1
|
Ijaz F, Sameeullah M, Farid A, Malik MS, Batool N, Mirza B, Timko MP, Liu H, Lössl AG, Waheed MT. In silico designing and characterization of outer membrane protein (OmpC) gene from Salmonella enterica and its expression in Nicotiana tabacum for developing a plant-based vaccine against salmonellosis. Microb Pathog 2025; 199:107225. [PMID: 39675439 DOI: 10.1016/j.micpath.2024.107225] [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: 06/29/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Salmonella, a gram-negative bacteria, is the leading cause of foodborne illness globally. Two serovars of Salmonella, S. enteritidis and S. typhimurium are responsible for the majority of human salmonellosis. Prolonged salmonellosis caused by Salmonella species leads to the development of colon cancer, which is 3rd most common cancer in the world. Porins in the outer membrane of Salmonella can be used to elicit immune response. The production of plant-based vaccine against salmonellosis and the subsequent colon cancer using outer membrane proteins can be helpful for the people of developing countries. In this study, OmpC protein from Salmonella enteritidis was subjected to various bioinformatics tools which exhibited OmpC vaccine construct to be sufficiently immunogenic, non-allergenic, non-toxic and non-homologous to human proteins. Docking analysis showed strong interaction of OmpC vaccine model with TLR-4. After in silico analysis, this vaccine construct was expressed in tobacco plants via Agrobacterium-mediated transformation. Gateway® cloning was used to clone OmpC gene. Transformation and integration of transgene within tobacco plants was confirmed through conventional PCR. qRT-PCR was done for expression analysis and copy number calculated was 2. The expressed OmpC protein accumulated up to 0.42 % of total soluble protein. Immunization of mice with total soluble protein (TSP) and purified OmpC protein generated significant level of anti-OmpC antibodies. The vaccine candidate also demonstrated significant protective effect in mice upon challenging with Salmonella typhimurium. To the best of our knowledge, this is the first study reporting the expression of OmpC antigen in plants for potential use as vaccine against salmonellosis.
Collapse
Affiliation(s)
- Fatima Ijaz
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Sameeullah
- Department of Field Crops, Faculty of Agriculture and Natural Sciences, Abant Izzet Baysal University, Golkoy Campus, Bolu, Turkey; Centre for Innovative Food Technologies Development, Application and Research, Bolu Abant Izzet Baysal University, Bolu, 14030, Türkiye
| | | | - Muhammad Suleman Malik
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Neelam Batool
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Bushra Mirza
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Michael P Timko
- Department of Biology, University of Virginia, Virginia, USA
| | - Hai Liu
- Department of Biology, University of Virginia, Virginia, USA
| | | | - Mohammad Tahir Waheed
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
2
|
Zhalbinova MR, Rakhimova SE, Kozhamkulov UA, Akilzhanov KR, Shaimardanov NK, Akilzhanova GA, Lee JH, Pya YV, Bekbossynova MS, Akilzhanova AR. The Impact of Genetic Polymorphism on Complication Development in Heart Failure Patients. J Clin Med 2024; 14:35. [PMID: 39797120 PMCID: PMC11721630 DOI: 10.3390/jcm14010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Despite the high progress that has been made in the field of cardiology, the left ventricular assist device (LVAD) can still cause complications (thrombosis/bleeding) in heart failure (HF) patients after implantation. Complications develop due to the incorrect dose of antithrombotic therapy, due to the influence of the non-physiological shear stress of the device, and also due to inherited genetic polymorphisms. Therefore, the aim of our study is to identify the influence of the genetic polymorphisms on complication development in HF patients with implanted LVADs with prescribed antiplatelet therapy. Methods: Our study investigated 98 HF patients with/without complications who were genotyped for 21 single-nucleotide polymorphisms (SNPs) associated with cardiovascular events, the coagulation system, and the metabolism of warfarin and aspirin drugs. This study performed a more detailed analysis on genetic polymorphism in the UGT1A6 gene and its influence on aspirin dose. Results: SNP rs2070959 in the UGT1A6 gene showed a significant association with the group of HF patients with complications [(OR (95% CI): 4.40 (1.06-18.20), p = 0.044]. The genetic polymorphism of rs2070959 in the UGT1A6 gene showed a significant association in HF patients who received aspirin treatment on the 12th month after LVAD implantation [OR (95% CI): 5.10 (1.31-19.87), p = 0.018]. Moreover, our genotype distribution analysis showed that the GG genotype of rs2070959 in the UGT1A6 gene was significantly higher in the group with aspirin treatment than without treatment after the 12th month of treatment (50.0% vs. 0%, p = 0.008), especially in the group of patients with complications. A higher frequency of the GG genotype with long-lasting aspirin therapy up to the 12th month showed that 100 mg of aspirin was not an effective dose in the group of patients with complications. Conclusions: Our study identified that genotyping for genetic polymorphism rs2070959 in the UGT1A6 gene could predict the recommended dose of aspirin in HF patients, which could help to prevent and predict complication development after LVAD implantation.
Collapse
Affiliation(s)
- Madina R. Zhalbinova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
| | - Saule E. Rakhimova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
- Eurasian Society of Personalize Medicine, Astana 010000, Kazakhstan
| | - Ulan A. Kozhamkulov
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
- Eurasian Society of Personalize Medicine, Astana 010000, Kazakhstan
| | - Kenes R. Akilzhanov
- Department of Medicine, Semey Medical University, Pavlodar Branch, Pavlodar 140000, Kazakhstan; (K.R.A.); (G.A.A.)
| | | | - Gulbanu A. Akilzhanova
- Department of Medicine, Semey Medical University, Pavlodar Branch, Pavlodar 140000, Kazakhstan; (K.R.A.); (G.A.A.)
| | - Joseph H. Lee
- Sergievsky Center, Taub Institute, Columbia University Medical Center, New York, NY 10032, USA;
| | - Yuriy V. Pya
- National Research Cardiac Surgery Center, Astana 010000, Kazakhstan; (Y.V.P.); (M.S.B.)
| | | | - Ainur R. Akilzhanova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
- Eurasian Society of Personalize Medicine, Astana 010000, Kazakhstan
- Department of General Biology and Genomics, L. N. Gumilyov Eurasian National University, Astana 010000, Kazakhstan
| |
Collapse
|
3
|
Tęcza K, Kalinowska-Herok M, Rusinek D, Zajkowicz A, Pfeifer A, Oczko-Wojciechowska M, Pamuła-Piłat J. Are the Common Genetic 3'UTR Variants in ADME Genes Playing a Role in Tolerance of Breast Cancer Chemotherapy? Int J Mol Sci 2024; 25:12283. [PMID: 39596349 PMCID: PMC11594993 DOI: 10.3390/ijms252212283] [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: 10/16/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
We studied the associations between 3'UTR genetic variants in ADME genes, clinical factors, and the risk of breast cancer chemotherapy toxicity. Those variants and factors were tested in relation to seven symptoms belonging to myelotoxicity (anemia, leukopenia, neutropenia), gastrointestinal side effects (vomiting, nausea), nephrotoxicity, and hepatotoxicity, occurring in overall, early, or recurrent settings. The cumulative risk of overall symptoms of anemia was connected with AKR1C3 rs3209896 AG, ERCC1 rs3212986 GT, and >6 cycles of chemotherapy; leukopenia was determined by ABCC1 rs129081 allele G and DPYD rs291593 allele T; neutropenia risk was correlated with accumulation of genetic variants of DPYD rs291583 allele G, ABCB1 rs17064 AT, and positive HER2 status. Risk of nephrotoxicity was determined by homozygote DPYD rs291593, homozygote AKR1C3 rs3209896, postmenopausal age, and negative ER status. Increased risk of hepatotoxicity was connected with NR1/2 rs3732359 allele G, postmenopausal age, and with present metastases. The risk of nausea and vomiting was linked to several genetic factors and premenopausal age. We concluded that chemotherapy tolerance emerges from the simultaneous interaction of many genetic and clinical factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jolanta Pamuła-Piłat
- Department of Clinical and Molecular Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (K.T.); (M.K.-H.); (D.R.); (A.Z.); (A.P.); (M.O.-W.)
| |
Collapse
|
4
|
Lai H, Liu Y, Wu J, Cai J, Jie H, Xu Y, Deng S. Targeting cancer-related inflammation with non-steroidal anti-inflammatory drugs: Perspectives in pharmacogenomics. Front Pharmacol 2022; 13:1078766. [PMID: 36545311 PMCID: PMC9760816 DOI: 10.3389/fphar.2022.1078766] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/25/2022] [Indexed: 12/11/2022] Open
Abstract
Inflammatory processes are essential for innate immunity and contribute to carcinogenesis in various malignancies, such as colorectal cancer, esophageal cancer and lung cancer. Pharmacotherapies targeting inflammation have the potential to reduce the risk of carcinogenesis and improve therapeutic efficacy of existing anti-cancer treatment. Non-steroidal anti-inflammatory drugs (NSAIDs), comprising a variety of structurally different chemicals that can inhibit cyclooxygenase (COX) enzymes and other COX-independent pathways, are originally used to treat inflammatory diseases, but their preventive and therapeutic potential for cancers have also attracted researchers' attention. Pharmacogenomic variability, including distinct genetic characteristics among different patients, can significantly affect pharmacokinetics and effectiveness of NSAIDs, which might determine the preventive or therapeutic success for cancer patients. Hence, a more comprehensive understanding in pharmacogenomic characteristics of NSAIDs and cancer-related inflammation would provide new insights into this appealing strategy. In this review, the up-to-date advances in clinical and experimental researches targeting cancer-related inflammation with NSAIDs are presented, and the potential of pharmacogenomics are discussed as well.
Collapse
Affiliation(s)
- Hongjin Lai
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Wu
- Department of Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Cai
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Jie
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuyang Xu
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Yuyang Xu, ; Senyi Deng,
| | - Senyi Deng
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Yuyang Xu, ; Senyi Deng,
| |
Collapse
|
5
|
Wu P, Guo Y. Susceptibility Loci in SLC15A1, UGT1A3, and CWC27 Genes Associated with Bladder Cancer in the Northeast Chinese Population. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2988159. [PMID: 36124064 PMCID: PMC9482523 DOI: 10.1155/2022/2988159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/24/2022]
Abstract
Bladder cancer (BCa) is an increasingly severe clinical and public health issue. Therefore, we aim to investigate BCa susceptibility loci in the Chinese population. In this study, 487 BCa patients and 563 controls were recruited from the First Affiliated Hospital of China Medical University from July 2015 to September 2020. A total of ten single-nucleotide polymorphisms (SNPs) in solute carrier family 15 member 1 (SLC15A1), CWC27 spliceosome associated cyclophilin (CWC27), or UDP glucuronosyltransferase family 1 member A3 (UGT1A3) genes were genotyped. The associations between the candidate SNPs and BCa were analyzed using genotype and haplotype analysis. The results demonstrated that Rs4646227 of SLC15A1 has a significant association with BCa. The patients with CG (OR =2.513, p < 0.05) and GG (OR =2.859, p < 0.05) genotypes had an increasing risk of BCa compared with the CC genotype. For the CWC27 gene, genotypic frequency analysis revealed that the GT or TT genotype of rs2042329 and the CT or TT genotype of rs1870437 were more frequent in BCa patients than those in the control group, indicating that these genotypes were associated with a higher risk of BCa (all p < 0.05). Haplotypes of SLC15A1, UGT1A3, and CWC27 genes found that the C-C-C haplotype of SLC15A1 was associated with a lower risk of BCa while the C-G-C haplotype was associated with a higher risk. For the UGT1A3 gene, a moderate protective effect was observed with the most frequent T-T-C haplotype, and for the CWC27 gene, most of the haplotypes showed no association with BCa, except the G-G-C-T haplotype (order of SNPs: rs2042329-rs7735338-rs1870437-rs2278351, OR =0.81, p =0.038). In sum, this study indicated that rs2042329 and rs1870437 in the CWC27 gene and rs4646227 in the SLC15A1 gene are independent indicators for BCa risk in Chinese people. Further large-scale studies are required to validate these findings. Also, this study provided the theoretical basis for developing new therapeutic drug targeting of BCa.
Collapse
Affiliation(s)
- Peihong Wu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang 110001, China
| | - Yaoxing Guo
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang 110001, China
| |
Collapse
|
6
|
Zhalbinova MR, Rakhimova SE, Kozhamkulov UA, Akilzhanova GA, Kaussova GK, Akilzhanov KR, Pya YV, Lee JH, Bekbossynova MS, Akilzhanova AR. Association of Genetic Polymorphisms with Complications of Implanted LVAD Devices in Patients with Congestive Heart Failure: A Kazakhstani Study. J Pers Med 2022; 12:jpm12050744. [PMID: 35629166 PMCID: PMC9143784 DOI: 10.3390/jpm12050744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023] Open
Abstract
The left ventricular assist device (LVAD) is one of the alternative treatments for heart failure (HF) patients. However, LVAD support is followed by thrombosis, and bleeding complications which are caused by high non-physiologic shear stress and antithrombotic/anticoagulant therapy. A high risk of complications occurs in the presence of the genotype polymorphisms which are involved in the coagulation system, hemostasis function and in the metabolism of the therapy. The aim of the study was to investigate the influence of single-nucleotide polymorphisms (SNP) in HF patients with LVAD complications. We analyzed 21 SNPs in HF patients (n = 98) with/without complications, and healthy controls (n = 95). SNPs rs9934438; rs9923231 in VKORC1, rs5918 in ITGB3 and rs2070959 in UGT1A6 demonstrated significant association with HF patients’ complications (OR (95% CI): 3.96 (1.42–11.02), p = 0.0057), (OR (95% CI): 3.55 (1.28–9.86), p = 0.011), (OR (95% CI): 5.37 (1.79–16.16), p = 0.0056) and OR (95% CI): 4.40 (1.06–18.20), p = 0.044]. Genotype polymorphisms could help to predict complications at pre- and post-LVAD implantation period, which will reduce mortality rate. Our research showed that patients can receive treatment with warfarin and aspirin with a personalized dosage and LVAD complications can be predicted by reference to their genotype polymorphisms in VKORC1, ITGB3 and UGT1A6 genes.
Collapse
Affiliation(s)
- Madina R. Zhalbinova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
- Department of General Biology and Genomics, L. N. Gumilyov Eurasian National University, Nur-Sultan 010000, Kazakhstan
| | - Saule E. Rakhimova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
| | - Ulan A. Kozhamkulov
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
| | - Gulbanu A. Akilzhanova
- Semey Medical University, Pavlodar Branch, Pavlodar 140000, Kazakhstan; (G.A.A.); (K.R.A.)
| | | | - Kenes R. Akilzhanov
- Semey Medical University, Pavlodar Branch, Pavlodar 140000, Kazakhstan; (G.A.A.); (K.R.A.)
| | - Yuriy V. Pya
- National Research Cardiac Surgery Center, Nur-Sultan 010000, Kazakhstan; (Y.V.P.); (M.S.B.)
| | - Joseph H. Lee
- Sergievsky Center, Taub Institute, Columbia University Irving Medical Centerx, 630 W, New York, NY 10032, USA;
| | | | - Ainur R. Akilzhanova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (M.R.Z.); (S.E.R.); (U.A.K.)
- Department of General Biology and Genomics, L. N. Gumilyov Eurasian National University, Nur-Sultan 010000, Kazakhstan
- Correspondence: ; Tel.: +7-7172-706501
| |
Collapse
|
7
|
Wang Y, Zhao YX, Zhang XW, Jiang YZ, Ma W, Zhang L, Dong W. USF1 Transcriptionally Regulates UGT1A3 and Promotes Lung Adenocarcinoma Progression by Regulating Neurotrophin Signaling Pathway. Front Mol Biosci 2022; 9:758968. [PMID: 35155573 PMCID: PMC8829114 DOI: 10.3389/fmolb.2022.758968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Lung cancer remains the leading cause of oncological death. There is an urgent need to discover new molecular targets and to develop new treatments. Our previous study showed that one of the UDP-glucuronosyltransferases (UGTs) family, UGT1A3, is an important prognostic factor for lung adenocarcinoma (LUAD), inhibiting UGT1A3 could significantly improve the efficacy of anti-tumor drugs. In this study, we aimed to explore the upstream transcriptional factor (USF1) of UGT1A3 and its way of playing a role in LUAD. Methods: The UGT1A3 promoter region was analyzed and dual-luciferase assay was involved to explore whether USF1 could bind to this region, and the possible regulation effects of USF1 to UGT1A3 was indicated by siRNA and recovery experiment. Then, the Cancer Genome Atlas database was used to analyze USF1 clinical features. The expression level of USF1 was detected by immunohistochemical assay and Western blotting. Cellular viability, proliferation, migration and invasion potential were also investigated. Meanwhile, the effect of USF1 in LUAD progression was detected in a mouse model. The downstream signaling pathway was analyzed by bioinformatic analysis and the expression of all related proteins was detected. Results: UGT1A3 was transcriptionally regulated by USF1, which was highly expressed in all investigated samples including patients’ tissues, studied cells lines, and mouse models. The knockdown of USF1 inhibited cells viability, proliferation, migration and invasion, and reduced the tumor volume. Moreover, USF1 promoted the progress of LUAD by regulating the neurotrophin signaling pathway. Conclusion: As an important transcriptional regulator of UGT1A3, USF1 was highly expressed in LUAD and promoted LUAD progression by regulating the neurotrophin signaling pathway. These findings provide a new theoretical data that could serve as a good foundation for the treatment of LUAD.
Collapse
Affiliation(s)
- Yu Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yun-Xia Zhao
- Department of Neurology, Shandong Provinacial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiang-Wei Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuan-Zhu Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Ma
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lin Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Dong
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Wei Dong,
| |
Collapse
|
8
|
Menati Rashno M, Mehraban H, Naji B, Radmehr M. Microbiome in human cancers. Access Microbiol 2021; 3:000247. [PMID: 34888478 PMCID: PMC8650843 DOI: 10.1099/acmi.0.000247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
A microbiome is defined as the aggregate of all microbiota that reside in human digestive system and other tissues. This microbiota includes viruses, bacteria, fungi that live in various human organs and tissues like stomach, guts, oesophagus, mouth cavity, urinary tract, vagina, lungs, and skin. Almost 20 % of malignant cancers worldwide are related to microbial infections including bacteria, parasites, and viruses. The human body is constantly being attacked by microbes during its lifetime and microbial pathogens that have tumorigenic effects in 15-20 % of reported cancer cases. Recent scientific advances and the discovery of the effect of microbes on cancer as a pathogen or as a drug have significantly contributed to our understanding of the complex relationship between microbiome and cancer. The aim of this study is to overview some microbiomes that reside in the human body and their roles in cancer.
Collapse
Affiliation(s)
| | - Hamed Mehraban
- Department of Biology, Payame Noor University (PNU), Tehran, Iran
| | - Behnaz Naji
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mohadeseh Radmehr
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
9
|
AlRuwaisan AU, Al-Anazi MR, Shafeai MI, Rudiny FH, Motaen AM, Bin Dajem SM, Alothaid H, Morsy K, Alkahtani S, Al-Qahtani AA. Associations of Single Nucleotide Polymorphisms in IL-18 Gene with Plasmodium falciparum-Associated Malaria. J Inflamm Res 2021; 14:3587-3619. [PMID: 34345179 PMCID: PMC8323861 DOI: 10.2147/jir.s314638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/23/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Interleukin-18 (IL-18) is a pro-inflammatory cytokine, reported to be involved in the initial immune responses against malaria. Genetic variations in the host are an important factor that influences the etiology of malaria at several disease levels. Polymorphisms within the IL-18 gene are associated with susceptibility and clinical outcome of several diseases. Methods We genotyped single nucleotide polymorphisms (SNPs) in IL-18 of patients infected with Plasmodium falciparum with varying extent of parasitemia and different age groups. Results SNPs rs5744292 (OR = 70.446; 95% CI = 4.318-1149.323; p<0.0001) and rs544354 (OR = 1.498; 95% CI = 1.088-2.063; p=0.013) were found to be significantly associated with parasitemia in P. falciparum-infected patients when compared with healthy control subjects. SNP rs5744292 (OR = 7.597; 95% CI=1.028-56.156; p=0.019) was associated with increased parasite density in infected patients. SNPs rs544354 (OR 0.407; 95% CI=0.204-0.812; p = 0.009) and rs360714 (OR of 0.256; 95% CI=0.119-0.554; p = 0.001) were significantly associated with parasite density in an age-dependent manner, with the risk alleles present more frequently among the younger (1-9 years) patients. Several haplotypes were found to have a significant association with parasitemia. In-vitro expression analysis using luciferase reporter assay showed that SNPs rs1946518 and rs187238 in the IL-18 gene promoter region and rs360728 and rs5744292 in the 3'-untranslated region of the IL-18 gene were associated with enhanced transcriptional activity. Conclusion Our results suggest that polymorphisms within the IL-18 gene are associated with the susceptibility to P. falciparum infection and related parasitemia among groups with different parasite density and across various age groups.
Collapse
Affiliation(s)
- Alaa U AlRuwaisan
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mashael R Al-Anazi
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | | | | | | | - Saad M Bin Dajem
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Saad Alkahtani
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Weismüller TJ, Zhou T, Kalthoff S, Lenzen H, Manns MP, Strassburg CP. Genetic variants of UDP-glucuronosyltransferase 1A genes are associated with disease presentation and outcome in primary sclerosing cholangitis. Liver Int 2020; 40:1645-1654. [PMID: 32378294 DOI: 10.1111/liv.14487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/09/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Primary sclerosing cholangitis (PSC) is a progressive cholestatic liver disease without a curative medical therapy. The human UDP-glucuronosyltransferases 1A play a major role in the detoxification and elimination of bilirubin, bile acids and xenobiotics. Whether genetic UGT1A variants determine course and outcome of PSC has not yet been described. METHODS A large cohort of German PSC patients with a long-term-follow-up was genotyped for UGT1A variants including UGT1A1*28, UGT1A3-66 T>C and UGT1A7 p.N129K/p.R131K using TaqMan 5'-nuclease assays. Results were correlated with clinical characteristics and transplant-free survival. RESULTS About 331 patients with PSC were included in the study (69.9% male, mean age at diagnosis 32.6 years). Median transplant-free survival was 14.9 years. Patients with wild-type alleles of all three UGT1A genes had a longer transplant-free survival (17.2 vs. 14.4 years, P = .048) than patients carrying a homozygous or heterozygous SNP variant in at least one of the UGT1A1, UGT1A3 or UGT1A7 genes. Additionally, we found that patients carrying wild-type alleles of all three UGT1A genes had lower serum bilirubin (25 vs. 38 µmol/L, P = .02) and serum cholesterol (195 vs. 223 mg/dL), P = .035) at first presentation. Furthermore, inflammatory bowel disease was found to be associated with wild-type UGT1A alleles (82.2% vs. 68.4%, P = .046). CONCLUSIONS This large cohort shows an association with single nucleotide polymorphisms of the UGT1A1, UGT1A3 and UGT1A7 genes and outcome in PSC. Thus, UGT1A variants may represent a tool for the prognostic stratification of PSC patients and establish a link between disease progression and the regulation of detoxification by glucuronidation in PSC.
Collapse
Affiliation(s)
| | - Taotao Zhou
- Department of Internal Medicine 1, University of Bonn, Bonn, Germany
| | - Sandra Kalthoff
- Department of Internal Medicine 1, University of Bonn, Bonn, Germany
| | - Henrike Lenzen
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
11
|
Beyerle J, Holowatyj AN, Haffa M, Frei E, Gigic B, Schrotz-King P, Boehm J, Habermann N, Stiborova M, Scherer D, Kölsch T, Skender S, Becker N, Herpel E, Schneider M, Ulrich A, Schirmacher P, Chang-Claude J, Brenner H, Hoffmeister M, Haug U, Owen RW, Ulrich CM. Expression Patterns of Xenobiotic-Metabolizing Enzymes in Tumor and Adjacent Normal Mucosa Tissues among Patients with Colorectal Cancer: The ColoCare Study. Cancer Epidemiol Biomarkers Prev 2019; 29:460-469. [PMID: 31740522 DOI: 10.1158/1055-9965.epi-19-0449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/15/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Xenobiotic-metabolizing enzymes (XME) play a critical role in the activation and detoxification of several carcinogens. However, the role of XMEs in colorectal carcinogenesis is unclear. METHODS We investigated the expression of XMEs in human colorectal tissues among patients with stage I-IV colorectal cancer (n = 71) from the ColoCare Study. Transcriptomic profiling using paired colorectal tumor and adjacent normal mucosa tissues of XMEs (GSTM1, GSTA1, UGT1A8, UGT1A10, CYP3A4, CYP2C9, GSTP1, and CYP2W1) by RNA microarray was compared using Wilcoxon rank-sum tests. We assessed associations between clinicopathologic, dietary, and lifestyle factors and XME expression with linear regression models. RESULTS GSTM1, GSTA1, UGT1A8, UGT1A10, and CYP3A4 were all statistically significantly downregulated in colorectal tumor relative to normal mucosa tissues (all P ≤ 0.03). Women had significantly higher expression of GSTM1 in normal tissues compared with men (β = 0.37, P = 0.02). By tumor site, CYP2C9 expression was lower in normal mucosa among patients with rectal cancer versus colon cancer cases (β = -0.21, P = 0.0005). Smokers demonstrated higher CYP2C9 expression levels in normal mucosa (β = 0.17, P = 0.02) when compared with nonsmokers. Individuals who used NSAIDs had higher GSTP1 tumor expression compared with non-NSAID users (β = 0.17, P = 0.03). Higher consumption of cooked vegetables (>1×/week) was associated with higher CYP3A4 expression in colorectal tumor tissues (β = 0.14, P = 0.007). CONCLUSIONS XMEs have lower expression in colorectal tumor relative to normal mucosa tissues and may modify colorectal carcinogenesis via associations with clinicopathologic, lifestyle, and dietary factors. IMPACT Better understanding into the role of drug-metabolizing enzymes in colorectal cancer may reveal biological differences that contribute to cancer development, as well as treatment response, leading to clinical implications in colorectal cancer prevention and management.
Collapse
Affiliation(s)
- Jolantha Beyerle
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Andreana N Holowatyj
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah.,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Mariam Haffa
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Eva Frei
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Petra Schrotz-King
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Juergen Boehm
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah.,Huntsman Cancer Institute, Salt Lake City, Utah
| | - Nina Habermann
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Dominique Scherer
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Torsten Kölsch
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Stephanie Skender
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Nikolaus Becker
- NCT Cancer Registry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | | | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Haug
- Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany.,Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| | - Robert W Owen
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Cornelia M Ulrich
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah. .,Huntsman Cancer Institute, Salt Lake City, Utah.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
12
|
Wang Y, Liu S, Dong W, Qu X, Huang C, Yan T, Du J. Combination of hesperetin and platinum enhances anticancer effect on lung adenocarcinoma. Biomed Pharmacother 2019; 113:108779. [DOI: 10.1016/j.biopha.2019.108779] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/03/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022] Open
|
13
|
Meech R, Hu DG, McKinnon RA, Mubarokah SN, Haines AZ, Nair PC, Rowland A, Mackenzie PI. The UDP-Glycosyltransferase (UGT) Superfamily: New Members, New Functions, and Novel Paradigms. Physiol Rev 2019; 99:1153-1222. [DOI: 10.1152/physrev.00058.2017] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UDP-glycosyltransferases (UGTs) catalyze the covalent addition of sugars to a broad range of lipophilic molecules. This biotransformation plays a critical role in elimination of a broad range of exogenous chemicals and by-products of endogenous metabolism, and also controls the levels and distribution of many endogenous signaling molecules. In mammals, the superfamily comprises four families: UGT1, UGT2, UGT3, and UGT8. UGT1 and UGT2 enzymes have important roles in pharmacology and toxicology including contributing to interindividual differences in drug disposition as well as to cancer risk. These UGTs are highly expressed in organs of detoxification (e.g., liver, kidney, intestine) and can be induced by pathways that sense demand for detoxification and for modulation of endobiotic signaling molecules. The functions of the UGT3 and UGT8 family enzymes have only been characterized relatively recently; these enzymes show different UDP-sugar preferences to that of UGT1 and UGT2 enzymes, and to date, their contributions to drug metabolism appear to be relatively minor. This review summarizes and provides critical analysis of the current state of research into all four families of UGT enzymes. Key areas discussed include the roles of UGTs in drug metabolism, cancer risk, and regulation of signaling, as well as the transcriptional and posttranscriptional control of UGT expression and function. The latter part of this review provides an in-depth analysis of the known and predicted functions of UGT3 and UGT8 enzymes, focused on their likely roles in modulation of levels of endogenous signaling pathways.
Collapse
Affiliation(s)
- Robyn Meech
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Ross A. McKinnon
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Siti Nurul Mubarokah
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Alex Z. Haines
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Pramod C. Nair
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Peter I. Mackenzie
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
14
|
Anon B, Perray C, Regnault D, Caulet M, Orain I, Godart B, Pages JC, Tallet A, Ouaissi M, Guyetant S, Barin-le Guellec C, Lecomte T. A study of the association between UGT1A1*28 variant allele of UGT1A1 gene and colonic phenotype of sporadic colorectal cancer. Dig Liver Dis 2019; 51:579-583. [PMID: 30583998 DOI: 10.1016/j.dld.2018.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/25/2018] [Accepted: 11/27/2018] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The transcriptional activity of the UGT1A1 gene is modulated by a variable number of repetitions of the dinucleotide (TA) within its promoter region. By comparison to the most common allele (TA)6 (UGT1A1*1), decreased activity is observed with increasing TA repetitions. The aim of this study was to determine whether the presence of the variant allele UGT1A1*28, harbouring seven TA repetitions, (TA)7, in the homozygous state, is associated with precancerous colonic lesions and/or with specific colorectal cancer characteristics. MATERIAL AND METHODS All patients treated for colorectal cancer in a tertiary care centre, between January 2009 and December 2013, who had routine UGT1A1 genotyping for irinotecan dose-adjustment were included. Data were retrospectively collected. RESULTS 292 patients were enrolled, including 23 UGT1A1*28/*28 homozygous (7.9%), 137 wild type homozygous (46.9%) and 132 heterozygous (45.2%). There were no significant differences in phenotypic colonic characteristics between homozygous and heterozygous patients carrying the UGT1A1*28 allele as compared to *1/*1 homozygous. Patients treated with aspirin were significantly more common in the UGT1A1*28/*28 homozygous group than in the other groups (7/23 (30.4%) compared to 22/269 (8.2%), p = 0.001). CONCLUSION Dinucleotide polymorphism in the promoter region of the UGT1A1 gene is not associated with a specific colonic phenotype in patients with sporadic colorectal cancer.
Collapse
Affiliation(s)
- Benjamin Anon
- Department of Hepatogastroenterology and Digestive Oncology, Tours, France.
| | - Clémence Perray
- Department of Hepatogastroenterology and Digestive Oncology, Tours, France
| | - David Regnault
- Department of Hepatogastroenterology and Digestive Oncology, Tours, France
| | - Morgane Caulet
- Department of Hepatogastroenterology and Digestive Oncology, Tours, France
| | | | - Bruno Godart
- Department of Hepatogastroenterology and Digestive Oncology, Tours, France
| | | | - Anne Tallet
- Platform of Somatic Tumour Molecular Genetics, Tours, France
| | | | | | - Chantal Barin-le Guellec
- University of Tours, Tours, France; Department of Molecular Biology, Tours, France; INSERM, UMR 1248, Université de Limoges, Limoges, France
| | - Thierry Lecomte
- Department of Hepatogastroenterology and Digestive Oncology, Tours, France; EA 7501 (GICC), Université de Tours, France
| |
Collapse
|
15
|
Charkoftaki G, Thompson DC, Golla JP, Garcia-Milian R, Lam TT, Engel J, Vasiliou V. Integrated multi-omics approach reveals a role of ALDH1A1 in lipid metabolism in human colon cancer cells. Chem Biol Interact 2019; 304:88-96. [PMID: 30851239 DOI: 10.1016/j.cbi.2019.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/16/2019] [Accepted: 02/28/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Georgia Charkoftaki
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520, USA
| | - David C Thompson
- Department of Clinical Pharmacy, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Jaya Prakash Golla
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520, USA
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, 06250, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, 06510, USA; Yale MS & Proteomics Resource, WM Keck Biotechnology Resource Laboratory, New Haven, CT, 06510, USA
| | - Jasper Engel
- Biometris, Wageningen University & Research, Wagenigen, the Netherlands
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
16
|
Sheth H, Northwood E, Ulrich CM, Scherer D, Elliott F, Barrett JH, Forman D, Wolf CR, Smith G, Jackson MS, Santibanez-Koref M, Haile R, Casey G, Jenkins M, Win AK, Hopper JL, Marchand LL, Lindor NM, Thibodeau SN, Potter JD, Burn J, Bishop DT. Interaction between polymorphisms in aspirin metabolic pathways, regular aspirin use and colorectal cancer risk: A case-control study in unselected white European populations. PLoS One 2018; 13:e0192223. [PMID: 29425227 PMCID: PMC5806861 DOI: 10.1371/journal.pone.0192223] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/19/2018] [Indexed: 12/21/2022] Open
Abstract
Regular aspirin use is associated with reduced risk of colorectal cancer (CRC). Variation in aspirin's chemoprevention efficacy has been attributed to the presence of single nucleotide polymorphisms (SNPs). We conducted a meta-analysis using two large population-based case-control datasets, the UK-Leeds Colorectal Cancer Study Group and the NIH-Colon Cancer Family Registry, having a combined total of 3325 cases and 2262 controls. The aim was to assess 42 candidate SNPs in 15 genes whose association with colorectal cancer risk was putatively modified by aspirin use, in the literature. Log odds ratios (ORs) and standard errors were estimated for each dataset separately using logistic regression adjusting for age, sex and study site, and dataset-specific results were combined using random effects meta-analysis. Meta-analysis showed association between SNPs rs6983267, rs11694911 and rs2302615 with CRC risk reduction (All P<0.05). Association for SNP rs6983267 in the CCAT2 gene only was noteworthy after multiple test correction (P = 0.001). Site-specific analysis showed association between SNPs rs1799853 and rs2302615 with reduced colon cancer risk only (P = 0.01 and P = 0.004, respectively), however neither reached significance threshold following multiple test correction. Meta-analysis of SNPs rs2070959 and rs1105879 in UGT1A6 gene showed interaction between aspirin use and CRC risk (Pinteraction = 0.01 and 0.02, respectively); stratification by aspirin use showed an association for decreased CRC risk for aspirin users having a wild-type genotype (rs2070959 OR = 0.77, 95% CI = 0.68-0.86; rs1105879 OR = 0.77 95% CI = 0.69-0.86) compared to variant allele cariers. The direction of the interaction however is in contrast to that published in studies on colorectal adenomas. Both SNPs showed potential site-specific interaction with aspirin use and colon cancer risk only (Pinteraction = 0.006 and 0.008, respectively), with the direction of association similar to that observed for CRC. Additionally, they showed interaction between any non-steroidal anti-inflammatory drugs (including aspirin) use and CRC risk (Pinteraction = 0.01 for both). All gene x environment (GxE) interactions however were not significant after multiple test correction. Candidate gene investigation indicated no evidence of GxE interaction between genetic variants in genes involved in aspirin pathways, regular aspirin use and colorectal cancer risk.
Collapse
Affiliation(s)
- Harsh Sheth
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Emma Northwood
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Cornelia M. Ulrich
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Dominique Scherer
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Faye Elliott
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Jennifer H. Barrett
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - David Forman
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - C. Roland Wolf
- School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Gillian Smith
- School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Michael S. Jackson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mauro Santibanez-Koref
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Robert Haile
- Stanford Cancer Institute, Stanford, California, United States of America
| | - Graham Casey
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Mark Jenkins
- Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Australia
| | - Aung Ko Win
- Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Australia
| | - John L. Hopper
- Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Australia
| | | | | | | | - John D. Potter
- Centre for Public Health Research, Massey University, Wellington, New Zealand
| | - John Burn
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - D. Timothy Bishop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
17
|
He BX, Qiao B, Lam AKY, Zhao XL, Zhang WZ, Liu H. Association between UDP-glucuronosyltransferase 2B7 tagSNPs and breast cancer risk in Chinese females. Clin Exp Pharmacol Physiol 2018; 45:437-443. [PMID: 29272031 DOI: 10.1111/1440-1681.12908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 11/18/2017] [Accepted: 12/06/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Bao-Xia He
- Department of Pharmacy; Affiliated Cancer Hospital of Zhengzhou University; Zhengzhou China
| | - Bin Qiao
- Department of Stomatology; The First Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology; School of Medicine; Menzies Health Institute Queensland; Griffith University; Gold Coast QLD Australia
| | - Xiu-Li Zhao
- Department of Pharmacy; Affiliated Cancer Hospital of Zhengzhou University; Zhengzhou China
| | - Wen-Zhou Zhang
- Department of Pharmacy; Affiliated Cancer Hospital of Zhengzhou University; Zhengzhou China
| | - Hui Liu
- Department of Breast Surgery; Affiliated Cancer Hospital of Zhengzhou University; Zhengzhou China
| |
Collapse
|
18
|
Mughini-Gras L, Schaapveld M, Kramers J, Mooij S, Neefjes-Borst EA, van Pelt W, Neefjes J. Increased colon cancer risk after severe Salmonella infection. PLoS One 2018; 13:e0189721. [PMID: 29342165 PMCID: PMC5771566 DOI: 10.1371/journal.pone.0189721] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/30/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Colon cancer constitutes one of the most frequent malignancies. Previous studies showed that Salmonella manipulates host cell signaling pathways and that Salmonella Typhimurium infection facilitates colon cancer development in genetically predisposed mice. This epidemiological study examined whether severe Salmonella infection, usually acquired from contaminated food, is associated with increased colon cancer risk in humans. METHODS AND FINDINGS We performed a nationwide registry-based study to assess colon cancer risk after diagnosed Salmonella infection. National infectious disease surveillance records (1999-2015) for Dutch residents aged ≥20 years when diagnosed with salmonellosis (n = 14,264) were linked to the Netherlands Cancer Registry. Salmonella-infected patients were laboratory-confirmed under medical consultation after 1-2 weeks of illness. These datasets also contained information on Salmonella serovar and type of infection. Colon cancer risk (overall and per colon subsite) among patients with a diagnosed Salmonella infection was compared with expected colon cancer risk in the general population. Data from the nationwide registry of histo- and cytopathology (PALGA) and Statistics Netherlands (CBS) allowed assessing potential effects of age, gender, latency, socioeconomic status, genetic predisposition, inflammatory bowel disease (IBD), and tumor features. We found that compared to the general population, colon cancer risk was significantly increased (standardized incidence ratio [SIR] 1.54; 95%CI 1.09-2.10) among patients with Salmonella infection diagnosed <60 years of age. Such increased risk concerned specifically the ascending/transverse colon (SIR 2.12; 95%CI 1.38-3.09) after S. Enteritidis infection (SIR 2.97; 95%CI 1.73-4.76). Salmonellosis occurred more frequently among colon cancer patients with pre-infectious IBD, a known risk factor for colon cancer. Colon tumors of patients with a history of Salmonella infection were mostly of low grade. CONCLUSIONS Patients diagnosed with severe salmonellosis have an increased risk of developing cancer in the ascending/transverse parts of the colon. This risk concerns particularly S. Enteritidis infection, suggesting a contribution of this major foodborne pathogen to colon cancer development.
Collapse
Affiliation(s)
- Lapo Mughini-Gras
- National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, Bilthoven, the Netherlands
- Department of Infectious Diseases and Immunology, Utrecht University, Yalelaan 1, Utrecht, the Netherlands
| | - Michael Schaapveld
- Division of Epidemiology, the Netherlands Cancer Institute (NKI), Plesmanlaan 121, Amsterdam, the Netherlands
| | - Jolanda Kramers
- National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, Bilthoven, the Netherlands
| | - Sofie Mooij
- National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, Bilthoven, the Netherlands
| | - E. Andra Neefjes-Borst
- Department of Pathology, Free University Medical Center (VUmc), Boelelaan 1117, Amsterdam, the Netherlands
| | - Wilfrid van Pelt
- National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, Bilthoven, the Netherlands
| | - Jacques Neefjes
- Department of Chemical Immunology, Leiden University Medical Center (LUMC), Einthovenweg 20, Leiden, the Netherlands
| |
Collapse
|
19
|
Yang N, Sun R, Liao X, Aa J, Wang G. UDP-glucuronosyltransferases (UGTs) and their related metabolic cross-talk with internal homeostasis: A systematic review of UGT isoforms for precision medicine. Pharmacol Res 2017; 121:169-183. [PMID: 28479371 DOI: 10.1016/j.phrs.2017.05.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/03/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022]
Abstract
UDP-glucuronosyltransferases (UGTs) are the primary phase II enzymes catalyzing the conjugation of glucuronic acid to the xenobiotics with polar groups for facilitating their clearance. The UGTs belong to a superfamily that consists of diverse isoforms possessing distinct but overlapping metabolic activity. The abnormality or deficiency of UGTs in vivo is highly associated with some diseases, efficacy and toxicity of drugs, and precisely therapeutic personality. Despite the great effects and fruitful results achieved, to date, the expression and functions of individual UGTs have not been well clarified, the inconsistency of UGTs is often observed in human and experimental animals, and the complex regulation factors affecting UGTs have not been systematically summarized. This article gives an overview of updated reports on UGTs involving the various regulatory factors in terms of the genetic, environmental, pathological, and physiological effects on the functioning of individual UGTs, in turn, the dysfunction of UGTs induced disease risk and endo- or xenobiotic metabolism-related toxicity. The complex cross-talk effect of UGTs with internal homeostasis is systematically summarized and discussed in detail, which would be of great importance for personalized precision medicine.
Collapse
Affiliation(s)
- Na Yang
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Runbin Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoying Liao
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Jiye Aa
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
20
|
Cassidy A, Minihane AM. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am J Clin Nutr 2017; 105:10-22. [PMID: 27881391 PMCID: PMC5183723 DOI: 10.3945/ajcn.116.136051] [Citation(s) in RCA: 337] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 10/13/2016] [Indexed: 01/10/2023] Open
Abstract
At a population level, there is growing evidence of the beneficial effects of dietary flavonoids on health. However, there is extensive heterogeneity in the response to increased intake, which is likely mediated via wide interindividual variability in flavonoid absorption and metabolism. Flavonoids are extensively metabolized by phase I and phase II metabolism (which occur predominantly in the gastrointestinal tract and liver) and colonic microbial metabolism. A number of factors, including age, sex, and genotype, may affect these metabolic processes. In addition, food composition and flavonoid source are likely to affect bioavailability, and emerging data suggest a critical role for the microbiome. This review will focus on the current knowledge for the main subclasses of flavonoids, including anthocyanins, flavonols, flavan-3-ols, and flavanones, for which there is growing evidence from prospective studies of beneficial effects on health. The identification of key factors that govern metabolism and an understanding of how the differential capacity to metabolize these bioactive compounds affect health outcomes will help establish how to optimize intakes of flavonoids for health benefits and in specific subgroups. We identify research areas that need to be addressed to further understand important determinants of flavonoid bioavailability and metabolism and to advance the knowledge base that is required to move toward the development of dietary guidelines and recommendations for flavonoids and flavonoid-rich foods.
Collapse
Affiliation(s)
- Aedín Cassidy
- Department of Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | | |
Collapse
|
21
|
Abstract
Aspirin (acetylsalicylic acid) has become one of the most commonly used drugs, given its role as an analgesic, antipyretic and agent for cardiovascular prophylaxis. Several decades of research have provided considerable evidence demonstrating its potential for the prevention of cancer, particularly colorectal cancer. Broader clinical recommendations for aspirin-based chemoprevention strategies have recently been established; however, given the known hazards of long-term aspirin use, larger-scale adoption of an aspirin chemoprevention strategy is likely to require improved identification of individuals for whom the protective benefits outweigh the harms. Such a precision medicine approach may emerge through further clarification of aspirin's mechanism of action.
Collapse
Affiliation(s)
- David A Drew
- Massachusetts General Hospital and Harvard Medical School, Clinical and Translational Epidemiology Unit, 55 Fruit Street, Bartlett Ext. 9, Boston, Massachusetts 02114, USA
| | - Yin Cao
- Massachusetts General Hospital and Harvard Medical School, Clinical and Translational Epidemiology Unit, and Harvard T.H. Chan School of Public Health, Department of Nutrition, 55 Fruit Street, Bartlett Ext. 9, Boston, Massachusetts 02114, USA
| | - Andrew T Chan
- Massachusetts General Hospital and Harvard Medical School, Clinical and Translational Epidemiology Unit, Division of Gastroenterology, GRJ-825C, Boston, Massachusetts 02114, USA
| |
Collapse
|
22
|
Hu DG, Mackenzie PI, McKinnon RA, Meech R. Genetic polymorphisms of human UDP-glucuronosyltransferase (UGT) genes and cancer risk. Drug Metab Rev 2016; 48:47-69. [DOI: 10.3109/03602532.2015.1131292] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
|
24
|
Pharmacogenomics of human uridine diphospho-glucuronosyltransferases and clinical implications. Clin Pharmacol Ther 2014; 96:324-39. [PMID: 24922307 DOI: 10.1038/clpt.2014.126] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/07/2014] [Indexed: 12/12/2022]
Abstract
Glucuronidation by uridine diphospho-glucuronosyltransferase enzymes (UGTs) is a major phase II biotransformation pathway and, complementary to phase I metabolism and membrane transport, one of the most important cellular defense mechanisms responsible for the inactivation of therapeutic drugs, other xenobiotics, and endogenous molecules. Interindividual variability in UGT pathways is significant and may have profound pharmacological and toxicological implications. Several genetic and genomic processes underlie this variability and are discussed in relation to drug metabolism and diseases such as cancer.
Collapse
|