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,Bioinformatics Unit, South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa.,DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa.,South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa.,Centre for Bioinformatics and Computational Biology, Stellenbosch University, Stellenbosch, South Africa
| | - Vikash Sewram
- African Cancer Institute, Division of Health Systems and Public Health, Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
10
|
Chen WC, Bye H, Matejcic M, Amar A, Govender D, Khew YW, Beynon V, Kerr R, Singh E, Prescott NJ, Lewis CM, Babb de Villiers C, Parker MI, Mathew CG. Association of genetic variants in CHEK2 with oesophageal squamous cell carcinoma in the South African Black population. Carcinogenesis 2019; 40:513-520. [PMID: 30753320 PMCID: PMC6556703 DOI: 10.1093/carcin/bgz026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/18/2018] [Accepted: 02/07/2019] [Indexed: 12/15/2022] Open
Abstract
Oesophageal squamous cell carcinoma (OSCC) has a high incidence in southern Africa and a poor prognosis. Limited information is available on the contribution of genetic variants in susceptibility to OSCC in this region. However, recent genome-wide association studies have identified multiple susceptibility loci in Asian and European populations. In this study, we investigated genetic variants from seven OSCC risk loci identified in non-African populations for association with OSCC in the South African Black population. We performed association studies in a total of 1471 cases and 1791 controls from two study sample groups, which included 591 cases and 852 controls from the Western Cape and 880 cases and 939 controls from the Johannesburg region in the Gauteng province. Thereafter, we performed a meta-analysis for 11 variants which had been genotyped in both studies. A single nucleotide polymorphism in the CHEK2 gene, rs1033667, was significantly associated with OSCC [P = 0.002; odds ratio (OR) = 1.176; 95% confidence interval (CI): 1.06-1.30]. However, single nucleotide polymorphisms in the CASP8/ALS2CR12, TMEM173, PLCE1, ALDH2, ATP1B2/TP53 and RUNX1 loci were not associated with the disease (P > 0.05). The lack of association of six of these loci with OSCC in South African populations may reflect different genetic risk factors in non-African and African populations or differences in the genetic architecture of African genomes. The association at CHEK2, a gene with key roles in cell cycle regulation and DNA repair, in an African population provides further support for the contribution of common genetic variants at this locus to the risk of oesophageal cancer.
Collapse
Affiliation(s)
- Wenlong C Chen
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, School of Pathology, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
| | - Hannah Bye
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Marco Matejcic
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ariella Amar
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Dhiren Govender
- Division of Anatomical Pathology, University of Cape Town, Cape Town, South Africa
| | - Yee Wen Khew
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Victoria Beynon
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Robyn Kerr
- Division of Human Genetics, School of Pathology, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
| | - Elvira Singh
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Natalie J Prescott
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Cathryn M Lewis
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Chantal Babb de Villiers
- Division of Human Genetics, School of Pathology, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
| | - M Iqbal Parker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Christopher G Mathew
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, School of Pathology, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
11
|
Abstract
Esophageal cancer (EC) is an extremely aggressive cancer with one of the highest mortality rates. The cancer is generally only diagnosed at the later stages and has a poor 5-year survival rate due to the limited treatment options. China and South Africa are two countries with a very high prevalence rate of EC. EC rates in South Africa have been on the increase, and esophageal squamous cell carcinoma is the predominant subtype and a primary cause of cancer-related deaths in the black and male mixed ancestry populations in South Africa. The incidence of EC is highest in the Eastern Cape Province, especially in the rural areas such as the Transkei, where the consumption of foods contaminated with Fusarium verticillioides is thought to play a major contributing role to the incidence of EC. China is responsible for almost half of all new cases of EC globally. In China, the prevalence of EC varies greatly. However, the two main areas of high prevalence are the southern Taihang Mountain area (Linxian, Henan Province) and the north Jiangsu area. In both countries, environmental toxins play a major role in increasing the chance that an individual will develop EC. These associative factors include tobacco use, alcohol consumption, nutritional deficiencies and exposure to environmental toxins. However, genetic polymorphisms also play a role in predisposing individuals to EC. These include single-nucleotide polymorphisms that can be found in both protein-coding genes and in non-coding sequences such as miRNAs. The aim of this review is to summarize the contribution of genetic polymorphisms to EC in South Africa and to compare and contrast this to the genetic polymorphisms observed in EC in the most comprehensively studied population group, the Chinese.
Collapse
Affiliation(s)
- Mohammed Alaouna
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rodney Hull
- Research, Innovation & Engagements Portfolio, Mangosuthu University of Technology, Durban, South Africa,
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Zodwa Dlamini
- Research, Innovation & Engagements Portfolio, Mangosuthu University of Technology, Durban, South Africa,
| |
Collapse
|
12
|
Wang S, Zhang J, Jun F, Bai Z. Glutathione S-transferase pi 1 variant and squamous cell carcinoma susceptibility: a meta-analysis of 52 case-control studies. BMC MEDICAL GENETICS 2019; 20:22. [PMID: 30665373 PMCID: PMC6340173 DOI: 10.1186/s12881-019-0750-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022]
Abstract
Background There are several meta-analyses on the genetic relationship between the rs1695 polymorphism within the GSTP1 (glutathione S-transferase pi 1) gene and the risk of different SCC (squamous cell carcinoma) diseases, such as ESCC (oesophageal SCC), HNSCC (head and neck SCC), LSCC (lung SCC), and SSCC (skin SCC). Nevertheless, no unified conclusions have been drawn. Methods Herein, an updated meta-analysis was performed to evaluate the probable impact of GSTP1 rs1695 on the susceptibility to different SCC diseases under six genetic models (allele, carrier, homozygote, heterozygote, dominant, and recessive). Three online databases, namely, PubMed, WOS (Web of Science), and Embase (Excerpta Medica Database), were searched. Results Initially, we obtained a total of 497 articles. Based on our selection criteria, we eventually included 52 case-control studies (9763 cases/15,028 controls) from 47 eligible articles. As shown in the pooling analysis, there was no difference in the risk of overall SCC disease between cases and controls [allele, Pa (P value of association test) = 0.601; carrier, Pa = 0.587; homozygote, Pa = 0.689; heterozygote, Pa = 0.167; dominant, Pa = 0.289; dominant, Pa = 0.548]. Similar results were obtained after stratification by race (Asian/Caucasian), genotyping, control source, and disease type (ESCC/HNSCC/LSCC/SSCC) (all Pa > 0.05). Conclusion The rs1695 polymorphism within the GSTP1 gene is not associated with the risk of overall SCC or a specific SCC type, including ESCC, HNSCC, LSCC, and SSCC.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Plastic and Burn Surgery, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Jingqi Zhang
- Department of Plastic and Burn Surgery, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Fan Jun
- Department of Plastic and Burn Surgery, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Zhijie Bai
- Department of Urology Surgery, Tianjin First Center Hospital, Tianjin, 300192, China.
| |
Collapse
|
13
|
High-performance liquid chromatography-based assay for glutathione transferase theta 2 activity: Application to characterize interindividual variability in human liver fractions. J Pharm Biomed Anal 2018; 156:181-188. [DOI: 10.1016/j.jpba.2018.04.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/13/2018] [Accepted: 04/22/2018] [Indexed: 02/06/2023]
|
14
|
He F, Liu C, Zhang R, Hao Z, Li Y, Zhang N, Zheng L. Association between the Glutathione-S-transferase T1 null genotype and esophageal cancer susceptibility: a meta-analysis involving 11,163 subjects. Oncotarget 2018; 9:15111-15121. [PMID: 29599931 PMCID: PMC5871102 DOI: 10.18632/oncotarget.24534] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/21/2017] [Indexed: 02/07/2023] Open
Abstract
Background Glutathione-S-Transferase T1 (GSTT1) null genotype has been shown to be associated with the risk of esophageal cancer. However, the results remain inconsistent. Thus a comprehensive meta-analysis was conducted to assess the strength of association between GSTT1 null genotype and the risk of esophageal cancer. Materials and Methods A literature search of PubMed, Embase, China National Knowledge Infrastructure (CNKI) and Wanfang databases up to March 31, 2017 was conducted and 30 eligible articles with 4482 cases and 6681 controls were finally recruited. The strength of correlation between GSTT1 polymorphism and the susceptibility of esophageal cancer was assessed by the crude odds ratios (ORs) with 95% confidence intervals (CIs). Subgroup analyses and sensitivity analyses were performed to further identify the association. Results GSTT1 null genotype significantly increased the risk of esophageal cancer (OR = 1.20; 95% CI 1.04-1.40; P < 0.05). In a subgroup analysis by ethnicity, GSTT1 null genotype was correlated with a significantly increased risk of esophageal cancer among Asians (OR = 1.33; 95% CI 1.12-1.58; P < 0.05), instead of Caucasians or Africans (OR = 0.91; 95% CI 0.65-1.26; P > 0.05 for Caucasians and OR = 1.32; 95% CI 0.98-1.77; P > 0.05 for Africans). In the analysis by histological type, GSTT1 null genotype was correlated with a significantly increased risk of esophageal squamous cell carcinoma (OR = 1.34; 95% CI 1.12-1.61; P < 0.05), particularly among Asians (OR = 1.54; 95% CI 1.30-1.82; P < 0.05), but not among Caucasians or Africans (OR = 0.87; 95% CI 0.48-1.57; P > 0.05 for Caucasians and OR = 1.32; 95% CI 0.98-1.77; P > 0.05 for Africans). In addition, there is no significant correlation between GSTT1 null genotype and the risk of esophageal adenocarcinoma (OR = 0.98; 95% CI 0.71-1.35; P > 0.05). Conclusions Our findings demonstrate that GSTT1 null genotype significantly increases esophageal cancer risk, particularly in Asians.
Collapse
Affiliation(s)
- Feng He
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Changyu Liu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ruijie Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhipeng Hao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yangkai Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Zheng
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| |
Collapse
|
15
|
Shi H, Wang Z. Atypical microdeletion in 22q11 deletion syndrome reveals new candidate causative genes: A case report and literature review. Medicine (Baltimore) 2018; 97:e9936. [PMID: 29465581 PMCID: PMC5841983 DOI: 10.1097/md.0000000000009936] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
RATIONALE 22q11 deletion syndrome, the most common chromosomal microdeletion disease, is caused by megabase-sized deletions on chromosome 22q11.2. It is characterized by a wide spectrum of congenital anomalies in velopharyngeal and facial, cardiac, genitourinary, vertebroskeletal, respiratory, digestive, and central nervous systems. Phenotype-genotype studies have revealed several causative genes that regulate the development of the third and fourth pharyngeal arches in human. However, the exact pathogenesis of this syndrome remains unknown. Herein, we report a case of 22q11 deletion syndrome with an atypical microdeletion of 125 kb. PATIENT CONCERNS A 15-year-old Chinese girl presented with symptoms of facial dysmorphia, cardiac defects, velopharyngeal insufficiency, splenomegaly, immunodeficiency, and thrombocytopenia. DIAGNOSES Microarray analysis revealed a 22q11.23 deletion of 125 kb (chromosome 22: 24276973-24402263), suggesting the diagnosis of 22q11 deletion syndrome. The haploinsufficient genes included GSTT2B, GSTT2, DDTL, DDT, GSTTP1, LOC391322, GSTT1, and GSTTP2. INTERVENTIONS The patient was administrated glucocorticoids and calcium supplements. OUTCOMES No epistaxis or petechiae episode occurred during the follow-up; her platelet count ranged between 60 × 10 and 80 × 10/L. LESSONS Although none of the previous reported causative genes were affected in the patient, her clinical manifestations were typical of 22q11 deletion syndrome, apart from her progressive splenomegaly. This case indicated 8 new candidate pathogenic genes for 22q11 deletion syndrome. Given that the loss of these genes was sufficient to induce 22q11DS defects, whether these genes directly influence the pathogenesis of 22q11DS or through interactions with known hotspot mutations is worthy of research.
Collapse
Affiliation(s)
- Huiping Shi
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zhaoyue Wang
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
16
|
Ma L, Lan B, Guo L, Nong S, Huang C, Wu Q, Huang Z. GSTM1 and GSTT1 Gene Polymorphisms, Gene-Gene Interaction, and Esophageal Carcinoma Risk: Evidence from an Updated Meta-Analysis. Genet Test Mol Biomarkers 2018; 22:11-19. [PMID: 29215312 DOI: 10.1089/gtmb.2017.0137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Liping Ma
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Biyang Lan
- Department of General Surgery, Guangxi Minzu Hospital, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Lingxiao Guo
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Shaoyun Nong
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Cuibo Huang
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Qiulong Wu
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| | - Zhihu Huang
- Department of Clinical Laboratory, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Abstract
In first part of this study, a systematic review was designed to explore the involvement of CYP1A1 and GSTP1 genes in breast cancerogenesis. Based on systematic review, we designed a study to screen CYP1A1 and GSTP1 genes for mutation and their possible association with breast carcinogenesis. A total of 400 individuals were collected and analyzed by PCR-SSCP. After sequence analysis of coding region of CYP1A1 we identified eleven mutations in different exons of respective gene. Among these eleven mutations, ~3 folds increased breast cancer risk was found associated with Asp82Glu mutation (OR 2.99; 95% CI 1.26-7.09), with Ser83Thr mutation (OR 2.99; 95% CI 1.26-7.09) and with Glu86Ala mutation (OR 3.18; 95% CI 1.27-7.93) in cancer patients compared to controls. Furthermore, ~4 folds increase in breast cancer risk was found associated with Asp347Glu, Phe398Tyr and 5178delT mutations (OR 3.92; 95% CI 1.35-11.3) in patients compared to controls. The sequence analysis of GSTP1 resulted in identification of total five mutations. Among these five mutations, ~3 folds increase in breast cancer risk was observed associated with 1860G>A mutation, with 1861-1876delCAGCCCTCTGGAGTGG mutation (OR 2.70; 95% CI 1.10-6.62) and with 1861C>A mutation (OR 2.97; 95% CI 1.01-8.45) in cancer patients compared to controls. Furthermore, ~5 folds increase in breast cancer risk was associated with 1883G>T mutation (OR 4.75; 95% CI 1.46-15.3) and ~6 folds increase in breast cancer risk was found associated with Iso105Val mutation (OR 6.43; 95% CI 1.41-29.3) in cancer patients compared to controls. Our finding, based on systematic review and experimental data suggest that the polymorphic CYP1A1 and GSTP1 genes may contribute to risk of developing breast cancer.
Collapse
|
18
|
Lu L, Pandey AK, Houseal MT, Mulligan MK. The Genetic Architecture of Murine Glutathione Transferases. PLoS One 2016; 11:e0148230. [PMID: 26829228 PMCID: PMC4734686 DOI: 10.1371/journal.pone.0148230] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 01/14/2016] [Indexed: 12/17/2022] Open
Abstract
Glutathione S-transferase (GST) genes play a protective role against oxidative stress and may influence disease risk and drug pharmacokinetics. In this study, massive multiscalar trait profiling across a large population of mice derived from a cross between C57BL/6J (B6) and DBA2/J (D2)—the BXD family—was combined with linkage and bioinformatic analyses to characterize mechanisms controlling GST expression and to identify downstream consequences of this variation. Similar to humans, mice show a wide range in expression of GST family members. Variation in the expression of Gsta4, Gstt2, Gstz1, Gsto1, and Mgst3 is modulated by local expression QTLs (eQTLs) in several tissues. Higher expression of Gsto1 in brain and liver of BXD strains is strongly associated (P < 0.01) with inheritance of the B6 parental allele whereas higher expression of Gsta4 and Mgst3 in brain and liver, and Gstt2 and Gstz1 in brain is strongly associated with inheritance of the D2 parental allele. Allele-specific assays confirmed that expression of Gsto1, Gsta4, and Mgst3 are modulated by sequence variants within or near each gene locus. We exploited this endogenous variation to identify coexpression networks and downstream targets in mouse and human. Through a combined systems genetics approach, we provide new insight into the biological role of naturally occurring variants in GST genes.
Collapse
Affiliation(s)
- Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Ashutosh K. Pandey
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
| | - M. Trevor Houseal
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
| | - Megan K. Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
- * E-mail:
| |
Collapse
|
19
|
Identification of transposable element-mediated deletions in 27 Korean individuals based on whole genome sequencing data. Genes Genomics 2016. [DOI: 10.1007/s13258-015-0370-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
20
|
Hu L, Wu Y, Guan X, Liang Y, Yao X, Tan D, Bai Y, Xiong G, Yang K. Germline copy number loss of UGT2B28 and gain of PLEC contribute to increased human esophageal squamous cell carcinoma risk in Southwest China. Am J Cancer Res 2015; 5:3056-3071. [PMID: 26693059 PMCID: PMC4656730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/02/2015] [Indexed: 06/05/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers worldwide with poor prognosis. Thus, identification of predictive biomarkers for early diagnosis and intervention is needed to improve patients' survival. Research shows that heritable mutations, such as single nucleotide polymorphisms (SNPs), contribute to human cancer susceptibility significantly. However, the association of copy number variations (CNVs), another major source of genetic variation, with ESCC risk remains poorly clarified. In this study, we aimed to identify ESCC risk-related CNVs based on candidate-gene strategy in a case-control study. A meta-analysis was first performed to identify the most variable chromosome regions of ESCC tissues. Bioinformatic analysis and dual-luciferase reporter assays were carried out to evaluate the properties of all recorded CNVs located on these regions. Six candidate CNVs located within well-known oncogenes and detoxification-associated enzymes were enrolled in the final analysis. A newly developed multiplex gene copy number quantitation method AccuCopy(TM) was employed to simultaneously genotype all six candidate sites in 404 ESCC patients and 402 cancer-free controls from Southwest China, and in 42 ESCC tissues. qRT-PCR was performed to measure UGT2B28 mRNA in cancerous and corresponding normal tissues. Unconditional logistic regression was applied to test association between germline CNV genotypes and ESCC risk. Relationship between germline copy number variation and somatic copy number alterations was further analyzed. Finally we found that copy number loss of UDP-glucuronosyltransferase family 2, polypeptide B28 (UGT2B28) and gain of plectin (PLEC) conferred increased ESCC risk (Adjusted OR = 2.085, 95% CI = 1.493-2.912, P < 0.001 for UGT2B28. Adjusted OR = 3.725, 95% CI = 1.026-13.533, P = 0.046 for PLEC). mRNA level was lower in UGT2B28 loss genotyped esophageal tissues than in two-copy tissues, indicating that UGT2B28 loss genotypes modify ESCC susceptibility perhaps by decreasing UGT2B28 expression level and enzyme activity. In addition, an association was drawn between germline copy number variations and somatic alterations for PLEC, UGT2B17 and UGT2B28, but not for other candidate loci.
Collapse
Affiliation(s)
- Liwen Hu
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical UniversityChongqing, PR China
- Department of Cardiothoracic Surgery, Jinling Hospital, School of Clinical Medicine, Nanjing UniversityNanjing, Jiangsu Province, PR China
| | - Yuanyuan Wu
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical UniversityChongqing, PR China
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical UniversityChongqing, PR China
| | - Yan Liang
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical UniversityChongqing, PR China
| | - Xinyue Yao
- Department of Microbiology, College of Basic Medical Science, Third Military Medical UniversityChongqing, PR China
| | - Deli Tan
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical UniversityChongqing, PR China
| | - Yun Bai
- Department of Medical Genetics, College of Basic Medical Science, Third Military Medical UniversityChongqing, PR China
| | - Gang Xiong
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical UniversityChongqing, PR China
| | - Kang Yang
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical UniversityChongqing, PR China
| |
Collapse
|
21
|
Abstract
Esophageal cancer (EC) is one of the most common malignancies in low- and medium-income countries and represents a disease of public health importance because of its poor prognosis and high mortality rate in these regions. The striking variation in the prevalence of EC among different ethnic groups suggests a significant contribution of population-specific environmental and dietary factors to susceptibility to the disease. Although individuals within a demarcated geographical area are exposed to the same environment and share similar dietary habits, not all of them will develop the disease; thus genetic susceptibility to environmental risk factors may play a key role in the development of EC. A wide range of xenobiotic-metabolizing enzymes are responsible for the metabolism of carcinogens introduced via the diet or inhaled from the environment. Such dietary or environmental carcinogens can bind to DNA, resulting in mutations that may lead to carcinogenesis. Genes involved in the biosynthesis of these enzymes are all subject to genetic polymorphisms that can lead to altered expression or activity of the encoded proteins. Genetic polymorphisms may, therefore, act as molecular biomarkers that can provide important predictive information about carcinogenesis. The aim of this review is to discuss our current knowledge on the genetic risk factors associated with the development of EC in different populations; it addresses mainly the topics of genetic polymorphisms, gene-environment interactions, and carcinogenesis. We have reviewed the published data on genetic polymorphisms of enzymes involved in the metabolism of xenobiotics and discuss some of the potential gene-environment interactions underlying esophageal carcinogenesis. The main enzymes discussed in this review are the glutathione S-transferases (GSTs), N-acetyltransferases (NATs), cytochrome P450s (CYPs), sulfotransferases (SULTs), UDP-glucuronosyltransferases (UGTs), and epoxide hydrolases (EHs), all of which have key roles in the detoxification of environmental and dietary carcinogens. Finally, we discuss recent advances in the study of genetic polymorphisms associated with EC risk, specifically with regard to genome-wide association studies, and examine possible challenges of case-control studies that need to be addressed to better understand the interaction between genetic and environmental factors in esophageal carcinogenesis.
Collapse
Affiliation(s)
- Marco Matejcic
- a International Centre for Genetic Engineering and Biotechnology, Cape Town Component , Observatory , Cape Town , South Africa , and
| | | |
Collapse
|
22
|
Iorio A, Polimanti R, Calandro M, Graziano ME, Piacentini S, Bucossi S, Squitti R, Lazzarin N, Scano G, Limbruno GM, Manfellotto D, Fuciarelli M. Explorative genetic association study of GSTT2B copy number variant in complex disease risks. Ann Hum Biol 2015. [PMID: 26207597 DOI: 10.3109/03014460.2015.1049206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Glutathione S-transferases (GSTs) are the main phase II enzymes involved in cellular detoxification. Through phase I and phase II detoxification reactions, the cell is able to detoxify endogenous and exogenous toxic compounds. AIMS This study focused attention on the GSTT2B copy number variant (CNV) in order to explore its involvement in the genetic pre-disposition to asthma, Alzheimer's disease (AD), allergic rhinitis (AR), essential hypertension (EH), hypothyroidism and recurrent miscarriage (RM). METHODS The study population consists of 1225 individuals divided into six case-control groups. The genotyping of the GSTT2B CNV was performed by using a duplex-PCR. Odds Ratios (ORs) were calculated, adjusting for the confounding variables, to estimate the association between GSTT2B CNV and the disease status. RESULTS The χ(2)-test and ORs did not show any association between this genetic marker and pathological phenotypes. CONCLUSION The data highlights that GSTT2B CNV is not associated with the investigated complex diseases in Italian patients. However, further investigations are necessary to replicate these findings in larger sample sizes and to explore other health-related phenotypes.
Collapse
Affiliation(s)
- Andrea Iorio
- a Department of Biology , University of Rome 'Tor Vergata' , Rome , Italy
| | - Renato Polimanti
- b Department of Psychiatry , Yale University School of Medicine , West Haven , CT , USA
| | - Melania Calandro
- a Department of Biology , University of Rome 'Tor Vergata' , Rome , Italy
| | | | - Sara Piacentini
- a Department of Biology , University of Rome 'Tor Vergata' , Rome , Italy
| | - Serena Bucossi
- c Department of Clinical Neuroscience , AFaR - 'San Giovanni Calibita' Fatebenefratelli Hospital , Isola Tiberina , Rome , Italy .,d Department of Neurology , 'Campus Bio-Medico' University , Rome , Italy
| | - Rosanna Squitti
- c Department of Clinical Neuroscience , AFaR - 'San Giovanni Calibita' Fatebenefratelli Hospital , Isola Tiberina , Rome , Italy .,e Laboratorio Neurodegenerazione , IRCCS San Raffaele Pisana , Rome , Italy
| | | | - Giuseppina Scano
- a Department of Biology , University of Rome 'Tor Vergata' , Rome , Italy
| | - Giancarlo Maria Limbruno
- g Clinical Pathology Department , AFaR - 'San Giovanni Calibita' Fatebenefratelli Hospital , Rome , Italy
| | | | - Maria Fuciarelli
- a Department of Biology , University of Rome 'Tor Vergata' , Rome , Italy
| |
Collapse
|
23
|
Tan X, Chen M. Association between glutathione S-transferases P1 Ile105Val polymorphism and susceptibility to esophageal cancer: evidence from 20 case-control studies. Mol Biol Rep 2015; 42:399-408. [PMID: 25280543 DOI: 10.1007/s11033-014-3781-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 09/24/2014] [Indexed: 02/06/2023]
Abstract
Available epidemiological studies had estimated the correlation between glutathione S-transferases P1 (GSTP1) Ile105Val polymorphism and esophageal cancer (EC) risk. However, the conclusions were controversial and inconclusive. An updated meta-analysis was conducted to explore whether GSTP1 polymorphism could be contributed to the EC risk. Ultimately, a total of 2,992 cases and 4,758 controls from 20 previous studies were included. Crude odds ratios (ORs) with 95 % confidence intervals (95 % CIs) were used to assess the strength of the associations. Pooled results suggested that GSTP1 Ile105Val polymorphism significantly increased the risk of developing EC in Caucasians under three genetic models (G vs. A, OR 1.146, 95 % CI 1.031-1.275, P = 0.012, I(2) = 30.40 %; GA vs. AA, OR 1.208, 95 % CI 1.036-1.408, P = 0.016, I(2) = 50.30 %; GG+GA vs. AA, OR 1.219, 95 % CI 1.053-1.410, P = 0.008, I(2) = 44.50 %). However, no significant correlation was found in Asians, African and mixed ethnicities analyses. Moreover, similar results were detected for any genetic model in esophageal squamous cell carcinoma and esophageal adenocarcinoma when stratifying for pathologic types. This meta-analysis provides new evidences that GSTP1 Ile105Val gene polymorphism contributes to EC susceptibility in Caucasians.
Collapse
Affiliation(s)
- Xiang Tan
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China,
| | | |
Collapse
|
24
|
Song Y, Du Y, Zhou Q, Ma J, Yu J, Tao X, Zhang F. Association of GSTP1 Ile105Val polymorphism with risk of esophageal cancer: a meta-analysis of 21 case-control studies. Int J Clin Exp Med 2014; 7:3215-3224. [PMID: 25419352 PMCID: PMC4238532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 08/28/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND The association of glutathione s-transferase P1 (GSTP1) Ile105Val polymorphism with risk of esophageal cancer (EC) has been evaluated in many studies; however, the results from these studies are controversial. Thus, further analysis on association between GSTP1 Ile105Val polymorphism and risk of EC is needed among a larger study population. METHOD We searched the relevant electronic databases and performed a meta-analysis based on 21 published case-control studies. The Chi-square based I(2)-statistic test was performed to evaluate possible heterogeneity across the studies. Additionally, random-effects models were used to calculate crude pooled odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS Overall, this meta-analysis did support a significant association between GSTP1 Ile105Val polymorphism and risk of EC (pooled OR 1.25, 95% CI, 1.05-1.49). Furthermore, the stratified analysis showed that, in comparison to GSTP1 Ile105Val Ile/Ile genotype, the Val/Val genotype was significantly associated with risk of esophageal squamous cell carcinoma (ESCC) (pooled OR 1.45, 95% CI, 1.07-1.96), particularly in the Caucasian population (pooled OR 1.41, 95% CI, 1.01-1.95). Such a significant association was not observed for esophageal adenocarcinoma (EAC) patients or subjects of an Asian ethnicity. Moreover, substantial evidence of heterogeneity among the studies was not observed. CONCLUSION The results from this meta-analysis support a significant association between the GSTP1 Ile105Val polymorphism and risk of EC, particularly in a subgroup with ESCC and in the Caucasian population. Further studies with larger sample sizes are needed to validate our findings.
Collapse
Affiliation(s)
- Yipeng Song
- Department of Radiation Oncology, Affiliated Yantai Yuhuangding Hospital, Qingdao UniversityYantai 264000, China
| | - Yuanna Du
- Department of Radiation Oncology, Affiliated Yantai Yuhuangding Hospital, Qingdao UniversityYantai 264000, China
| | - Qi Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital, Soochow UniversityChangzhou 213003, Jiangsu Province, China
| | - Jinbo Ma
- Department of Radiation Oncology, Affiliated Yantai Yuhuangding Hospital, Qingdao UniversityYantai 264000, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer HospitalJinan 250012, Shandong, China
| | - Xiaofeng Tao
- Radiology Department of Shanghai Ninth People’s Hospital Affiliated Shanghai Jiao Tong University School of MedicineShanghai 200011, China
| | - Fenghua Zhang
- Department of General Surgery, Hebei General HospitalShijiazhuang 050051, Hebei, China
| |
Collapse
|
25
|
Godoy FR, Costa EOA, da Silva Reis AA, Batista MP, de Melo AV, Gonçalves MW, Cruz AS, de Araújo Melo CO, Minasi LB, Ribeiro CL, da Cruz AD, de Melo E Silva D. Do GSTT1 and GSTM1 polymorphisms influence intoxication events in individuals occupationally exposed to pesticides? ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2014; 21:3706-3712. [PMID: 24281680 DOI: 10.1007/s11356-013-2349-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/04/2013] [Indexed: 06/02/2023]
Abstract
This study evaluated the variability of GSTM1 and GSTT1 polymorphisms in individuals occupationally exposed to pesticides in ten Goias municipalities that present intense agricultural activity. We evaluated blood samples of 235 individuals, which 120 were rural workers occupationally exposed to pesticides and 115 formed the control group, analyzing GST polymorphisms by quantitative polymerase chain reaction (qPCR).The exposed group consisted of 111 men and nine women only getting an average of 39 ± 9 years. These workers were from ten rural municipalities situated at Goias state. It was found that 18 % of the exposed individuals had the GSTT1 null genotype and 49 % had the GSTM1 null genotype, and 10 % had both null genotypes. Data as intoxication (42 %), use of Personal Protection Equipment (PPE; 52 %) and if the worker prepared the pesticide (7 %), or if just applied the pesticide (22 %) or if the worker prepared and applied (71 %) have all been correlated with genetic polymorphisms. There were no statistically significant differences between the GSTM1 and GSTT1 polymorphisms between control and exposed groups. Finally, we could not associate a null GSTT1 or null GSTM1 polymorphisms or both to intoxication events caused by pesticides, but instead we presented the importance to use PPE to prevent such harm, once we found a statistically significant association between the use of PPE and events of intoxication (p ≤ 0.001).
Collapse
|
26
|
Iorio A, Piacentini S, Polimanti R, De Angelis F, Calderon R, Fuciarelli M. Functional variability of glutathione S-transferases in basque populations. Am J Hum Biol 2014; 26:361-6. [DOI: 10.1002/ajhb.22520] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/11/2014] [Accepted: 01/21/2014] [Indexed: 01/12/2023] Open
Affiliation(s)
- Andrea Iorio
- Department of Biology; University of Rome “Tor Vergata,”; Rome Italy
- Clinical Pathophysiology Center; AFaR-“San Giovanni Calibita” Fatebenefratelli Hospital; Isola Tiberina Rome Italy
| | - Sara Piacentini
- Department of Biology; University of Rome “Tor Vergata,”; Rome Italy
| | - Renato Polimanti
- Department of Biology; University of Rome “Tor Vergata,”; Rome Italy
| | - Flavio De Angelis
- Department of Biology; University of Rome “Tor Vergata,”; Rome Italy
| | - Rosario Calderon
- Departamento de Zoologia y Antropologìa Fìsica; Facultad de Biologìa, Universidad Complutense; Madrid Spain
| | - Maria Fuciarelli
- Department of Biology; University of Rome “Tor Vergata,”; Rome Italy
| |
Collapse
|
27
|
Dura P, Salomon J, Te Morsche RHM, Roelofs HMJ, Kristinsson JO, Wobbes T, Witteman BJM, Tan ACITL, Drenth JPH, Peters WHM. No role for glutathione S-transferase genotypes in Caucasian esophageal squamous cell or adenocarcinoma etiology: an European case-control study. BMC Gastroenterol 2013; 13:97. [PMID: 23731957 PMCID: PMC3706247 DOI: 10.1186/1471-230x-13-97] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 05/22/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Identifying and monitoring high-risk patients can aid the prevention of esophageal cancer (EC). The interaction of environmental risk factor exposure and genetic susceptibility may contribute to the etiology of EC. Biotransformation enzymes such as Glutathione S-Transferases (GSTs ) detoxify mutagenic and genotoxic compounds and therefore control the rate of detoxification of carcinogens. Functional polymorphisms in the genes coding for GSTs alter their enzyme activity in vitro, and were reported to modify EC risk in Asians. We hypothesized that altered enzyme activity GST genotypes influence the susceptibility for esophageal adeno- (EAC) and squamous cell carcinoma (ESCC) in Caucasians. METHODS We performed a case-control study including 440 Caucasian patients with EC and 592 healthy Caucasian controls matched for age and sex. Functional polymorphisms were selected and genotypes were determined in GST classes Alpha, Mu, Theta and Pi by means of polymerase chain reaction. Genotypes were classified into predicted high, intermediate and low enzyme activity categories based on in vitro activity data. The distribution of the activity genotypes were compared between patients with EAC or ESCC, and controls. Odds ratios (OR) with 95% confidence intervals (CI) were calculated by logistic regression analyses. Gene-gene interactions were tested and for comparison purposes, the predicted low and intermediate activity genotypes were combined. Genotypes with similar risks for EAC or ESCC were combined and analyzed for multiplicative effects. RESULTS Our analyses includes 327 patients with EAC and 106 patients with ESCC. Low or intermediate activity enzyme genotypes for GSTM1, GSTA1, GSTP1 I105V and A114V as well as for GSTT1, did not significantly modify the risk for ESCC or EAC in our Dutch population. CONCLUSION Functional genotypes in GST genes are not involved in EAC or ESCC susceptibility in Caucasians, in contrast to results on ESCC from Asia or Africa.
Collapse
Affiliation(s)
- Polat Dura
- Department of Gastroenterology, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cai Y, Wang J. Significant association of glutathione S-transferase T1 null genotype with esophageal cancer risk: a meta-analysis. Mol Biol Rep 2012; 40:2397-403. [PMID: 23238916 DOI: 10.1007/s11033-012-2320-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 11/19/2012] [Indexed: 12/27/2022]
Abstract
Recent studies on the association between glutathione S-transferase T1 (GSTT1) polymorphism and risk of esophageal cancer showed inconclusive results. To clarify this possible association, we conducted a meta-analysis of published studies. Data were collected from the following electronic databases: Pubmed, Embase, and Chinese Biomedical Database (CBM). The odds ratio (OR) and its 95 % confidence interval (95 % CI) was used to assess the strength of this association. We summarized the data on the association between GSTT1 null genotype and risk of esophageal cancer in the overall population, and performed subgroup analyses by ethnicity. Finally, a total of 24 independent studies including a total of 7,801 subjects (2,965 cases and 4,836 controls) were eligible for meta-analysis. In the overall analysis, there was no significant association between GSTT1 null genotype and esophageal cancer risk (OR = 1.15, 95 % CI 0.99-1.33, P = 0.067). However, meta-analysis of adjusted ORs showed a significant association between GSTT1 null genotype and increased risk of esophageal cancer (OR = 1.30, 95 % CI 1.08-1.56, P = 0.005). Subgroup analyses by ethnicity showed there was an obvious association between GSTT1 null genotype and increased risk of esophageal cancer in East Asians (OR = 1.24, 95 % CI 1.10-1.39, P < 0.001), but not in Caucasians (OR = 0.89, 95 % CI 0.71-1.11, P = 0.300). There was no obvious risk of publication bias in this meta-analysis (Egger's test, P = 0.784). This meta-analysis demonstrates that GSTT1 null genotype is independently associated with increased risk of esophageal cancer, and a race-specific effect may exist in this association.
Collapse
Affiliation(s)
- Yong Cai
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai, 200433, China.
| | | |
Collapse
|
29
|
Board PG, Menon D. Glutathione transferases, regulators of cellular metabolism and physiology. Biochim Biophys Acta Gen Subj 2012. [PMID: 23201197 DOI: 10.1016/j.bbagen.2012.11.019] [Citation(s) in RCA: 275] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The cytosolic glutathione transferases (GSTs) comprise a super family of proteins that can be categorized into multiple classes with a mixture of highly specific and overlapping functions. SCOPE OF REVIEW The review covers the genetics, structure and function of the human cytosolic GSTs with particular attention to their emerging roles in cellular metabolism. MAJOR CONCLUSIONS All the catalytically active GSTs contribute to the glutathione conjugation or glutathione dependant-biotransformation of xenobiotics and many catalyze glutathione peroxidase or thiol transferase reactions. GSTs also catalyze glutathione dependent isomerization reactions required for the synthesis of several prostaglandins and steroid hormones and the catabolism of tyrosine. An increasing body of work has implicated several GSTs in the regulation of cell signaling pathways mediated by stress-activated kinases like Jun N-terminal kinase. In addition, some members of the cytosolic GST family have been shown to form ion channels in intracellular membranes and to modulate ryanodine receptor Ca(2+) channels in skeletal and cardiac muscle. GENERAL SIGNIFICANCE In addition to their well established roles in the conjugation and biotransformation of xenobiotics, GSTs have emerged as significant regulators of pathways determining cell proliferation and survival and as regulators of ryanodine receptors that are essential for muscle function. This article is part of a Special Issue entitled Cellular functions of glutathione.
Collapse
Affiliation(s)
- Philip G Board
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | | |
Collapse
|
30
|
Shi Y, Luo GJ, Zhang L, Shi J, Zhang DQ, Chen JM, Chen XB, Li ZD, Zhao Q. Interaction between alcohol consumption and CYP 2C19 gene polymorphism in relation to oesophageal squamous cell carcinoma. PLoS One 2012; 7:e43412. [PMID: 22984423 PMCID: PMC3439450 DOI: 10.1371/journal.pone.0043412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 07/20/2012] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES The purpose of this study is to explore the relationship between the interactions of CYP2C19 gene polymorphisms and several environmental factors and oesophageal squamous cell carcinoma (OSCC). METHODS In a case-control study of OSCC patients (n = 350) and healthy controls (n = 350), we investigated the roles of polymorphism in the CYP2C19 gene by the use of polymerase chain reaction--restriction fragment length polymorphism (PCR - RFLP) analysis. RESULTS The CYP2C19(*)3 AG+AA genotype was significantly more prevalent in OSCC patients (10.0% versus 3.43%; P<0.01). Multiple logistic regression analysis showed drinking (OR: 5.603, 95% CI: 3.431-11.112; P = 0.005) and smoking (OR: 4.341, 95% CI: 3.425-10.241; P = 0.001) was the independent risk factor of OSCC respectively, and there were significant interaction between CYP2C19(*)3 and drinking (OR: 8.747, 95% CI: 6.321-18.122; P = 0.009). CONCLUSIONS The CYP2C19(*)3 polymorphism and OSCC were synergistically and significantly associated in Chinese Han patients.
Collapse
Affiliation(s)
- Yun Shi
- Department of Chest Surgery; Kunming General Hospital of Chengdu Military Region, Kunming, Yunnan Province, China
| | - Guo-jun Luo
- Department of Chest Surgery; Kunming General Hospital of Chengdu Military Region, Kunming, Yunnan Province, China
- * E-mail:
| | - Li Zhang
- Department of Chest Surgery; Kunming General Hospital of Chengdu Military Region, Kunming, Yunnan Province, China
| | - Ji Shi
- Department of Chest Surgery; Kunming General Hospital of Chengdu Military Region, Kunming, Yunnan Province, China
| | - Dao-quan Zhang
- Department of Chest Surgery, The Sixtieth Central Hospital of the People's Liberation Army, Dali, Yunnan Province, China
| | - Jian-min Chen
- Department of Chest Surgery, The Fifty-ninth Central Hospital of the People's Liberation Army, Kaiyuan, Yunnan Province, China
| | - Xiao-bo Chen
- Department of Chest Surgery; Kunming General Hospital of Chengdu Military Region, Kunming, Yunnan Province, China
| | - Zhuo-dong Li
- Department of Chest Surgery; Kunming General Hospital of Chengdu Military Region, Kunming, Yunnan Province, China
| | - Qing Zhao
- Department of Chest Surgery; Kunming General Hospital of Chengdu Military Region, Kunming, Yunnan Province, China
| |
Collapse
|
31
|
Bye H, Prescott NJ, Lewis CM, Matejcic M, Moodley L, Robertson B, Rensburg CV, Parker MI, Mathew CG. Distinct genetic association at the PLCE1 locus with oesophageal squamous cell carcinoma in the South African population. Carcinogenesis 2012; 33:2155-61. [PMID: 22865593 DOI: 10.1093/carcin/bgs262] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Oesophageal squamous cell carcinoma (OSCC) has a high prevalence in the Black and Mixed Ancestry populations of South Africa. Recently, three genome-wide association studies in Chinese populations identified five new OSCC susceptibility loci, including variants at PLCE1, C20orf54, PDE4D, RUNX1 and UNC5CL, but their contribution to disease risk in other populations is unknown. In this study, we report testing variants from these five loci for association with OSCC in the South African Black (407 cases and 849 controls) and Mixed Ancestry (257 cases and 860 controls) populations. The RUNX1 variant rs2014300, which reduced risk in the Chinese population, was associated with an increased risk of OSCC in the Mixed Ancestry population [odds ratio (OR) = 1.33, 95% confidence interval (CI) = 1.09-1.63, P = 0.0055], and none of the five loci were associated in the Black population. Since PLCE1 variants increased the risk of OSCC in all three Chinese studies, this gene was investigated further by sequencing in 46 Black South Africans. This revealed 48 variants, 10 of which resulted in amino acid substitutions, and much lower linkage disequilibrium across the PLCE1 locus than in the Chinese population. We genotyped five PLCE1 variants in cases and controls, and found association of Arg548Leu (rs17417407) with a reduced risk of OSCC (OR = 0.74, 95% CI = 0.60-0.93, P = 0.008) in the Black population. These findings indicate several differences in the genetic contribution to OSCC between the South African and Chinese populations that may be related to differences in their genetic architecture.
Collapse
Affiliation(s)
- Hannah Bye
- Department of Medical and Molecular Genetics, King's College London, King's Health Partners, Guy's Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Marotta M, Piontkivska H, Tanaka H. Molecular trajectories leading to the alternative fates of duplicate genes. PLoS One 2012; 7:e38958. [PMID: 22720000 PMCID: PMC3375281 DOI: 10.1371/journal.pone.0038958] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 05/14/2012] [Indexed: 11/21/2022] Open
Abstract
Gene duplication generates extra gene copies in which mutations can accumulate without risking the function of pre-existing genes. Such mutations modify duplicates and contribute to evolutionary novelties. However, the vast majority of duplicates appear to be short-lived and experience duplicate silencing within a few million years. Little is known about the molecular mechanisms leading to these alternative fates. Here we delineate differing molecular trajectories of a relatively recent duplication event between humans and chimpanzees by investigating molecular properties of a single duplicate: DNA sequences, gene expression and promoter activities. The inverted duplication of the Glutathione S-transferase Theta 2 (GSTT2) gene had occurred at least 7 million years ago in the common ancestor of African great apes and is preserved in chimpanzees (Pan troglodytes), whereas a deletion polymorphism is prevalent in humans. The alternative fates are associated with expression divergence between these species, and reduced expression in humans is regulated by silencing mutations that have been propagated between duplicates by gene conversion. In contrast, selective constraint preserved duplicate divergence in chimpanzees. The difference in evolutionary processes left a unique DNA footprint in which dying duplicates are significantly more similar to each other (99.4%) than preserved ones. Such molecular trajectories could provide insights for the mechanisms underlying duplicate life and death in extant genomes.
Collapse
Affiliation(s)
- Michael Marotta
- Department of Molecular Genetics, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
| | - Helen Piontkivska
- Department of Biological Sciences, Kent State University, Kent, Ohio, United States of America
| | - Hisashi Tanaka
- Department of Molecular Genetics, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
| |
Collapse
|
33
|
The cumulative effects of polymorphisms in the DNA mismatch repair genes and tobacco smoking in oesophageal cancer risk. PLoS One 2012; 7:e36962. [PMID: 22623965 PMCID: PMC3356375 DOI: 10.1371/journal.pone.0036962] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 04/11/2012] [Indexed: 12/15/2022] Open
Abstract
The DNA mismatch repair (MMR) enzymes repair errors in DNA that occur during normal DNA metabolism or are induced by certain cancer-contributing exposures. We assessed the association between 10 single-nucleotide polymorphisms (SNPs) in 5 MMR genes and oesophageal cancer risk in South Africans. Prior to genotyping, SNPs were selected from the HapMap database, based on their significantly different genotypic distributions between European ancestry populations and four HapMap populations of African origin. In the Mixed Ancestry group, the MSH3 rs26279 G/G versus A/A or A/G genotype was positively associated with cancer (OR = 2.71; 95% CI: 1.34–5.50). Similar associations were observed for PMS1 rs5742938 (GG versus AA or AG: OR = 1.73; 95% CI: 1.07–2.79) and MLH3 rs28756991 (AA or GA versus GG: OR = 2.07; 95% IC: 1.04–4.12). In Black individuals, however, no association between MMR polymorhisms and cancer risk was observed in individual SNP analysis. The interactions between MMR genes were evaluated using the model-based multifactor-dimensionality reduction approach, which showed a significant genetic interaction between SNPs in MSH2, MSH3 and PMS1 genes in Black and Mixed Ancestry subjects, respectively. The data also implies that pathogenesis of common polymorphisms in MMR genes is influenced by exposure to tobacco smoke. In conclusion, our findings suggest that common polymorphisms in MMR genes and/or their combined effects might be involved in the aetiology of oesophageal cancer.
Collapse
|