1
|
Zhang H, Yan Y, Yi S, Sun Q. Advancements in targeting CD30 for lymphoma therapy: a historical perspective and future directions. Expert Rev Hematol 2025:1-14. [PMID: 40227173 DOI: 10.1080/17474086.2025.2492936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION CD30 is a transmembrane protein of the tumor necrosis factor receptor superfamily. It is expressed on a small subset of activated T and B lymphocytes, and various lymphoid neoplasms, including classical Hodgkin lymphoma and many non-Hodgkin lymphomas in both pediatric and adult populations. AREAS COVERED This review delves into the significance of CD30 as a therapeutic target and a prognostic indicator for various lymphomas. It provides a comprehensive overview of anti-CD30 therapeutic interventions developed to date, offering insights into the future direction of lymphoma treatment research. Literature search was conducted from January 1987 to December 2024 using PubMed, Scopus, and Web of Science databases to identify relevant studies. EXPERT OPINION CD30 has emerged as a critical marker of diagnosis, prognosis, and therapeutic strategies of lymphomas. The introduction of brentuximab vedotin (BV) (Adcetris), an antibody-drug conjugate targeting CD30, has significantly advanced the treatment landscape for multiple lymphoma types, demonstrating enhanced efficacy and manageable safety profiles in CD30+ lymphomas patients. However, drug resistance is observed in few patients. Concurrently, innovative therapeutic strategies targeting CD30, such as chimeric antigen receptor T-cells therapies and bispecific antibodies, are in development. This underscores a strong and ongoing research effort aimed at improving the management of patients with CD30+ lymphomas.
Collapse
Affiliation(s)
- Hongju Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Hematologic Pathology Center, Tianjin Institutes of Health Science, Tianjin, China
| | - Yuting Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Lymphoma Diagnosis and Treatment Center, Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Lymphoma Diagnosis and Treatment Center, Tianjin Institutes of Health Science, Tianjin, China
| | - Qi Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Hematologic Pathology Center, Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
2
|
Li Z, Guo W, Bai O. Mechanism of action and therapeutic targeting of CD30 molecule in lymphomas. Front Oncol 2023; 13:1301437. [PMID: 38188299 PMCID: PMC10767573 DOI: 10.3389/fonc.2023.1301437] [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] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
At present, the treatment of lymphoma has entered the era of precision medicine, and CD30, as a transmembrane protein, has become an important marker to help the diagnosis and formulation of treatment plans for lymphomas. This protein is widely expressed in various types of lymphomas and can play a role through nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), and other pathways, and ultimately lead to the up-regulation of CD30 expression to give tumor cells a survival advantage. Brentuximab vedotin (BV), as an antibody-drug conjugate (ADC) targeting CD30, is one of the first new drugs to significantly improve survival in patients with CD30+lymphomas. However, the biological function of CD30 has not been fully elucidated. Therefore, this review highlights the CD30-mediated tumor-promoting mechanisms and the molecular factors that regulate CD30 expression. We hope that a better understanding of CD30 biology will provide new insights into clinical treatment and improve the survival and quality of life of lymphoma patients.
Collapse
Affiliation(s)
| | | | - Ou Bai
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Katsin M, Dormeshkin D, Meleshko A, Migas A, Dubovik S, Konoplya N. CAR-T Cell Therapy for Classical Hodgkin Lymphoma. Hemasphere 2023; 7:e971. [PMID: 38026793 PMCID: PMC10656097 DOI: 10.1097/hs9.0000000000000971] [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: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 12/01/2023] Open
Abstract
Classical Hodgkin lymphoma (cHL) is a malignancy characterized by the presence of Hodgkin and Reed-Sternberg (HRS) cells within a complex tumor microenvironment (TME). Despite advances in conventional therapies, a subset of cHL patients experience relapse or refractory disease, necessitating the exploration of novel treatment strategies. Chimeric antigen receptor T cell (CAR-T cell) therapy has emerged as a promising approach for the management of cHL, harnessing the power of genetically modified T cells to recognize and eliminate tumor cells. In this article, we provide an overview of the pathogenesis of cHL, highlighting the key molecular and cellular mechanisms involved. Additionally, we discuss the rationale for the development of CAR-T cell therapy in cHL, focusing on the identification of suitable targets on HRS cells (such as CD30, CD123, LMP1, and LMP2A), clonotypic lymphoma initiating B cells (CD19, CD20), and cells within the TME (CD123, CD19, CD20) for CAR-T cell design. Furthermore, we explore various strategies employed to enhance the efficacy and safety of CAR-T cell therapies in the treatment of cHL. Finally, we present an overview of the results obtained from clinical trials evaluating the efficacy of CAR-T cell therapies in cHL, highlighting their potential as a promising therapeutic option. Collectively, this article provides a comprehensive review of the current understanding of cHL pathogenesis and the rationale for CAR-T cell therapy development, offering insights into the future directions of this rapidly evolving field.
Collapse
Affiliation(s)
- Mikalai Katsin
- Vitebsk Regional Clinical Cancer Centre, Vitebsk, Belarus
| | - Dmitri Dormeshkin
- Institute of Bioorganic Chemistry of the National academy of Sciences of Belarus, Minsk, Belarus
| | - Alexander Meleshko
- Belarusian Research Center for Pediatric Oncology and Hematology, Minsk, Belarus
| | | | - Simon Dubovik
- Institute of Bioorganic Chemistry of the National academy of Sciences of Belarus, Minsk, Belarus
| | - Natalya Konoplya
- N.N. Alexandrov National Cancer Center of Belarus, Minsk, Belarus
| |
Collapse
|
4
|
Ren FJ, Cai XY, Yao Y, Fang GY. JunB: a paradigm for Jun family in immune response and cancer. Front Cell Infect Microbiol 2023; 13:1222265. [PMID: 37731821 PMCID: PMC10507257 DOI: 10.3389/fcimb.2023.1222265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Jun B proto-oncogene (JunB) is a crucial member of dimeric activator protein-1 (AP-1) complex, which plays a significant role in various physiological processes, such as placental formation, cardiovascular development, myelopoiesis, angiogenesis, endochondral ossification and epidermis tissue homeostasis. Additionally, it has been reported that JunB has great regulatory functions in innate and adaptive immune responses by regulating the differentiation and cytokine secretion of immune cells including T cells, dendritic cells and macrophages, while also facilitating the effector of neutrophils and natural killer cells. Furthermore, a growing body of studies have shown that JunB is involved in tumorigenesis through regulating cell proliferation, differentiation, senescence and metastasis, particularly affecting the tumor microenvironment through transcriptional promotion or suppression of oncogenes in tumor cells or immune cells. This review summarizes the physiological function of JunB, its immune regulatory function, and its contribution to tumorigenesis, especially focusing on its regulatory mechanisms within tumor-associated immune processes.
Collapse
Affiliation(s)
- Fu-jia Ren
- Department of Pharmacy, Hangzhou Women’s Hospital, Hangzhou, Zhejiang, China
| | - Xiao-yu Cai
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Yao
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guo-ying Fang
- Department of Pharmacy, Hangzhou Women’s Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Nakashima M, Uchimaru K. CD30 Expression and Its Functions during the Disease Progression of Adult T-Cell Leukemia/Lymphoma. Int J Mol Sci 2023; 24:ijms24108731. [PMID: 37240076 DOI: 10.3390/ijms24108731] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
CD30, a member of the tumor necrosis factor receptor superfamily, plays roles in pro-survival signal induction and cell proliferation in peripheral T-cell lymphoma (PTCL) and adult T-cell leukemia/lymphoma (ATL). Previous studies have identified the functional roles of CD30 in CD30-expressing malignant lymphomas, not only PTCL and ATL, but also Hodgkin lymphoma (HL), anaplastic large cell lymphoma (ALCL), and a portion of diffuse large B-cell lymphoma (DLBCL). CD30 expression is often observed in virus-infected cells such as human T-cell leukemia virus type 1 (HTLV-1). HTLV-1 is capable of immortalizing lymphocytes and producing malignancy. Some ATL cases caused by HTLV-1 infection overexpress CD30. However, the molecular mechanism-based relationship between CD30 expression and HTLV-1 infection or ATL progression is unclear. Recent findings have revealed super-enhancer-mediated overexpression at the CD30 locus, CD30 signaling via trogocytosis, and CD30 signaling-induced lymphomagenesis in vivo. Successful anti-CD30 antibody-drug conjugate (ADC) therapy for HL, ALCL, and PTCL supports the biological significance of CD30 in these lymphomas. In this review, we discuss the roles of CD30 overexpression and its functions during ATL progression.
Collapse
Affiliation(s)
- Makoto Nakashima
- Laboratory of Tumor Cell Biology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo 1088639, Japan
| | - Kaoru Uchimaru
- Laboratory of Tumor Cell Biology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo 1088639, Japan
| |
Collapse
|
6
|
Sun G, Yang Y, Liu J, Gao Z, Xu T, Chai J, Xu J, Fan Z, Xiao T, Jia Q, Li M. Cancer stem cells in esophageal squamous cell carcinoma. Pathol Res Pract 2022; 237:154043. [DOI: 10.1016/j.prp.2022.154043] [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: 06/21/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023]
|
7
|
Wang Y, He J, Xu M, Xue Q, Zhu C, Liu J, Zhang Y, Shi W. Holistic View of ALK TKI Resistance in ALK-Positive Anaplastic Large Cell Lymphoma. Front Oncol 2022; 12:815654. [PMID: 35211406 PMCID: PMC8862178 DOI: 10.3389/fonc.2022.815654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/04/2022] [Indexed: 11/23/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase expressed at early stages of normal development and in various cancers including ALK-positive anaplastic large cell lymphoma (ALK+ ALCL), in which it is the main therapeutic target. ALK tyrosine kinase inhibitors (ALK TKIs) have greatly improved the prognosis of ALK+ALCL patients, but the emergence of drug resistance is inevitable and limits the applicability of these drugs. Although various mechanisms of resistance have been elucidated, the problem persists and there have been relatively few relevant clinical studies. This review describes research progress on ALK+ ALCL including the application and development of new therapies, especially in relation to drug resistance. We also propose potential treatment strategies based on current knowledge to inform the design of future clinical trials.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China.,Nantong University School of Medicine, Nantong, China
| | - Jing He
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China.,Nantong University School of Medicine, Nantong, China
| | - Manyu Xu
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, China
| | - Qingfeng Xue
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Cindy Zhu
- Department of Psychology, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Juan Liu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China.,Nantong University School of Medicine, Nantong, China
| | - Yaping Zhang
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wenyu Shi
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
8
|
Wu Z, Nicoll M, Ingham RJ. AP-1 family transcription factors: a diverse family of proteins that regulate varied cellular activities in classical hodgkin lymphoma and ALK+ ALCL. Exp Hematol Oncol 2021; 10:4. [PMID: 33413671 PMCID: PMC7792353 DOI: 10.1186/s40164-020-00197-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/07/2023] Open
Abstract
Classical Hodgkin lymphoma (cHL) and anaplastic lymphoma kinase-positive, anaplastic large cell lymphoma (ALK+ ALCL) are B and T cell lymphomas respectively, which express the tumour necrosis factor receptor superfamily member, CD30. Another feature shared by cHL and ALK+ ALCL is the aberrant expression of multiple members of the activator protein-1 (AP-1) family of transcription factors which includes proteins of the Jun, Fos, ATF, and Maf subfamilies. In this review, we highlight the varied roles these proteins play in the pathobiology of these lymphomas including promoting proliferation, suppressing apoptosis, and evading the host immune response. In addition, we discuss factors contributing to the elevated expression of these transcription factors in cHL and ALK+ ALCL. Finally, we examine therapeutic strategies for these lymphomas that exploit AP-1 transcriptional targets or the signalling pathways they regulate.
Collapse
Affiliation(s)
- Zuoqiao Wu
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.17063.330000 0001 2157 2938Present Address: Department of Medicine, University of Toronto, Toronto, Canada
| | - Mary Nicoll
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada ,grid.14709.3b0000 0004 1936 8649Present Address: Department of Biology, McGill University, Montreal, Canada
| | - Robert J. Ingham
- grid.17089.37Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| |
Collapse
|
9
|
Larose H, Burke GAA, Lowe EJ, Turner SD. From bench to bedside: the past, present and future of therapy for systemic paediatric ALCL, ALK. Br J Haematol 2019; 185:1043-1054. [PMID: 30681723 DOI: 10.1111/bjh.15763] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Anaplastic large cell lymphoma (ALCL) is a T cell Non-Hodgkin Lymphoma that mainly presents in paediatric and young adult patients. The majority of cases express a chimeric fusion protein resulting in hyperactivation of anaplastic lymphoma kinase (ALK) as the consequence of a chromosomal translocation. Rarer cases lack expression of ALK fusion proteins and are categorised as ALCL, ALK-. An adapted regimen of an historic chemotherapy backbone is still used to this day, yielding overall survival (OS) of over 90% but with event-free survival (EFS) at an unacceptable 70%, improving little over the past 30 years. It is clear that continued adaption of current therapies will probably not improve these statistics and, for progress to be made, integration of biology with the design and implementation of future clinical trials is required. Indeed, advances in our understanding of the biology of ALCL are outstripping our ability to clinically translate them; laboratory-based research has highlighted a plethora of potential therapeutic targets but, with high survival rates combined with a scarcity of funding and patients to implement paediatric trials of novel agents, progress is slow. However, advances must be made to reduce the side-effects of intensive chemotherapy regimens whilst maintaining, if not improving, OS and EFS.
Collapse
Affiliation(s)
- Hugo Larose
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, UK.,European Research Initiative for ALK-related malignancies (www.erialcl.net), Cambridge, UK
| | - G A Amos Burke
- Department of paediatric oncology, Addenbrooke's Hospital, Cambridge, UK
| | - Eric J Lowe
- Division of Pediatric Hematology-Oncology, Children's Hospital of the Kings Daughter, Norfolk, Virginia, USA
| | - Suzanne D Turner
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge, UK.,European Research Initiative for ALK-related malignancies (www.erialcl.net), Cambridge, UK
| |
Collapse
|
10
|
Gonzalez-Fierro A, Dueñas-González A. Emerging DNA methylation inhibitors for cancer therapy: challenges and prospects. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019; 4:27-35. [DOI: 10.1080/23808993.2019.1571906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/16/2019] [Indexed: 10/27/2022]
Affiliation(s)
| | - Alfonso Dueñas-González
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM/Instituto Nacional de Can cerología, México City, Mexico
| |
Collapse
|
11
|
Wezler X, Dübel S, Schirrmann T. Antibody fusion proteins with human ribonucleases 1 to 8. Hum Antibodies 2018; 26:177-192. [PMID: 29689715 DOI: 10.3233/hab-180337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
ImmunoRNases combine tumor targeting by antibodies with the cytotoxic action of ribonucleases from the RNase A superfamily. This study investigated for the first time all catalytic active human RNase A family members (1 to 8) as effector components of antibody fusion proteins. ImmunoRNase fusion proteins were constructed using the CD30-specific bivalent recombinant scFv-Fc antibody SH313-B5. Production of the resulting entirely human immunoRNases 1 to 8 was done in mammalian cells by secretion of active forms. The immunoRNases mediated CD30-specific cell binding and showed ribonucleolytic activity. Interestingly, immunoRNases 1 and 2 were active in the presence of up to 5-/20-fold molar excess of the pancreatic RNase inhibitor (RI), which is supposed to efficiently inhibit all human RNase A activity. ImmunoRNases 3, 4, 6 and 7 were only inhibited by several fold molar excess of RI, whereas immunoRNases 5 and 8 were already completely inactive at equimolar RI concentrations. Compared to free RNases, activity and RI sensitivity were not significantly changed by antibody fusion or dimerisation. ImmunoRNase3 and 5 mediated tumor growth inhibition at low nanomolar concentrations. Anti-tumor activity was antigen-specific and did not show any correlation with ribonucleolytic activity or RI sensitivity.
Collapse
Affiliation(s)
- Xenia Wezler
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, 38106 Braunschweig, Germany
| | - Stefan Dübel
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, 38106 Braunschweig, Germany
| | | |
Collapse
|
12
|
Wei J, Li G, Zhang J, Zhou Y, Dang S, Chen H, Wu Q, Liu M. Integrated analysis of genome-wide DNA methylation and gene expression profiles identifies potential novel biomarkers of rectal cancer. Oncotarget 2018; 7:62547-62558. [PMID: 27566576 PMCID: PMC5308745 DOI: 10.18632/oncotarget.11534] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 08/08/2016] [Indexed: 12/18/2022] Open
Abstract
DNA methylation was regarded as the promising biomarker for rectal cancer diagnosis. However, the optimal methylation biomarkers with ideal diagnostic performance for rectal cancer are still limited. To identify new molecular markers for rectal cancer, we mapped DNA methylation and transcriptomic profiles in the six rectal cancer and paired normal samples. Further analysis revealed the hypermethylated probes in cancer prone to be located in gene promoter. Meanwhile, transcriptome analysis presented 773 low-expressed and 1,161 over-expressed genes in rectal cancer. Correction analysis identified a panel of 36 genes with an inverse correlation between methylation and gene expression levels, including 10 known colorectal cancer related genes. From the other 26 novel marker genes, GFRA1 and GSTM2 were selected for further analysis on the basis of their biological functions. Further experiment analysis confirmed their methylation and expression status in a larger number (44) of rectal cancer samples, and ROC curves showed higher AUC than SEPT9, which has been used as a biomarker in rectal cancer. Our data suggests that aberrant DNA methylation of contiguous CpG sites in methylation array may be potential diagnostic markers of rectal cancer.
Collapse
Affiliation(s)
- Jiufeng Wei
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China.,Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China
| | - Guodong Li
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China.,Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China
| | - Jinning Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China.,Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China
| | - Yuhui Zhou
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China.,Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China
| | - Shuwei Dang
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China.,Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China
| | - Hongsheng Chen
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China.,Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150001, P.R. China
| | - Ming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China.,Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, P.R. China
| |
Collapse
|
13
|
Wang Q, Pan M, Wei J, Liu X, Wang F. Evaluation of DNA Methyltransferase Activity and Inhibition via Isothermal Enzyme-Free Concatenated Hybridization Chain Reaction. ACS Sens 2017; 2:932-939. [PMID: 28750535 DOI: 10.1021/acssensors.7b00168] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Methyltransferase (MTase)-catalyzed DNA methylation plays a vital role in the biological epigenetic processes of key diseases and has attracted increasing attention, making the amplified detection of MTase activity of great significance in clinical disease diagnosis and treatment. Herein, we developed an isothermal, enzyme-free, and autonomous strategy for analyzing MTase activity based on concatenated hybridization chain reaction (C-HCR)-mediated Förster resonance energy transfer (FRET). In a typical C-HCR procedure without MTase (Dam), Y-shaped initiator DNA activates upstream HCR-1 to assemble a double-stranded DNA (dsDNA) copolymeric nanowire consisting of multiple tandem DNA trigger units that motivate downstream HCR-2 to successively bring a fluorophore donor/acceptor (FAM/TAMRA) pair into close proximity, leading to the generation of an amplified FRET readout signal. The target Dam MTase and auxiliary DpnI endonuclease can sequentially and specifically recognize/methylate and cleave the Y-shaped initiator oligonucleotide, respectively, and thus prohibit the C-HCR process and FRET signal generation, resulting in the construction of a signal-on sensing platform for MTase assay. Our proposed isothermal enzyme-free C-HCR amplification approach was further utilized for screening MTase inhibitors. Furthermore, the proposed C-HCR approach can be easily adapted for probing other different MTases and for screening the corresponding inhibitors just by changing the recognition sequence of Y-shaped initiator DNA through a "plug-and-play" format. It provides a versatile and robust tool for highly sensitive detection of various biotransformations and thus holds great promise in clinical assessment and diagnosis.
Collapse
Affiliation(s)
- Qing Wang
- Key Laboratory of Analytical
Chemistry for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Min Pan
- Key Laboratory of Analytical
Chemistry for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Jie Wei
- Key Laboratory of Analytical
Chemistry for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Xiaoqing Liu
- Key Laboratory of Analytical
Chemistry for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Fuan Wang
- Key Laboratory of Analytical
Chemistry for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
14
|
Papoudou-Bai A, Hatzimichael E, Barbouti A, Kanavaros P. Expression patterns of the activator protein-1 (AP-1) family members in lymphoid neoplasms. Clin Exp Med 2016; 17:291-304. [PMID: 27600282 DOI: 10.1007/s10238-016-0436-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 08/23/2016] [Indexed: 12/22/2022]
Abstract
The activator protein-1 (AP-1) is a dimeric transcription factor composed of proteins belonging to the Jun (c-Jun, JunB and JunD), Fos (c-Fos, FosB, Fra1 and Fra2) and activating transcription factor protein families. AP-1 is involved in various cellular events including differentiation, proliferation, survival and apoptosis. Deregulated expression of AP-1 transcription factors is implicated in the pathogenesis of various lymphomas such as classical Hodgkin lymphomas, anaplastic large cell lymphomas, diffuse large B cell lymphomas and adult T cell leukemia/lymphoma. The main purpose of this review is the analysis of the expression patterns of AP-1 transcription factors in order to gain insight into the histophysiology of lymphoid tissues and the pathology of lymphoid malignancies.
Collapse
Affiliation(s)
| | | | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
15
|
Singh R, Lillard JW, Singh S. Epigenetic Changes and Potential Targets in Pancreatic Cancer. EPIGENETIC ADVANCEMENTS IN CANCER 2016:27-63. [DOI: 10.1007/978-3-319-24951-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Atsaves V, Zhang R, Ruder D, Pan Y, Leventaki V, Rassidakis GZ, Claret FX. Constitutive control of AKT1 gene expression by JUNB/CJUN in ALK+ anaplastic large-cell lymphoma: a novel crosstalk mechanism. Leukemia 2015; 29:2162-2172. [PMID: 25987255 PMCID: PMC4633353 DOI: 10.1038/leu.2015.127] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 01/10/2023]
Abstract
Anaplastic lymphoma kinase-positive (ALK+) anaplastic large-cell lymphoma (ALCL) is an aggressive T-cell non-Hodgkin lymphoma characterized by the t(2;5), resulting in the overexpression of nucleophosmin (NPM)-ALK, which is known to activate the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, resulting in cell cycle and apoptosis deregulation. ALK+ ALCL is also characterized by strong activator protein-1 (AP-1) activity and overexpression of two AP-1 transcription factors, CJUN and JUNB. Here, we hypothesized that a biologic link between AP-1 and AKT kinase may exist, thus contributing to ALCL oncogenesis. We show that JUNB and CJUN bind directly to the AKT1 promoter, inducing AKT1 transcription in ALK+ ALCL. Knockdown of JUNB and CJUN in ALK+ ALCL cell lines downregulated AKT1 mRNA and promoter activity and was associated with lower AKT1 protein expression and activation. We provide evidence that this is a transcriptional control mechanism shared by other cell types even though it may operate in a way that is cell context-specific. In addition, STAT3 (signal transducer and activator of transcription 3)-induced control of AKT1 transcription was functional in ALK+ ALCL and blocking of STAT3 and AP-1 signaling synergistically affected cell proliferation and colony formation. Our findings uncover a novel transcriptional crosstalk mechanism that links AP-1 and AKT kinase, which coordinate uncontrolled cell proliferation and survival in ALK+ ALCL.
Collapse
Affiliation(s)
- Vassilios Atsaves
- Department of Systems Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- G.P. Livanos and M. Simou Laboratories, First Department of Critical Care Medicine & Pulmonary Services, Medical School of Athens University, "Evangelismos" Hospital, Athens, Greece
| | - Ronghua Zhang
- Department of Systems Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Dennis Ruder
- Department of Systems Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yunbao Pan
- Department of Systems Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Wuxi Medical School and Affiliated Hospital, Jiangnan University, Wuxi 214122, China
| | - Vasiliki Leventaki
- Department of Pathology, Saint Jude Children’s Hospital, Memphis, TN, USA
| | - George Z. Rassidakis
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Department of Pathology and Cytology, Karolinska University Hospital & Karolinska Institute, Stockholm, Sweden
| | - Francois X. Claret
- Department of Systems Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Experimental Therapeutics Academic Program and Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
17
|
Schirrmann T, Steinwand M, Wezler X, Ten Haaf A, Tur MK, Barth S. CD30 as a therapeutic target for lymphoma. BioDrugs 2015; 28:181-209. [PMID: 24043362 DOI: 10.1007/s40259-013-0068-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hodgkin's lymphoma (HL) and ALK(+) anaplastic large-cell lymphoma (ALCL) have become highly curable due to the success of modern regimens of chemotherapy and radiotherapy. However, up to one-third of the patients experience relapse or do not respond to first-line therapy, and half of them relapse again after secondary therapy with limited options for further treatment. In the last 15 years, monoclonal antibodies (mAbs) directed to surface receptors became a new and valuable therapeutic option in many hematologic malignancies. Due to its restricted expression on normal activated lymphocytes and its high expression on malignant cells, CD30 represents an attractive target molecule for HL and ALCL therapy. However, unconjugated CD30 mAbs have demonstrated a lack of objective clinical responses in patients with recurrent HL. CD30 exhibits complex signaling pathways, and binding of its natural ligand or anti-CD30 mAbs can induce apoptosis but may also promote proliferation and activation depending on the cellular context. Moreover, CD30 rapidly internalizes after crosslinking, which counteracts efficient recruitment of immunologic effectors but also provides the opportunity to transfer cytotoxic payloads coupled to CD30-specific mAbs into the tumor cells. Several tumor targeting approaches have been studied, including radio-immunoconjugates, immunotoxins, immunoRNases, immunokinases, and antibody drug conjugates (ADCs). In 2011, the ADC brentuximab-vedotin, consisting of the CD30-specific chimeric mAb cAC10 and the potent tubulin toxin monomethyl auristatin E, gained regulatory approval as a well tolerated and highly active drug in patients with refractory and relapsed HL and ALCL. SGN-35 is on the way to being incorporated in the standard management of CD30(+) lymphoma with significant therapeutic impact. This review gives a critical overview about anti-CD30 therapies with unconjugated, engineered, and conjugated mAbs and the therapeutic challenges of treatment of CD30(+) lymphoma.
Collapse
Affiliation(s)
- Thomas Schirrmann
- Department of Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany,
| | | | | | | | | | | |
Collapse
|
18
|
Boddicker RL, Kip NS, Xing X, Zeng Y, Yang ZZ, Lee JH, Almada LL, Elsawa SF, Knudson RA, Law ME, Ketterling RP, Cunningham JM, Wu Y, Maurer MJ, O'Byrne MM, Cerhan JR, Slager SL, Link BK, Porcher JC, Grote DM, Jelinek DF, Dogan A, Ansell SM, Fernandez-Zapico ME, Feldman AL. The oncogenic transcription factor IRF4 is regulated by a novel CD30/NF-κB positive feedback loop in peripheral T-cell lymphoma. Blood 2015; 125:3118-27. [PMID: 25833963 PMCID: PMC4432006 DOI: 10.1182/blood-2014-05-578575] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 03/21/2015] [Indexed: 12/15/2022] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) are generally aggressive non-Hodgkin lymphomas with poor overall survival rates following standard therapy. One-third of PTCLs express interferon regulatory factor-4 (IRF4), a tightly regulated transcription factor involved in lymphocyte growth and differentiation. IRF4 drives tumor growth in several lymphoid malignancies and has been proposed as a candidate therapeutic target. Because direct IRF4 inhibitors are not clinically available, we sought to characterize the mechanism by which IRF4 expression is regulated in PTCLs. We demonstrated that IRF4 is constitutively expressed in PTCL cells and drives Myc expression and proliferation. Using an inhibitor screen, we identified nuclear factor κB (NF-κB) as a candidate regulator of IRF4 expression and cell proliferation. We then demonstrated that the NF-κB subunits p52 and RelB were transcriptional activators of IRF4. Further analysis showed that activation of CD30 promotes p52 and RelB activity and subsequent IRF4 expression. Finally, we showed that IRF4 transcriptionally regulates CD30 expression. Taken together, these data demonstrate a novel positive feedback loop involving CD30, NF-κB, and IRF4; further evidence for this mechanism was demonstrated in human PTCL tissue samples. Accordingly, NF-κB inhibitors may represent a clinical means to disrupt this feedback loop in IRF4-positive PTCLs.
Collapse
MESH Headings
- Adult
- Aged
- Cell Line, Tumor
- Cell Proliferation
- DNA Copy Number Variations
- Female
- Gene Expression Regulation, Neoplastic
- Genes, myc
- Germ Cells/metabolism
- Humans
- Interferon Regulatory Factors/genetics
- Ki-1 Antigen/metabolism
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/metabolism
- Male
- Middle Aged
- Models, Biological
- NF-kappa B/metabolism
- Polymorphism, Genetic
- Transcription, Genetic
Collapse
Affiliation(s)
| | - N Sertac Kip
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Xiaoming Xing
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Pathology, Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Yu Zeng
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | | | - Jeong-Heon Lee
- Epigenomics Translational Program, Center for Individualized Medicine
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, and
| | - Sherine F Elsawa
- Schulze Center for Novel Therapeutics, Division of Oncology Research, and
| | - Ryan A Knudson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Mark E Law
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Rhett P Ketterling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Julie M Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Yanhong Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Matthew J Maurer
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Megan M O'Byrne
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - James R Cerhan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Susan L Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Brian K Link
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA; and
| | | | | | - Diane F Jelinek
- Division of Hematology, Department of Immunology, Mayo Clinic, Rochester, MN
| | - Ahmet Dogan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | | | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| |
Collapse
|
19
|
Papoudou-Bai A, Goussia A, Batistatou A, Stefanou D, Malamou-Mitsi V, Kanavaros P. The expression levels of JunB, JunD and p-c-Jun are positively correlated with tumor cell proliferation in diffuse large B-cell lymphomas. Leuk Lymphoma 2015; 57:143-50. [PMID: 25813203 DOI: 10.3109/10428194.2015.1034704] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We analyzed the expression of Jun family in relation to CD30 expression, cell proliferation and B-cell differentiation immunophenotypes [Germinal Center and non-Germinal Center] in diffuse large B-cell lymphomas (DLBCL). Expression and high expression of phosphorylated-c-Jun (p-c-Jun), JunB, JunD and CD30 (cut-off scores 20% and 50%, respectively) was found in 18/103, 49/103, 72/101 and 26/102 cases, respectively, and in 6/103, 27/103, 60/101 and 21/102 cases, respectively. The following significant positive correlations were observed: (a) JunB with cyclin A (p = 0.046), cyclin B1 (p = 0.033), cyclin E (p = 0.003), MUM-1 (p = 0.002) and CD30 (p < 0.001), (b) JunD with Ki67 (p = 0.002) and cyclin E (p = 0.014), (c) p-c-Jun with CD30 (p = 0.015), and (d) high p-c-Jun with cyclin A (p = 0.034). The positive correlation between expression of JunB, JunD and p-c-Jun and tumor cell proliferation in DLBCL, suggests that increased JunB, JunD and p-c-Jun expression may be involved in the pathogenesis of DLBCL by increasing tumor cell proliferation.
Collapse
Affiliation(s)
| | - Anna Goussia
- a Department of Pathology , Medical Faculty , University of Ioannina, Ioannina , Greece
| | - Anna Batistatou
- a Department of Pathology , Medical Faculty , University of Ioannina, Ioannina , Greece
| | - Dimitrios Stefanou
- a Department of Pathology , Medical Faculty , University of Ioannina, Ioannina , Greece
| | | | - Panagiotis Kanavaros
- b Department of Anatomy-Histology-Embryology , Medical Faculty , University of Ioannina, Ioannina , Greece
| |
Collapse
|
20
|
Jeon SH, Jeon EH, Lee JY, Kim YS, Yoon HJ, Hong SP, Lee JH. The potential of interleukin 12 receptor beta 2 (IL12RB2) and tumor necrosis factor receptor superfamily member 8 (TNFRSF8) gene as diagnostic biomarkers of oral lichen planus (OLP). Acta Odontol Scand 2015; 73:588-94. [PMID: 25915578 DOI: 10.3109/00016357.2014.967719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE This study evaluated the potential of interleukin 12 receptor beta 2 and tumor necrosis factor receptor superfamily member 8 as diagnostic biomarkers of oral lichen planus (OLP). MATERIALS AND METHODS The mRNA expression of IL12RB2 and TNFRSF8 in FFPE OLP samples (OLP group, n = 38) were investigated with quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) analysis and compared to those of chronic non-specific mucositis (Non-OLP group, n = 25) and normal mucosa (Normal group, n = 18). Predictive modeling of the expression of IL12RB2 and TNFRSF8 was constructed using support vector machine (SVM), random forest (RF), linear discriminant analysis (LDA), neural network (NN) and naive Bayes (NB) methods. RESULTS Normalized expression of IL12RB2 in the OLP group (3.78 ± 1.67) was significantly higher than the Normal group (1.97 ± 1.12), but lower than the Non-OLP group (6.86 ± 1.67). TNFRSF8 gene expression in the OLP group (7.46 ± 1.51) was significantly higher than the Normal group (2.90 ± 1.61), but no significant difference was found between the OLP and Non-OLP groups. The ratio of IL12RB2/TNFRSF8 in the OLP group (0.52 ± 0.23) was significantly lower than the Normal group (0.74 ± 0.39) and the Non-OLP group (1.07 ± 0.38). In the predictive modeling, the area under receiver operating characteristic (ROC) curves (AUC) ranged from 0.83-0.92 and their accuracy was higher than 0.75 in all methods. CONCLUSIONS The IL12RB2/TNFRSF8 ratio can be a useful diagnostic tool for OLP.
Collapse
Affiliation(s)
- Seung-Ho Jeon
- Department of Oral and Maxillofacial Surgery, School of Dentistry
| | | | | | | | | | | | | |
Collapse
|
21
|
Li K, Lian L, Yang N, Qu L. Temporal expression and DNA hypomethylation profile of CD30 in Marek's disease virus-infected chicken spleens. Poult Sci 2015; 94:1165-9. [PMID: 25840961 DOI: 10.3382/ps/pev100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2015] [Indexed: 01/28/2023] Open
Abstract
Marek's disease (MD) is a viral neoplastic disease of chickens caused by Marek's disease virus (MDV), which is serious threat to worldwide poultry industry. Our previous studies showed that the CD30 gene was hypomethylated in MD lymphoma. In this study, we further analyzed differential expression patterns and methylation levels of the CD30 gene between MDV-infected and noninfected spleens at 4, 7, 14, 21, and 28 d postinfection (dpi). The results showed that the expression of CD30 in MDV-infected spleens was significantly lower than that in noninfected spleens at 4 dpi. The expression of CD30 did not present significant difference between MDV-infected and noninfected spleens at 7 and 14 dpi. However, an increased expression of CD30 was presented in MDV-infected spleens at both 21 and 28 dpi. Simultaneously, CD30 showed a lower DNA methylation level in MDV-infected spleens at 14, 21, and 28 dpi. The results indicated that CD30 gene was involved in the whole process of MD tumorigenesis and upregulated expression of CD30 in MDV-infected spleens might be attributed to the hypomethylation of promoter of CD30 gene.
Collapse
Affiliation(s)
- Kaiyang Li
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ling Lian
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ning Yang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lujiang Qu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
22
|
Gill K, Ariyan C, Wang X, Brady MS, Pulitzer M. CD30-positive lymphoproliferative disorders arising after regional therapy for recurrent melanoma: A report of two cases and analysis of CD30 expression. J Surg Oncol 2014; 110:258-64. [DOI: 10.1002/jso.23636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 04/05/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Kamraan Gill
- Department of Pathology; Memorial Sloan Kettering Cancer Center; New York New York
| | - Charlotte Ariyan
- Department of Surgery; Memorial Sloan Kettering Cancer Center; New York New York
| | - Xuan Wang
- Department of Surgery; Memorial Sloan Kettering Cancer Center; New York New York
| | - Mary Sue Brady
- Department of Surgery; Memorial Sloan Kettering Cancer Center; New York New York
| | - Melissa Pulitzer
- Department of Pathology; Memorial Sloan Kettering Cancer Center; New York New York
| |
Collapse
|
23
|
Banerjee N, Paul S, Sau TJ, Das JK, Bandyopadhyay A, Banerjee S, Giri AK. Epigenetic Modifications of DAPK and p16 Genes Contribute to Arsenic-Induced Skin Lesions and Nondermatological Health Effects. Toxicol Sci 2013; 135:300-8. [DOI: 10.1093/toxsci/kft163] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
24
|
Zhang W, Qu L, Xu G, Lian L, Zheng J, Yang N. Hypomethylation upregulates the expression of CD30 in lymphoma induced by Marek's disease virus. Poult Sci 2012; 91:1610-8. [PMID: 22700506 DOI: 10.3382/ps.2011-02086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Epigenetic modification is widely known to be involved in embryo development, aging, tumorigenesis, and many complex diseases. Both hypermethylation of CpG islands at the gene promoters and global hypomethylation are involved in the initiation and progression of carcinogenesis. However, only a small portion of hypomethylation occurs at gene promoters and leads to the overexpression of certain oncogenes. To determine whether DNA methylation plays a role in tumorigenesis of Marek's disease, we selected one putative oncogene and 8 tumor suppressor genes from the gene expression profile for the analysis of DNA methylation variation. Four normal spleen tissues and 4 Marek's disease virus-infected tumor spleen tissues were collected, and the methylation level of the promoter region of each gene was analyzed using MassARRAY. As a result, the promoter region of CD30 was hypomethylated and displayed a significantly higher expression in Marek's disease virus-infected tumor spleen tissues compared with normal ones (P < 0.05). In neoplastic cells, CD30 was known to promote the survival and proliferation of T-cell lymphomas. This result suggests that activation of CD30 is possibly associated with the tumorigenesis of Marek's disease.
Collapse
Affiliation(s)
- W Zhang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, China
| | | | | | | | | | | |
Collapse
|
25
|
Lipoprotein lipase expression in unmutated CLL patients is the consequence of a demethylation process induced by the microenvironment. Leukemia 2012; 27:721-5. [PMID: 22828442 DOI: 10.1038/leu.2012.212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
26
|
Greve B, Kelsch R, Spaniol K, Eich HT, Götte M. Flow cytometry in cancer stem cell analysis and separation. Cytometry A 2012; 81:284-93. [PMID: 22311742 DOI: 10.1002/cyto.a.22022] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 12/21/2011] [Accepted: 01/07/2012] [Indexed: 12/19/2022]
Abstract
In recent years, a special type of cancer cell--the cancer stem cell (CSC)--has been identified and characterized for different tumors. CSCs may be responsible for the recurrence of a tumor following a primarily successful therapy and are thought to bear a high metastatic potential. For the development of efficient treatment strategies, the establishment of reliable methods for the identification and effective isolation of CSCs is imperative. Similar to their stem cell counterparts in bone marrow or small intestine, different cluster of differentiation surface antigens have been characterized, thus enabling researchers to identify them within the tumor bulk and to determine their degree of differentiation. In addition, functional properties characteristic of stem cells can be measured. Side population analysis is based on the stem cell-specific activity of certain ATP-binding cassette transporter proteins, which are able to transport fluorescent dyes out of the cells. Furthermore, the stem cell-specific presence of aldehyde dehydrogenase isoform 1 can be used for CSC labeling. However, the flow cytometric analysis of these CSC functional features requires specific technical adjustments. This review focuses on the principles and strategies of the flow cytometric analysis of CSCs and provides an overview of current protocols as well as technical requirements and pitfalls. A special focus is set on side population analysis and analysis of ALDH activity. Flow cytometry-based sorting principles and future flow cytometric applications for CSC analysis are also discussed.
Collapse
Affiliation(s)
- Burkhard Greve
- Department of Radiotherapy, University Hospital, 48149 Münster, Germany.
| | | | | | | | | |
Collapse
|
27
|
Watanabe M, Itoh K, Togano T, Kadin ME, Watanabe T, Higashihara M, Horie R. Ets-1 activates overexpression of JunB and CD30 in Hodgkin's lymphoma and anaplastic large-cell lymphoma. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:831-8. [PMID: 22107829 DOI: 10.1016/j.ajpath.2011.10.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 09/23/2011] [Accepted: 10/13/2011] [Indexed: 01/20/2023]
Abstract
Overexpression of CD30 and JunB is a hallmark of tumor cells in Hodgkin's lymphoma (HL) and anaplastic large-cell lymphoma (ALCL). We reported that CD30-extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein kinase (MAPK) signaling induces JunB, which maintains constitutive activation of the CD30 promoter. Herein, we localize a cis-acting enhancer in the JunB promoter that is regulated by Ets-1. We show that E26 transformation-specific-1 (Ets-1) (-146 to -137) enhances JunB promoter activation in a manner that is dependent on CD30 or the nucleophosmin-anaplastic lymphoma kinase (NPM-ALK)-ERK1/2 MAPK pathway. Ets-1 knockdown reduces the expression of both JunB and CD30, and CD30 knockdown significantly reduces JunB expression in HL and ALCL cell lines. NPM-ALK knockdown also reduces JunB expression in ALCL cell lines expressing NPM-ALK. Collectively, these results indicate that CD30 and NPM-ALK cooperate to activate the ERK1/2 MAPK-Ets-1 pathway. Ets-1, constitutively activated by ERK1/2-MAPK, plays a central role in the overexpression of JunB and CD30, which are both involved in the pathogenesis of HL and ALCL.
Collapse
Affiliation(s)
- Mariko Watanabe
- Department of Hematology, School of Medicine, Kitasato University, Kanagawa, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Chung TL, Turner JP, Thaker NY, Kolle G, Cooper-White JJ, Grimmond SM, Pera MF, Wolvetang EJ. Ascorbate promotes epigenetic activation of CD30 in human embryonic stem cells. Stem Cells 2011; 28:1782-93. [PMID: 20715184 DOI: 10.1002/stem.500] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells have the ability to adapt to various culture conditions. Phenotypic and epigenetic changes brought about by the culture conditions can, however, have significant impacts on their use in research and in clinical applications. Here, we show that diploid hESCs start to express CD30, a biomarker for malignant cells in Hodgkin's disease and embryonal carcinoma cells, when cultured in knockout serum replacement (KOSR)-based medium, but not in fetal calf serum containing medium. We identify the commonly used medium additive, ascorbate, as the sole medium component in KOSR responsible for CD30 induction. Our data show that this epigenetic activation of CD30 expression in hESCs by ascorbate occurs through a dramatic loss of DNA methylation of a CpG island in the CD30 promoter. Analysis of the phenotype and transcriptome of hESCs that overexpress the CD30 signaling domain reveals that CD30 signaling leads to inhibition of apoptosis, enhanced single-cell growth, and transcriptome changes that are associated with cell signaling, lipid metabolism, and tissue development. Collectively, our data show that hESC culture media that contain ascorbate trigger CD30 expression through an epigenetic mechanism and that this provides a survival advantage and transcriptome changes that may help adapt hESCs to in vitro culture conditions.
Collapse
Affiliation(s)
- Tung-Liang Chung
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Conerly M, Grady WM. Insights into the role of DNA methylation in disease through the use of mouse models. Dis Model Mech 2010; 3:290-7. [PMID: 20427558 DOI: 10.1242/dmm.004812] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epigenetics was originally defined as the interaction of genes with their environment that brings the phenotype into being. It now refers to the study of heritable changes in gene expression that occur without a change in DNA sequence. To date, the best understood epigenetic mechanisms are CpG DNA methylation and histone modifications. DNA methylation in particular has been the subject of intense interest because of its recently recognized role in disease, as well as in the development and normal function of organisms. Much of the focus of disease-related research has been on cancer because of the recognition that epigenetic alterations are common in cancer and probably cooperate with genetic alterations to drive cancer formation. Our understanding of epigenetic mechanisms in controlling gene expression has resulted from the study of cell line systems and simple model systems, such as Arabidopsis thaliana. We are now moving into an era of more complex model systems, such as transgenic and knockout mouse models, which will lead to further insight into epigenetics in development and human disease. The current models have revealed complex, tissue-specific effects of epigenetic mechanisms and have further informed our understanding of the role of DNA methylation and histone modifications on disease and development. The current state of these models is the subject of this Commentary.
Collapse
Affiliation(s)
- Melissa Conerly
- Basic Science Division, University of Washington Medical School, 1959 Northeast Pacific Street, Seattle, WA 98195, USA
| | | |
Collapse
|
30
|
Pogribny IP, Beland FA. DNA hypomethylation in the origin and pathogenesis of human diseases. Cell Mol Life Sci 2009; 66:2249-61. [PMID: 19326048 PMCID: PMC11115809 DOI: 10.1007/s00018-009-0015-5] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 02/25/2009] [Accepted: 03/06/2009] [Indexed: 12/15/2022]
Abstract
The pathogenesis of any given human disease is a complex multifactorial process characterized by many biologically significant and interdependent alterations. One of these changes, specific to a wide range of human pathologies, is DNA hypomethylation. DNA hypomethylation signifies one of the major DNA methylation states that refers to a relative decrease from the "normal" methylation level. It is clear that disease by itself can induce hypomethylation of DNA; however, a decrease in DNA methylation can also have an impact on the predisposition to pathological states and disease development. This review presents evidence suggesting the involvement of DNA hypomethylation in the pathogenesis of several major human pathologies, including cancer, atherosclerosis, Alzheimer's disease, and psychiatric disorders.
Collapse
Affiliation(s)
- Igor P Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA.
| | | |
Collapse
|
31
|
Ho DS, Rea AJ, Abraham LJ. Functional aspects of the CD30 gene in Hodgkin’s lymphoma and anaplastic large cell lymphoma. Oncol Rev 2009. [DOI: 10.1007/s12156-009-0012-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
32
|
Iacobuzio-Donahue CA. Epigenetic changes in cancer. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2009; 4:229-49. [PMID: 18840073 DOI: 10.1146/annurev.pathol.3.121806.151442] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cancer is as much an epigenetic disease as it is a genetic disease, and epigenetic alterations in cancer often serve as potent surrogates for genetic mutations. Normal epigenetic modifications of DNA encompass three types of changes: chromatin modifications, DNA methylation, and genomic imprinting, each of which is altered in cancer cells. This review addresses the various epigenetic modifications that are pervasive among human tumors and traces the history of cancer epigenetics from the first observations of altered global methylation content to the recently proposed epigenetic progenitor model, which provides a common unifying mechanism for cancer development.
Collapse
|
33
|
Butcher LM, Beck S. Future impact of integrated high-throughput methylome analyses on human health and disease. J Genet Genomics 2009; 35:391-401. [PMID: 18640619 DOI: 10.1016/s1673-8527(08)60057-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 06/08/2008] [Accepted: 06/09/2008] [Indexed: 10/21/2022]
Abstract
A spate of high-powered genome-wide association studies (GWAS) have recently identified numerous single-nucleotide polymorphisms (SNPs) robustly linked with complex disease. Despite interrogating the majority of common human variation, these SNPs only account for a small proportion of the phenotypic variance, which suggests genetic factors are acting in concert with non-genetic factors. Although environmental measures are logical covariants for genotype-phenotype investigations, another non-genetic intermediary exists: epigenetics. Epigenetics is the analysis of somatically-acquired and, in some cases, transgenerationally inherited epigenetic modifications that regulate gene expression, and offers to bridge the gap between genetics and environment to understand phenotype. The most widely studied epigenetic mark is DNA methylation. Aberrant methylation at gene promoters is strongly implicated in disease etiology, most notably cancer. This review will highlight the importance of DNA methylation as an epigenetic regulator, outline techniques to characterize the DNA methylome and present the idea of reverse phenotyping, where multiple layers of analysis are integrated at the individual level to create personalized digital phenotypes and, at a phenotype level, to identify novel molecular signatures of disease.
Collapse
Affiliation(s)
- Lee M Butcher
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK
| | | |
Collapse
|
34
|
Zhu J, Yao X. Use of DNA methylation for cancer detection: Promises and challenges. Int J Biochem Cell Biol 2009; 41:147-54. [DOI: 10.1016/j.biocel.2008.09.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 09/08/2008] [Accepted: 09/10/2008] [Indexed: 01/12/2023]
|