1
|
Guo S, Zhu W, Bian Y, Li Z, Zheng H, Li W, Yang Y, Ji X, Zhang B. Developing diagnostic biomarkers for Alzheimer's disease based on histone lactylation-related gene. Heliyon 2024; 10:e37807. [PMID: 39315143 PMCID: PMC11417585 DOI: 10.1016/j.heliyon.2024.e37807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Background Research underscores the significant influence of histone lactylation pathways in the progression of Alzheimer's disease (AD), though the molecular mechanisms associated with histone lactylation-related genes (HLRGs) in AD are still insufficiently investigated. Methods This study employed datasets GSE85426 and GSE97760 to identify candidate genes by intersecting weighted gene co-expression network analysis (WGCNA) module genes with AD-control differentially expressed genes (DEGs). Subsequently, machine learning refined key genes, validated by receiver operating characteristic (ROC) curve performance. Gene-set enrichment analysis (GSEA) explored the molecular mechanisms of these diagnostic markers. Concurrently, the association between the diagnostic genes and both differential immune cells and immune responses was examined. Furthermore, a ceRNA and gene-drug network was developed. Finally, the expression of the selected genes was validated using brain tissues from AD model mice. Results This study identified five genes (ARID5B, NSMCE4A, SESN1, THADA, and XPA) with significant diagnostic utility, primarily enriched in olfactory transduction and N-glycan biosynthesis pathways. Correlation analysis demonstrated a strong positive association between all diagnostic genes and naive B cells. The ceRNA regulatory network comprised 7 miRNAs, 2 mRNAs, and 25 lncRNAs. Additionally, 33 drugs targeting the diagnostic genes were predicted. Following expression validation through training and validation sets, three genes (ARID5B, SESN1, XPA) were ultimately confirmed as biomarkers for this study. RT-qPCR and Western blot analyses revealed upregulated expression of ARID5B, SESN1, and XPA in the cerebral tissue of AD model mice. Conclusion Three histone lactylation-linked genes (ARID5B, SESN1, XPA) were identified as potential AD biomarkers, indicating a strong association with disease progression.
Collapse
Affiliation(s)
- Shaobo Guo
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Wenhui Zhu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yuting Bian
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Zhikai Li
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Heng Zheng
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
- Zhenjiang Hospital of Chinese Traditional And Western Medicine, Zhenjiang, China
| | - Wenlong Li
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
- Liyang Hospital of Chinese Medicine, Liyang, China
| | - Yi Yang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Xuzheng Ji
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Biao Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Martino D, Kresoje N, Amenyogbe N, Ben-Othman R, Cai B, Lo M, Idoko O, Odumade OA, Falsafi R, Blimkie TM, An A, Shannon CP, Montante S, Dhillon BK, Diray-Arce J, Ozonoff A, Smolen KK, Brinkman RR, McEnaney K, Angelidou A, Richmond P, Tebbutt SJ, Kampmann B, Levy O, Hancock REW, Lee AHY, Kollmann TR. DNA Methylation signatures underpinning blood neutrophil to lymphocyte ratio during first week of human life. Nat Commun 2024; 15:8167. [PMID: 39289350 PMCID: PMC11408723 DOI: 10.1038/s41467-024-52283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Understanding of newborn immune ontogeny in the first week of life will enable age-appropriate strategies for safeguarding vulnerable newborns against infectious diseases. Here we conducted an observational study exploring the immunological profile of infants longitudinally throughout their first week of life. Our Expanded Program on Immunization - Human Immunology Project Consortium (EPIC-HIPC) studies the epigenetic regulation of systemic immunity using small volumes of peripheral blood samples collected from West African neonates on days of life (DOL) 0, 1, 3, and 7. Genome-wide DNA methylation and single nucleotide polymorphism markers are examined alongside matched transcriptomic and flow cytometric data. Integrative analysis reveals that a core network of transcription factors mediates dynamic shifts in neutrophil-to-lymphocyte ratios (NLR), which are underpinned by cell-type specific methylation patterns in the two cell types. Genetic variants are associated with lower NLRs at birth, and healthy newborns with lower NLRs at birth are more likely to subsequently develop sepsis. These findings provide valuable insights into the early-life determinants of immune system development.
Collapse
Affiliation(s)
- David Martino
- The Kids Research Institute Australia, Perth, WA, Australia.
- University of Western Australia, Crawley, WA, Australia.
| | - Nina Kresoje
- The Kids Research Institute Australia, Perth, WA, Australia
| | - Nelly Amenyogbe
- The Kids Research Institute Australia, Perth, WA, Australia
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Bing Cai
- BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Mandy Lo
- BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Olubukola Idoko
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul, Gambia
| | - Oludare A Odumade
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Reza Falsafi
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Travis M Blimkie
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Andy An
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Casey P Shannon
- PROOF Centre of Excellence, Providence Research, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | | | - Bhavjinder K Dhillon
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kinga K Smolen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Kerry McEnaney
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Asimenia Angelidou
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neonatology, Beth Israel Deaconess Medical Centre, Boston, MA, USA
| | - Peter Richmond
- The Kids Research Institute Australia, Perth, WA, Australia
- University of Western Australia, Crawley, WA, Australia
| | - Scott J Tebbutt
- PROOF Centre of Excellence, Providence Research, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Beate Kampmann
- Vaccines & Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul, Gambia
- Centre for Global Health and Institute for International Health, Charite Universitatsmedizin, Berlin, Germany
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert E W Hancock
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Amy H Y Lee
- Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Tobias R Kollmann
- The Kids Research Institute Australia, Perth, WA, Australia
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
3
|
Tagawa Y, Saito T, Iwai H, Sato M, Noda S, Yamamoto A, Ota M, Endo K, Koga H, Takahara Y, Sugimoto K, Sekiya I, Fujio K, Kawakami E, Mizoguchi F, Yasuda S. ARID5B is a negative modulator of IL-6 production in rheumatoid arthritis synovial fibroblasts. Immunol Med 2024; 47:176-185. [PMID: 38747454 DOI: 10.1080/25785826.2024.2346956] [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: 12/06/2023] [Accepted: 03/06/2024] [Indexed: 08/23/2024] Open
Abstract
Recent single-cell RNA-sequencing analysis of rheumatoid arthritis (RA) synovial tissues revealed the heterogeneity of RA synovial fibroblasts (SFs) with distinct functions such as high IL-6 production. The molecular mechanisms responsible for high IL-6 production will become a promising drug target of RASFs to treat RA. In this study, we performed siRNA screening of 65 transcription factors (TFs) differentially expressed among RASF subsets to identify TFs involved in IL-6 production. The siRNA screening identified 7 TFs including ARID5B, a RA risk gene, that affected IL-6 production. Both long and short isoforms of ARID5B were expressed and negatively regulated by TNF-α in RASFs. The siRNA knockdown and lentiviral overexpression of long and short isoforms of ARID5B revealed that the long isoform suppressed IL-6 production stimulated with TNF-α. eQTL analysis using 58 SFs demonstrated that RA risk allele, rs10821944, in intron 4 of the ARID5B gene had a trend of eQTL effects to the expression of long isoform of ARID5B in SFs treated with TNF-α. ARID5B was found to be a negative modulator of IL-6 production in RASFs. The RA risk allele of ARID5B intron may cause high IL-6 production, suggesting that ARID5B will become a promising drug target to treat RA.
Collapse
Affiliation(s)
- Yasuhiro Tagawa
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuya Saito
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hideyuki Iwai
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Motohiko Sato
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Seiji Noda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akio Yamamoto
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kentaro Endo
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | | | - Kazutaka Sugimoto
- Department of Orthopedics, Sonodakai Joint Replacement Center Hospital
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eiryo Kawakami
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Advanced Data Science Project (ADSP), RIKEN Information R&D and Strategy Headquarters, Yokohama, Kanagawa, Japan
- NEXT-Ganken Program, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan
| | - Fumitaka Mizoguchi
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
4
|
Rahmayanti SU, Amalia R, Rusdiana T. Systematic review: genetic polymorphisms in the pharmacokinetics of high-dose methotrexate in pediatric acute lymphoblastic leukemia patients. Cancer Chemother Pharmacol 2024; 94:141-155. [PMID: 39002021 DOI: 10.1007/s00280-024-04694-0] [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: 03/30/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024]
Abstract
Variations in pharmacokinetic responses to high-dose methotrexate are essential for the prognosis and management of toxicity in the treatment of pediatric acute lymphoblastic leukemia (ALL) patients. This systematic review aimed to identify and evaluate genetic polymorphisms that are significantly associated with the pharmacokinetic parameters of methotrexate during the consolidation phase of pediatric ALL treatment. Using the Preferred Reporting Items for Systematic Reviews (PRISMA) guidelines, we systematically reviewed the literature from 2013 to 2023. The databases used were PubMed and Scopus. The outcomes of interest are the study design, patient characteristics, sample size, chemotherapy protocol utilized, pharmacokinetic parameters identified, and genetic polymorphisms implicated. We included 31 articles in the qualitative synthesis and found that the SLCO1B1, ABCB1, ABCC2, and MTHFR genes appear to play significant roles in MTX metabolism and clearance. Among these, variations in SLCO1B1 have the most significant and consistent impact on methotrexate clearance. These implicated variants may contribute to the precision and tailoring of HD-MTX treatment in pediatric ALL patients.
Collapse
Affiliation(s)
- Siti Utami Rahmayanti
- Master Program in Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, West Java, 45363, Indonesia
| | - Riezki Amalia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, 45363, Indonesia
| | - Taofik Rusdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Padjadjaran University, Sumedang, 45363, Indonesia.
| |
Collapse
|
5
|
Algarni A. Genetic Insights Into Leukemia Susceptibility in the Arab Population: A Scoping Review. Cureus 2024; 16:e67421. [PMID: 39310620 PMCID: PMC11415027 DOI: 10.7759/cureus.67421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
As per the Global Cancer Observatory, the WHO Eastern Mediterranean region (which includes the Arabic countries) ranks highest for age-standardized mortality rate at 4 per 100,000, thus indicating a probable role of genetic associations. Identifying the genes associated with leukemia in the Arab population is crucial for effective preventive and treatment strategies. This scoping review aimed to determine the nature and extent of research available on the genes associated with the major types of leukemia among the Arab population. As per the scoping review guidelines, a comprehensive search was conducted in PUBMED and Google Scholar for articles published before 01/10/2023 and focused on leukemia-related genes among the Arab population. In total 119 studies, focusing on genes associated with leukemia met the inclusion criteria. On reviewing these studies, 27 genes were found to be associated with ALL, 33 genes with AML, seven genes with CLL, and 14 genes with CML. The majority of these genes were associated with an increased risk for the disease. Notably, the 119 studies covered only nine out of the 22 Arab countries, with 56 studies carried out in Egypt, exhibiting an imbalance in the regional distribution of the research landscape. Thus, indicating the inadequacy of research on leukemia genetics in the Arab region in comparison to the Western studies. This finding highlights the need for extensive research in the Middle Eastern region to gain geographically heterogeneous genetic information about the Arab population. In conclusion, this scoping study highlights the genes associated with the major types of leukemia among the Arab population and also indicates the need for comprehensive and regionally balanced research on leukemia genetics in Middle Eastern countries. Addressing this gap is essential to provide robust genetic data that can be used for targeted interventions to improve leukemia outcomes in the Middle East. Increased research efforts in all Middle Eastern countries will contribute to a greater understanding of genetic predisposition and help develop effective prevention strategies and treatments tailored to this population.
Collapse
Affiliation(s)
- Abdulrahman Algarni
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar, SAU
| |
Collapse
|
6
|
Garcia C, Miller-Awe MD, Witkowski MT. Concepts in B cell acute lymphoblastic leukemia pathogenesis. J Leukoc Biol 2024; 116:18-32. [PMID: 38243586 DOI: 10.1093/jleuko/qiae015] [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: 10/26/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
B cell acute lymphoblastic leukemia (B-ALL) arises from genetic alterations impacting B cell progenitors, ultimately leading to clinically overt disease. Extensive collaborative efforts in basic and clinical research have significantly improved patient prognoses. Nevertheless, a subset of patients demonstrate resistance to conventional chemotherapeutic approaches and emerging immunotherapeutic interventions. This review highlights the mechanistic underpinnings governing B-ALL transformation. Beginning with exploring normative B cell lymphopoiesis, we delineate the influence of recurrent germline and somatic genetic aberrations on the perturbation of B cell progenitor differentiation and protumorigenic signaling, thereby facilitating the neoplastic transformation underlying B-ALL progression. Additionally, we highlight recent advances in the multifaceted landscape of B-ALL, encompassing metabolic reprogramming, microbiome influences, inflammation, and the discernible impact of socioeconomic and racial disparities on B-ALL transformation and patient survival.
Collapse
Affiliation(s)
- Clarissa Garcia
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| | - Megan D Miller-Awe
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| | - Matthew T Witkowski
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| |
Collapse
|
7
|
Chalise JP, Hu Z, Li M, Shepphird JK, Gu Z, Gyawali P, Itakura K, Larson GP. Identification of an alternative short ARID5B isoform associated with B-ALL survival. Biochem Biophys Res Commun 2024; 703:149659. [PMID: 38382358 DOI: 10.1016/j.bbrc.2024.149659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
Utilizing RNA sequence (RNA-Seq) splice junction data from a cohort of 1841 B-cell acute lymphoblastic leukemia (B-ALL) patients we define transcriptionally distinct isoforms of ARID5B, a risk-associated gene identified in genome wide association studies (GWAS), which associate with disease survival. Short (S) and long (L) ARID5B transcripts, which differ in an encoded BAH-like chromatin interaction domain, show remarkable correlation to the isoform splicing pattern. Testing of the ARID5B proximal promoter of the S & L isoforms indicated that both are functionally independent in luciferase reporter assays. Increased short isoform expression is associated with decreased event-free and overall survival. The abundance of short and long transcripts strongly correlates to B-ALL prognostic stratification, where B-ALL subtypes with poor outcomes express a higher proportion of the S-isoform. These data demonstrate that the analysis of independent promoters and alternative splicing events are essential for improved risk stratification and a more complete understanding of disease pathology.
Collapse
Affiliation(s)
- Jaya P Chalise
- Center for RNA Biology and Therapeutics, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Zunsong Hu
- Department of Systems Biology, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Min Li
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Jennifer K Shepphird
- Clinical Translational Project Development, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Zhaohui Gu
- Department of Systems Biology, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA; Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Purnima Gyawali
- Center for RNA Biology and Therapeutics, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Keiichi Itakura
- Center for RNA Biology and Therapeutics, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA
| | - Garrett P Larson
- Center for RNA Biology and Therapeutics, Beckman Research Institute of the City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
8
|
Deng Y, Dong Y, Wu L, Zhang Q, Yang L. ARID5B promoted the histone demethylation of SORBS2 and hampered the metastasis of ovarian cancer. Pathol Res Pract 2023; 252:154911. [PMID: 37948999 DOI: 10.1016/j.prp.2023.154911] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/18/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023]
Abstract
Ovarian cancer (OVCA) is the 4th most common female tumor after breast cancer, cervical cancer, and endometrial cancer, and now is mainly treated with debulking surgery and postoperative cisplatin and paclitaxel-based combination chemotherapy regimens. However, OVCA is insidious in its development and recurrence occurred in some patients after treatment. It is of great significance to study the pathogenesis of ovarian cancer and identify more biomarkers. Recently, the role of histone methyltransferase (HMT) and histone demethylase (HDM) in oncogenesis and development of malignant tumors has raised attention. Unlike other JMJC demethylases that have both JMJC and ARID domains in a single molecule, PHF2 requires assembly into a complex with a DNA-binding subunit (ARID5B) and exerts its enzymatic activity. Therefore, the aim of this manuscript is to investigate the role of histone demethylases ARID5B-PHF2 complex in the metastasis of OVCA. As result, we found ARID5B and PHF2 are both low expressed in OVCA tumor tissues and cell lines and associated with diagnosis and prognosis. Also, ARID5B suppressed rearrangement of the cytoskeleton in the process of EMT in OVCA cell lines. The role of PHF2 as a tumor suppressor was also confirmed both in vivo and in vitro. SORBS2 is low expressed in OVCA tumor tissues and cell lines and associated with diagnosis and prognosis. The expression of SORBS2 is positively corelated with the expression of ARID5B and PHF2. The promoter of SORBS2 is proved combined with ARID5B. The expression of SORBS2 was increased due to ARID5B-PHF2 complex promoted the histone demethylation by mainly binding in site H3K36me2 and therefore promoting the transcription of SORBS2. In conclusion, ARID5B-PHF2 complex promoted the histone demethylation of SORBS2 by mainly bind in site H3K36me2 and therefore promote the transcription of SORBS2 then hampered the process of EMT and tumor generation of OVCA. These results provided a new perspective on the molecular mechanisms of OVCA development and offered a new target of clinical diagnose and treatment of OVCA.
Collapse
Affiliation(s)
- Yue Deng
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Ying Dong
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Lu Wu
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Qin Zhang
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Lihua Yang
- Department of Gynecology,The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China.
| |
Collapse
|
9
|
Zhong C, Li S, Arroyo K, Morimoto LM, de Smith AJ, Metayer C, Ma X, Kogan SC, Gauderman WJ, Wiemels JL. Gene-Environment Analyses Reveal Novel Genetic Candidates with Prenatal Tobacco Exposure in Relation to Risk for Childhood Acute Lymphoblastic Leukemia. Cancer Epidemiol Biomarkers Prev 2023; 32:1707-1715. [PMID: 37773025 DOI: 10.1158/1055-9965.epi-23-0258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 08/02/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Associations between maternal tobacco exposure during pregnancy and childhood acute lymphoblastic leukemia (ALL) have yielded mixed results. This may be due to biases in self-reported smoking or other differences in individual-level risk factors. We utilized a biological marker of maternal tobacco exposure to evaluate the association between maternal tobacco exposure during pregnancy, genetics, and subsequent childhood ALL risk in two large population-based studies of childhood ALL in California. METHODS Maternal exposure to tobacco smoke was assessed with a validated methylation marker (cg05575921) of the aryl hydrocarbon receptor repressor (AHRR) gene in newborn dried blood spots. We adjusted for sex, birthweight, gestational age, mode of delivery, year of birth, AHRR quantitative trait locus (mQTL) rs77111113, and a polygenetic risk score for childhood ALL. We additionally adjusted for principal components in a gene-environment interaction testing method that incorporates gene-only and environment-only effects along with interactions. RESULTS AHRR hypomethylation overall was not associated with childhood ALL. In gene-environment interaction testing, several genetic variants displayed significant interaction with AHRR hypomethylation and childhood ALL. CONCLUSIONS Our results suggest that novel candidates in PTPRK and DPP6 may play a role in tobacco-related leukemogenesis. Further research is necessary to better understand the effects of tobacco and these variants on childhood ALL risk. IMPACT Despite the lack of an overall "main effect," tobacco exposure during pregnancy affects childhood ALL risk depending on specific genetic variants.
Collapse
Affiliation(s)
- Charlie Zhong
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Shaobo Li
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Katti Arroyo
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Libby M Morimoto
- School of Public Health, University of California, Berkeley, Berkeley, California
| | - Adam J de Smith
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Catherine Metayer
- School of Public Health, University of California, Berkeley, Berkeley, California
| | - Xiaomei Ma
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Scott C Kogan
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California
| | - W James Gauderman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Joseph L Wiemels
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
10
|
Al-Zayan NR, Ashour MJ, Abuwarda HN, Sharif FA. ARID5B, IKZF1, GATA3, CEBPE, and CDKN2A germline polymorphisms and predisposition to childhood acute lymphoblastic leukemia. Pediatr Hematol Oncol 2023; 41:103-113. [PMID: 37578068 DOI: 10.1080/08880018.2023.2234946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/10/2023] [Accepted: 06/21/2023] [Indexed: 08/15/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most frequent type of pediatric cancer. Germline single nucleotide polymorphisms (SNPs), including ARID5B (rs10821936 T/C), IKZF1 (rs4132601 T/G), GATA3 (rs3824662 G/T), CEBPE (rs2239633 G/A), and CDKN2A (rs3731217 A/C) have been linked to pediatric ALL in different populations. Hitherto, no previous studies have tested the relationship between these SNPs and pediatric ALL in Gaza strip. Therefore, we investigated the association between these polymorphisms and the occurrence of childhood ALL in this part of Palestine. This case-control study recruited 100 healthy controls and 78 ALL patients. Allele-specific PCR (AS-PCR) technique was used for SNPs genotyping. Relevant statistical tests were used and the multifactor dimensionality reduction (MDR) approach was applied in the analysis of gene-gene interactions. Minor alleles of ARID5B rs10821936 T/C (p = 0.007) and IKZF1 rs4132601 T/G (p = 0.045) were significantly higher in ALL patients. The homozygous (TT) genotype of GATA3 rs3824662 G/T (p = 0.038), (CC) of ARID5B rs10821936 T/C (p = 0.008), and (AC and CC) genotypes of CDKN2A rs3731217 A/C (p < 0.0001) were significantly higher in ALL cases. On MDR analysis, the best model for ALL risk was the five-factor model combination of the examined SNPs (CVC = 10/10; TBA = 0.632; p < 0.0001). This work demonstrates the association of ARID5B rs10821936 T/C, IKZF1 rs4132601 T/G, GATA3 rs3824662 G/T, and CDKN2A rs3731217 A/C polymorphisms with increased risk of pediatric ALL among a patient cohort from Gaza Strip. Further studies with a larger sample size are needed in order to confirm these findings and test the value of these SNPs in prognosis and treatment sensitivity.
Collapse
Affiliation(s)
| | | | | | - Fadel A Sharif
- Department of Medical Laboratory Sciences-IUG, Gaza, Palestine
| |
Collapse
|
11
|
Chalise JP, Ehsani A, Lemecha M, Hung YW, Zhang G, Larson GP, Itakura K. ARID5B regulates fatty acid metabolism and proliferation at the Pre-B cell stage during B cell development. Front Immunol 2023; 14:1170475. [PMID: 37483604 PMCID: PMC10360657 DOI: 10.3389/fimmu.2023.1170475] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
During B cell development in bone marrow, large precursor B cells (large Pre-B cells) proliferate rapidly, exit the cell cycle, and differentiate into non-proliferative (quiescent) small Pre-B cells. Dysregulation of this process may result in the failure to produce functional B cells and pose a risk of leukemic transformation. Here, we report that AT rich interacting domain 5B (ARID5B), a B cell acute lymphoblastic leukemia (B-ALL) risk gene, regulates B cell development at the Pre-B stage. In both mice and humans, we observed a significant upregulation of ARID5B expression that initiates at the Pre-B stage and is maintained throughout later stages of B cell development. In mice, deletion of Arid5b in vivo and ex vivo exhibited a significant reduction in the proportion of immature B cells but an increase in large and small Pre-B cells. Arid5b inhibition ex vivo also led to an increase in proliferation of both Pre-B cell populations. Metabolic studies in mouse and human bone marrow revealed that fatty acid uptake peaked in proliferative B cells then decreased during non-proliferative stages. We showed that Arid5b ablation enhanced fatty acid uptake and oxidation in Pre-B cells. Furthermore, decreased ARID5B expression was observed in tumor cells from B-ALL patients when compared to B cells from non-leukemic individuals. In B-ALL patients, ARID5B expression below the median was associated with decreased survival particularly in subtypes originating from Pre-B cells. Collectively, our data indicated that Arid5b regulates fatty acid metabolism and proliferation of Pre-B cells in mice, and reduced expression of ARID5B in humans is a risk factor for B cell leukemia.
Collapse
Affiliation(s)
- Jaya Prakash Chalise
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Ali Ehsani
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Mengistu Lemecha
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Yu-Wen Hung
- Immunology and Theranostics, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Guoxiang Zhang
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Garrett P. Larson
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Keiichi Itakura
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, CA, United States
| |
Collapse
|
12
|
Liu X, Xiao M, Xing Z, Jiang H, Zhu C, Zhang X, Li W, Wang Z, Wu F, Chen Y. Contributions of ARID5B, IKZF1, PIP4K2A, and GATA3 Gene Polymorphisms to Childhood Acute Lymphoblastic Leukemia in a Chinese Population. J Pediatr Hematol Oncol 2023; 45:123-129. [PMID: 36952466 DOI: 10.1097/mph.0000000000002646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/16/2023] [Indexed: 03/25/2023]
Abstract
Various studies have shown that single nucleotide polymorphisms in the AT-rich interaction domain 5B (ARID5B), IKAROS family zinc finger 1 (IKZF1), phosphatidylinositol-5-phosphate 4-kinase type 2 alpha (PIP4K2A), and GATA binding protein 3 (GATA3) genes may be associated with the susceptibility and prognosis of childhood acute lymphoblastic leukemia (ALL). The present study aimed to investigate the association of ARID5B rs10821936, IKZF1 rs4132601, PIP4K2A rs7088318, and GATA3 rs3824662 gene polymorphisms with the susceptibility and prognosis of childhood ALL in China. We found that the C allele of rs10821936 (ARID5B) and the A allele of rs3824662 (GATA3) were associated with an increased risk of childhood ALL in the Chinese population. There was no significant difference in frequencies of rs4132601 (IKZF1) and rs7088318 (PIP4K2A) genotypes and alleles between the childhood ALL and control groups. We observed that CC genotype of rs10821936 (ARID5B) was associated with increased rates of high-risk and moderate-risk childhood ALL. The rs10821936 (ARID5B) could serve as a potential biomarker for assessing the risk of childhood ALL in Chinese children.
Collapse
Affiliation(s)
| | - Min Xiao
- Department of Laboratory, Cancer Hospital Chinese Academy of Medical Science, Shenzhen Center, Shenzhen, Guangdong People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Tomás-Daza L, Rovirosa L, López-Martí P, Nieto-Aliseda A, Serra F, Planas-Riverola A, Molina O, McDonald R, Ghevaert C, Cuatrecasas E, Costa D, Camós M, Bueno C, Menéndez P, Valencia A, Javierre BM. Low input capture Hi-C (liCHi-C) identifies promoter-enhancer interactions at high-resolution. Nat Commun 2023; 14:268. [PMID: 36650138 PMCID: PMC9845235 DOI: 10.1038/s41467-023-35911-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Long-range interactions between regulatory elements and promoters are key in gene transcriptional control; however, their study requires large amounts of starting material, which is not compatible with clinical scenarios nor the study of rare cell populations. Here we introduce low input capture Hi-C (liCHi-C) as a cost-effective, flexible method to map and robustly compare promoter interactomes at high resolution. As proof of its broad applicability, we implement liCHi-C to study normal and malignant human hematopoietic hierarchy in clinical samples. We demonstrate that the dynamic promoter architecture identifies developmental trajectories and orchestrates transcriptional transitions during cell-state commitment. Moreover, liCHi-C enables the identification of disease-relevant cell types, genes and pathways potentially deregulated by non-coding alterations at distal regulatory elements. Finally, we show that liCHi-C can be harnessed to uncover genome-wide structural variants, resolve their breakpoints and infer their pathogenic effects. Collectively, our optimized liCHi-C method expands the study of 3D chromatin organization to unique, low-abundance cell populations, and offers an opportunity to uncover factors and regulatory networks involved in disease pathogenesis.
Collapse
Affiliation(s)
- Laureano Tomás-Daza
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
- Barcelona Supercomputing Center, Barcelona, Barcelona, Spain
| | - Llorenç Rovirosa
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
| | - Paula López-Martí
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
- Barcelona Supercomputing Center, Barcelona, Barcelona, Spain
| | | | - François Serra
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
| | | | - Oscar Molina
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
| | | | - Cedric Ghevaert
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Esther Cuatrecasas
- Pediatric Institute of Rare Diseases, Sant Joan de Déu Hospital, Esplugues de Llobregat, Barcelona, Spain
| | - Dolors Costa
- Hospital Clinic, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer, Barcelona, Spain
- Cancer Network Biomedical Research Center, Barcelona, Spain
| | - Mireia Camós
- Sant Joan de Déu Research Institute, Esplugues de Llobregat, Barcelona, Spain
- Sant Joan de Déu Hospital, Esplugues de Llobregat, Barcelona, Spain
- Center for Biomedical Research in the Rare Diseases Network (CIBERER), Carlos III Health Institute, Madrid, Spain
| | - Clara Bueno
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Alfonso Valencia
- Barcelona Supercomputing Center, Barcelona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Biola M Javierre
- Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain.
- Institute for Health Science Research Germans Trias i Pujol, Badalona, Barcelona, Spain.
| |
Collapse
|
14
|
Gamaleldin MA, Imbaby SAE. Association of two ARID5B gene variant single nucleotide polymorphisms with acute lymphoblastic leukemia in the Egyptian population. Asian Pac J Cancer Prev 2023; 24:337-343. [PMID: 36708585 PMCID: PMC10152858 DOI: 10.31557/apjcp.2023.24.1.337] [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: 09/10/2022] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND ARID5B SNPs have been linked to ALL in many research studies in which it was identified as a risk factor. From this context, we had great interest to investigate the relationship between ARID5B rs4948488 and ARID5B rs2893881 genotypes and ALL susceptibility and relapse in this study. MATERIALS AND METHODS Peripheral blood mononuclear cells were analyzed for ARID5B rs4948488 and rs2893881 gene polymorphisms by real-time quantitative polymerase chain reaction in 80 ALL patients and 80 controls. RESULTS Our results showed that the C/C genotype of ARID5B rs4948488 and A/G genotype and G-allele of rs2893881 were linked to higher ALL incidence. Regarding the relapse of ALL, rs4948488 C/C genotype and C-alleles were significantly associated with relapse of ALL. Meanwhile, rs4948488 C/C genotype and rs2893881 A/A genotype and A-allele are associated with T-ALL, while rs2893881 A/G genotype and G-allele are associated with B-ALL. CONCLUSION The results of our study suggested that ARID5B rs4948488 and rs2893881 SNPs might be used risk factors for genetic susceptibility for B-ALL and T-ALL, and that ARID5B s4948488 is related to relapse in ALL patients.<br />.
Collapse
|
15
|
Identification of Potential Treatments for Acute Lymphoblastic Leukemia through Integrated Genomic Network Analysis. Pharmaceuticals (Basel) 2022; 15:ph15121562. [PMID: 36559013 PMCID: PMC9786277 DOI: 10.3390/ph15121562] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The advancement of high-throughput sequencing and genomic analysis revealed that acute lymphoblastic leukemia (ALL) is a genetically heterogeneous disease. The abundance of such genetic data in ALL can also be utilized to identify potential targets for drug discovery and even drug repurposing. We aimed to determine potential genes for drug development and further guide the identification of candidate drugs repurposed for treating ALL through integrated genomic network analysis. Genetic variants associated with ALL were retrieved from the GWAS Catalog. We further applied a genomic-driven drug repurposing approach based on the six functional annotations to prioritize crucial biological ALL-related genes based on the scoring system. Lastly, we identified the potential drugs in which the mechanisms overlapped with the therapeutic targets and prioritized the candidate drugs using Connectivity Map (CMap) analysis. Forty-two genes were considered biological ALL-risk genes with ARID5B topping the list. Based on potentially druggable genes that we identified, palbociclib, sirolimus, and tacrolimus were under clinical trial for ALL. Additionally, chlorprothixene, sirolimus, dihydroergocristine, papaverine, and tamoxifen are the top five drug repositioning candidates for ALL according to the CMap score with dasatinib as a comparator. In conclusion, this study determines the practicability and the potential of integrated genomic network analysis in driving drug discovery in ALL.
Collapse
|
16
|
Xu M, Wu S, Wang Y, Zhao Y, Wang X, Wei C, Liu X, Hao F, Hu C. Association between high-dose methotrexate-induced toxicity and polymorphisms within methotrexate pathway genes in acute lymphoblastic leukemia. Front Pharmacol 2022; 13:1003812. [PMID: 36532750 PMCID: PMC9748425 DOI: 10.3389/fphar.2022.1003812] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/21/2022] [Indexed: 09/22/2023] Open
Abstract
Methotrexate (MTX) is a folic acid antagonist, the mechanism of action is to inhibit DNA synthesis, repair and cell proliferation by decreasing the activities of several folate-dependent enzymes. It is widely used as a chemotherapy drug for children and adults with malignant tumors. High-dose methotrexate (HD-MTX) is an effective treatment for extramedullary infiltration and systemic consolidation in children with acute lymphoblastic leukemia (ALL). However, significant toxicity results in most patients treated with HD-MTX, which limits its use. HD-MTX-induced toxicity is heterogeneous, and this heterogeneity may be related to gene polymorphisms in related enzymes of the MTX intracellular metabolic pathway. To gain a deeper understanding of the differences in toxicity induced by HD-MTX in individuals, the present review examines the correlation between HD-MTX-induced toxicity and the gene polymorphisms of related enzymes in the MTX metabolic pathway in ALL. In this review, we conclude that only the association of SLCO1B1 and ARID5B gene polymorphisms with plasma levels of MTX and MTX-related toxicity is clearly described. These results suggest that SLCO1B1 and ARID5B gene polymorphisms should be evaluated before HD-MTX treatment. In addition, considering factors such as age and race, the other exact predictor of MTX induced toxicity in ALL needs to be further determined.
Collapse
Affiliation(s)
- Meng Xu
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
- School of Laboratory Medicine, Beihua University, Jilin, China
| | - Shuangshuang Wu
- Department of Pediatric Hematology, The First Hospital of Jilin University, Changchun, China
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yue Wang
- Department of Pediatric Hematology, The First Hospital of Jilin University, Changchun, China
| | - Yundong Zhao
- School of Laboratory Medicine, Beihua University, Jilin, China
| | - Ximin Wang
- Jilin Drug Inspection Center, Changchun, China
| | - Changhong Wei
- Department of Hematology, The Linyi Central Hospital, Linyi, China
| | - Xueying Liu
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
- School of Laboratory Medicine, Beihua University, Jilin, China
| | - Feng Hao
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Cheng Hu
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| |
Collapse
|
17
|
David K, Friedlander G, Pellegrino B, Radomir L, Lewinsky H, Leng L, Bucala R, Becker-Herman S, Shachar I. CD74 as a regulator of transcription in normal B cells. Cell Rep 2022; 41:111572. [DOI: 10.1016/j.celrep.2022.111572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/07/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
|
18
|
Cavazos TB, Kachuri L, Graff RE, Nierenberg JL, Thai KK, Alexeeff S, Van Den Eeden S, Corley DA, Kushi LH, Hoffmann TJ, Ziv E, Habel LA, Jorgenson E, Sakoda LC, Witte JS. Assessment of genetic susceptibility to multiple primary cancers through whole-exome sequencing in two large multi-ancestry studies. BMC Med 2022; 20:332. [PMID: 36199081 PMCID: PMC9535845 DOI: 10.1186/s12916-022-02535-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Up to one of every six individuals diagnosed with one cancer will be diagnosed with a second primary cancer in their lifetime. Genetic factors contributing to the development of multiple primary cancers, beyond known cancer syndromes, have been underexplored. METHODS To characterize genetic susceptibility to multiple cancers, we conducted a pan-cancer, whole-exome sequencing study of individuals drawn from two large multi-ancestry populations (6429 cases, 165,853 controls). We created two groupings of individuals diagnosed with multiple primary cancers: (1) an overall combined set with at least two cancers across any of 36 organ sites and (2) cancer-specific sets defined by an index cancer at one of 16 organ sites with at least 50 cases from each study population. We then investigated whether variants identified from exome sequencing were associated with these sets of multiple cancer cases in comparison to individuals with one and, separately, no cancers. RESULTS We identified 22 variant-phenotype associations, 10 of which have not been previously discovered and were significantly overrepresented among individuals with multiple cancers, compared to those with a single cancer. CONCLUSIONS Overall, we describe variants and genes that may play a fundamental role in the development of multiple primary cancers and improve our understanding of shared mechanisms underlying carcinogenesis.
Collapse
Affiliation(s)
- Taylor B Cavazos
- Biological and Medical Informatics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Linda Kachuri
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, USA.,Department of Epidemiology and Population Health, Stanford University, Alway Building, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Rebecca E Graff
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, USA.,Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Jovia L Nierenberg
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, USA.,Regeneron Genetics Center, Tarrytown, NY, 10591, USA
| | - Khanh K Thai
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Stacey Alexeeff
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Stephen Van Den Eeden
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Douglas A Corley
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Lawrence H Kushi
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | | | - Thomas J Hoffmann
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Elad Ziv
- Regeneron Genetics Center, Tarrytown, NY, 10591, USA
| | - Laurel A Habel
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Eric Jorgenson
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Lori C Sakoda
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA.,Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, 91101, USA
| | - John S Witte
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, USA. .,Department of Epidemiology and Population Health, Stanford University, Alway Building, 300 Pasteur Drive, Stanford, CA, 94305, USA. .,Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
19
|
Chen Z, Fu S, Shan Y, Li H, Wang H, Liu J, Wang W, Huang Y, Huang H, Wang J, Ding M. Hsa_circ_0102485 inhibits the growth of cancer cells by regulating the miR-188-3p/ARID5B/AR axis in prostate carcinoma. Pathol Res Pract 2022; 237:154052. [DOI: 10.1016/j.prp.2022.154052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/17/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022]
|
20
|
Comprehensive Landscape of ARID Family Members and Their Association with Prognosis and Tumor Microenvironment in Hepatocellular Carcinoma. J Immunol Res 2022; 2022:1688460. [PMID: 35402625 PMCID: PMC8986425 DOI: 10.1155/2022/1688460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
As one of the most lethal forms of cancers, hepatocellular carcinoma (HCC) claims many lives around the world, and it is especially common in China. The ARID family plays key roles in the pathogenesis and development of human cancers. The potential of several functional genes used as novel biomarkers has attracted more and more attention. However, the prognostic values of the ARID family in HCC patients are rarely known by people. In this study, we performed comprehensive analysis using TCGA datasets, finding that the expressions of ARID4B, ARID2, ARID3B, JARID2, ARID1A, ARID1B, and ARID3A were increased in HCC specimens compared to nontumor specimens, while the expressions of ARID4A and ARID3C were decreased in HCC specimens. According to the Pearson correlation data, the methylation levels of the majority of ARID members were negatively correlated. Upregulation of ARID3A, ARID5B, and ARID1A was related to a poor HCC outcome according to the data of multivariate assays. Then, we built a LASSO Cox regression model based on ARID3A, ARID5B, and ARID1A in HCC. Overall survival rates were considerably lower for those with high risk scores compared to those with low risk scores. Finally, we studied the associations between risk scores and several types of infiltrating immune cells. The data revealed that the risk score was positively related to the infiltration of CD8+ T cells, B cell, T cell CD8+, neutrophil, macrophage, and myeloid dendritic cell. This study conducted a thorough analysis of the ARID members, resulting in new insights for further examination of the ARID family members as prospective targets in the treatment of HCC.
Collapse
|
21
|
Huang D, Ovcharenko I. Enhancer-silencer transitions in the human genome. Genome Res 2022; 32:437-448. [PMID: 35105669 PMCID: PMC8896465 DOI: 10.1101/gr.275992.121] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 01/27/2022] [Indexed: 11/24/2022]
Abstract
Dual-function regulatory elements (REs), acting as enhancers in some cellular contexts and as silencers in others, have been reported to facilitate the precise gene regulatory response to developmental signals in Drosophila melanogaster. However, with few isolated examples detected, dual-function REs in mammals have yet to be systematically studied. We herein investigated this class of REs in the human genome and profiled their activity across multiple cell types. Focusing on enhancer–silencer transitions specific to the development of T cells, we built an accurate deep learning classifier of REs and identified about 12,000 silencers active in primary peripheral blood T cells that act as enhancers in embryonic stem cells. Compared with regular silencers, these dual-function REs are evolving under stronger purifying selection and are enriched for mutations associated with disease phenotypes and altered gene expression. In addition, they are enriched in the loci of transcriptional regulators, such as transcription factors (TFs) and chromatin remodeling genes. Dual-function REs consist of two intertwined but largely distinct sets of binding sites bound by either activating or repressing TFs, depending on the type of RE function in a given cell line. This indicates the recruitment of different TFs for different regulatory modes and a complex DNA sequence composition of these REs with dual activating and repressive encoding. With an estimated >6% of cell type–specific human silencers acting as dual-function REs, this overlooked class of REs requires a specific investigation on how their inherent functional plasticity might be a contributing factor to human diseases.
Collapse
|
22
|
García-Padilla C, Dueñas Á, García-López V, Aránega A, Franco D, Garcia-Martínez V, López-Sánchez C. Molecular Mechanisms of lncRNAs in the Dependent Regulation of Cancer and Their Potential Therapeutic Use. Int J Mol Sci 2022; 23:764. [PMID: 35054945 PMCID: PMC8776057 DOI: 10.3390/ijms23020764] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 12/31/2021] [Accepted: 01/08/2022] [Indexed: 12/16/2022] Open
Abstract
Deep whole genome and transcriptome sequencing have highlighted the importance of an emerging class of non-coding RNA longer than 200 nucleotides (i.e., long non-coding RNAs (lncRNAs)) that are involved in multiple cellular processes such as cell differentiation, embryonic development, and tissue homeostasis. Cancer is a prime example derived from a loss of homeostasis, primarily caused by genetic alterations both in the genomic and epigenetic landscape, which results in deregulation of the gene networks. Deregulation of the expression of many lncRNAs in samples, tissues or patients has been pointed out as a molecular regulator in carcinogenesis, with them acting as oncogenes or tumor suppressor genes. Herein, we summarize the distinct molecular regulatory mechanisms described in literature in which lncRNAs modulate carcinogenesis, emphasizing epigenetic and genetic alterations in particular. Furthermore, we also reviewed the current strategies used to block lncRNA oncogenic functions and their usefulness as potential therapeutic targets in several carcinomas.
Collapse
Affiliation(s)
- Carlos García-Padilla
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (Á.D.); (A.A.); (D.F.)
- Department of Human Anatomy and Embryology, University of Extremadura, 06006 Badajoz, Spain; (V.G.-L.); (V.G.-M.)
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Ángel Dueñas
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (Á.D.); (A.A.); (D.F.)
- Department of Human Anatomy and Embryology, University of Extremadura, 06006 Badajoz, Spain; (V.G.-L.); (V.G.-M.)
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Virginio García-López
- Department of Human Anatomy and Embryology, University of Extremadura, 06006 Badajoz, Spain; (V.G.-L.); (V.G.-M.)
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Amelia Aránega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (Á.D.); (A.A.); (D.F.)
- Fundación Medina, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (Á.D.); (A.A.); (D.F.)
- Fundación Medina, 18016 Granada, Spain
| | - Virginio Garcia-Martínez
- Department of Human Anatomy and Embryology, University of Extremadura, 06006 Badajoz, Spain; (V.G.-L.); (V.G.-M.)
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Carmen López-Sánchez
- Department of Human Anatomy and Embryology, University of Extremadura, 06006 Badajoz, Spain; (V.G.-L.); (V.G.-M.)
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| |
Collapse
|
23
|
Taylor ZL, Vang J, Lopez-Lopez E, Oosterom N, Mikkelsen T, Ramsey LB. Systematic Review of Pharmacogenetic Factors That Influence High-Dose Methotrexate Pharmacokinetics in Pediatric Malignancies. Cancers (Basel) 2021; 13:cancers13112837. [PMID: 34200242 PMCID: PMC8201112 DOI: 10.3390/cancers13112837] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Methotrexate (MTX) is a mainstay therapeutic agent administered at high doses for the treatment of pediatric and adult malignancies, such as acute lymphoblastic leukemia, osteosarcoma, and lymphoma. Despite the vast evidence for clinical efficacy, high-dose MTX displays significant inter-individual pharmacokinetic variability. Delayed MTX clearance can lead to prolonged, elevated exposure, causing increased risks for nephrotoxicity, mucositis, seizures, and neutropenia. Numerous pharmacogenetic studies have investigated the effects of several genes and polymorphisms on MTX clearance in an attempt to better understand the pharmacokinetic variability and improve patient outcomes. To date, several genes and polymorphisms that affect MTX clearance have been identified. However, evidence for select genes have conflicting results or lack the necessary replication and validation needed to confirm their effects on MTX clearance. Therefore, we performed a systematic review to identify and then summarize the pharmacogenetic factors that influence high-dose MTX pharmacokinetics in pediatric malignancies. Using the PRISMA guidelines, we analyzed 58 articles and 24 different genes that were associated with transporter pharmacology or the folate transport pathway. We conclude that there is only one gene that reliably demonstrates an effect on MTX pharmacokinetics: SLCO1B1.
Collapse
Affiliation(s)
- Zachary L. Taylor
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH 45267, USA;
- Division of Research in Patient Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jesper Vang
- Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark;
- Paediatric Oncology Research Laboratory, University Hospital of Copenhagen, Rigshospitalet Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Elixabet Lopez-Lopez
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain;
- Pediatric Oncology Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Natanja Oosterom
- Princess Máxima Center for Pediatric Oncology, 3720 Utrecht, The Netherlands;
| | - Torben Mikkelsen
- Department of Pediatric Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Laura B. Ramsey
- Division of Research in Patient Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Correspondence: ; Tel.: +1-513-803-8963
| |
Collapse
|
24
|
Hao Q, Cao M, Zhang C, Yin D, Wang Y, Ye Y, Zhao S, Yang Y, Chen KL, Ying B, Wang L, Zhang Y, Xu C, Zhu Y, Wu Y, Gao J, Zhao JN, Zhang Y, Lu X. Age-related differences of genetic susceptibility to patients with acute lymphoblastic leukemia. Aging (Albany NY) 2021; 13:12456-12465. [PMID: 33891562 PMCID: PMC8148462 DOI: 10.18632/aging.202903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/05/2021] [Indexed: 02/05/2023]
Abstract
Inherited predispositions to acute lymphoblastic leukemia have been well investigated in pediatric patients, but studies on adults, particularly Chinese patients, are limited. In this study, we conducted a genome-wide association study in 466 all-age Chinese patients with Acute lymphoblastic leukemia (ALL) and 1,466 non-ALL controls to estimate the impact of age on ALL susceptibility in the Chinese population. Among the 17 reported loci, 8 have been validated in pediatric and 1 in adult patients. The strongest association signal was identified at ARID5B locus and gradually decreased with age, while the signal at GATA3 exhibited the opposite trend and significantly impact on adult patients. With genome-wide approaches, germline variants at 2q14.3 rank as the top inherited predisposition to adult patients (e.g., rs73956024, P = 4.3 × 10-5) and separate the genetic risk of pediatric vs. adult patients (P = 3.6 × 10-6), whereas variants at 15q25.3 (e.g., rs11638062) have a similar impact on patients in different age groups (overall P = 2.9 × 10-7). Our analysis highlights the impact of age on genetic susceptibility to ALL in Chinese patients.
Collapse
Affiliation(s)
- Qing Hao
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,College of Pharmaceutical Sciences, Southwest Medical University, Luzhou, China.,Sichuan Center for Translational Medicine of Traditional Chinese Medicine, Institute of Translational Pharmacology, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Minyuan Cao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chunlan Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Dandan Yin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yuelan Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanxin Ye
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Zhao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yunfan Yang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Ke-Ling Chen
- Digestive Surgery Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lanlan Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yiguan Zhang
- Sichuan Center for Translational Medicine of Traditional Chinese Medicine, Institute of Translational Pharmacology, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Caigang Xu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yiping Zhu
- Department of Hematology/Oncology, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yu Wu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Ju Gao
- Department of Hematology/Oncology, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jun-Ning Zhao
- Sichuan Center for Translational Medicine of Traditional Chinese Medicine, Institute of Translational Pharmacology, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Yan Zhang
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Lu
- Department of Hematology/Oncology, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Regulation of the MIE Locus During HCMV Latency and Reactivation. Pathogens 2020; 9:pathogens9110869. [PMID: 33113934 PMCID: PMC7690695 DOI: 10.3390/pathogens9110869] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesviral pathogen that results in life-long infection. HCMV maintains a latent or quiescent infection in hematopoietic cells, which is broadly defined by transcriptional silencing and the absence of de novo virion production. However, upon cell differentiation coupled with immune dysfunction, the virus can reactivate, which leads to lytic replication in a variety of cell and tissue types. One of the mechanisms controlling the balance between latency and reactivation/lytic replication is the regulation of the major immediate-early (MIE) locus. This enhancer/promoter region is complex, and it is regulated by chromatinization and associated factors, as well as a variety of transcription factors. Herein, we discuss these factors and how they influence the MIE locus, which ultimately impacts the phase of HCMV infection.
Collapse
|