1
|
Saloni, Sachan M, Rahul, Verma RS, Patel GK. SOXs: Master architects of development and versatile emulators of oncogenesis. Biochim Biophys Acta Rev Cancer 2025; 1880:189295. [PMID: 40058508 DOI: 10.1016/j.bbcan.2025.189295] [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: 10/02/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Transcription factors regulate a variety of events and maintain cellular homeostasis. Several transcription factors involved in embryonic development, has been shown to be closely associated with carcinogenesis when deregulated. Sry-like high mobility group box (SOX) proteins are potential transcription factors which are evolutionarily conserved. They regulate downstream genes to determine cell fate, via various signaling pathways and cellular processes essential for tissue and organ development. Dysregulation of SOXs has been reported to promote or suppress tumorigenesis by modulating cellular reprogramming, growth, proliferation, angiogenesis, metastasis, apoptosis, immune modulation, lineage plasticity, maintenance of the stem cell pool, therapy resistance and cancer relapse. This review provides a crucial understanding of the molecular mechanism by which SOXs play multifaceted roles in embryonic development and carcinogenesis. It also highlights their potential in advancing therapeutic strategies aimed at targeting SOXs and their downstream effectors in various malignancies.
Collapse
Affiliation(s)
- Saloni
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Rahul
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rama Shanker Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| |
Collapse
|
2
|
Zhou Y, Chen X, Zu X. ZBTB7A as a therapeutic target for cancer. Biochem Biophys Res Commun 2024; 736:150888. [PMID: 39490153 DOI: 10.1016/j.bbrc.2024.150888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
ZBTB7A, alternatively referred to Pokemon, FBI-1, LRF, and OCZF, is classified as a member of POK/ZBTB protein family of transcriptional repressors. ZBTB7A binds to targeted DNA via C-terminal zinc fingers and recruits co-compression complexes through N-terminal BTB ⁄ POZ domain to impede transcription. ZBTB7A regulates a range of fundamental biological processes such as cell proliferation, differentiation and apoptosis, B- and T-lymphocyte fate determination and thymic insulin expression and self-tolerance. Accumulating evidence has demonstrated an important role of ZBTB7A in the initiation and advancement of tumors, thus making ZBTB7A emerge as an appealing target. This review examines the functions and regulatory mechanisms of ZBTB7A in a range of common solid tumors, including hepatocellular carcinoma, breast cancer, prostate cancer and lung cancer, as well as hematological malignancies. Notably, the review concludes with a summary of the recent applications of targeting ZBTB7A in clinical treatments through gene silencing, immunotherapy and chemotherapeutic approaches to halt or slow tumor progression. We focus on the functional role and regulatory mechanisms of ZBTB7A in cancer with the goal of providing new insights for the development of more effective cancer therapeutic strategies.
Collapse
Affiliation(s)
- Ying Zhou
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xisha Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, China.
| |
Collapse
|
3
|
Kambe G, Kobayashi M, Ishikita H, Koyasu S, Hammond EM, Harada H. ZBTB7A forms a heterodimer with ZBTB2 and inhibits ZBTB2 homodimerization required for full activation of HIF-1. Biochem Biophys Res Commun 2024; 733:150604. [PMID: 39197198 DOI: 10.1016/j.bbrc.2024.150604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Hypoxia-inducible factor 1 (HIF-1), recognized as a master transcription factor for adaptation to hypoxia, is associated with malignant characteristics and therapy resistance in cancers. It has become clear that cofactors such as ZBTB2 are critical for the full activation of HIF-1; however, the mechanisms downregulating the ZBTB2-HIF-1 axis remain poorly understood. In this study, we identified ZBTB7A as a negative regulator of ZBTB2 by analyzing protein sequences and structures. We found that ZBTB7A forms a heterodimer with ZBTB2, inhibits ZBTB2 homodimerization necessary for the full expression of ZBTB2-HIF-1 downstream genes, and ultimately delays the proliferation of cancer cells under hypoxic conditions. The Cancer Genome Atlas (TCGA) analyses revealed that overall survival is better in patients with high ZBTB7A expression in their tumor tissues. These findings highlight the potential of targeting the ZBTB7A-ZBTB2 interaction as a novel therapeutic strategy to inhibit HIF-1 activity and improve treatment outcomes in hypoxia-related cancers.
Collapse
Affiliation(s)
- Gouki Kambe
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hiroshi Ishikita
- Department of Applied Chemistry, The University of Tokyo, Tokyo, Japan; Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Sho Koyasu
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
4
|
Bergez-Hernández F, Irigoyen-Arredondo M, Martínez-Camberos A. A systematic review of mechanisms of PTEN gene down-regulation mediated by miRNA in prostate cancer. Heliyon 2024; 10:e34950. [PMID: 39144981 PMCID: PMC11320309 DOI: 10.1016/j.heliyon.2024.e34950] [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: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
Background The Phosphatase and Tensin Homolog gene (PTEN) is pivotal in regulating diverse cellular processes, including growth, differentiation, proliferation, and cell survival, mainly by modulating the PI3K/AKT/mTOR pathway. Alterations in the expression of the PTEN gene have been associated with epigenetic mechanisms, particularly the regulation by small non-coding RNAs, such as miRNAs. Modifications in the expression levels of miRNAs that control PTEN have been shown to lead to its underexpression. This underexpression, in turn, impacts the PI3K/AKT/mTOR pathway, thereby influencing crucial mechanisms like proliferation and apoptosis, playing an important role in the initiation and progression of prostate cancer (PCa). Thus, we aimed to systematically reviewed available information concerning the regulation of PTEN mediated by miRNA in PCa. Methods Electronic databases were searched to identify studies assessing PTEN regulation via PCa miRNAs, the search included combination of the words microRNAs, PTEN and prostatic neoplasms. The quality assessment of the articles included was carried out using an adapted version of SYRCLE and CASP tool. Results We included 39 articles that measured the relative gene expression of miRNAs in PCa and their relationship with PTEN regulation. A total of 42 miRNAs were reported involved in the development and progression of PCa via PTEN dysregulation (34 miRNAs up-regulated and eight miRNAs down-regulated). Sixteen miRNAs were shown as the principal regulators for genetic interactions leading to carcinogenesis, being the miR-21 the most reported in PCa associated with PTEN down-regulation. We showed the silencing of PTEN could be promoted by a loop between miR-200b and DNMT1 or by direct targeting of PTEN by microRNAs, leading to the constitutive activation of PI3K/AKT/mTOR and interactions with intermediary genes support apoptosis inhibition, proliferation, invasion, and metastasis in PCa. Conclusion According to our review, dysregulation of PTEN mediated mainly by miR-21, -20a, -20b, -93, -106a, and -106b up-regulation has a central role in PCa development and could be potential biomarkers for diagnosis, prognostic, and therapeutic targets.
Collapse
Affiliation(s)
| | | | - Alejandra Martínez-Camberos
- Laboratorio de Biomedicina y Biología Molecular. Lic. en Ciencias Biomédicas, Universidad Autónoma de Occidente. Av del Mar 1200, Tellerías, 82100, Mazatlán, Sinaloa, Mexico
| |
Collapse
|
5
|
Cao F, Li Y, Peng T, Li Y, Yang L, Hu L, Zhang H, Wang J. PTEN in kidney diseases: a potential therapeutic target in preventing AKI-to-CKD transition. Front Med (Lausanne) 2024; 11:1428995. [PMID: 39165377 PMCID: PMC11333338 DOI: 10.3389/fmed.2024.1428995] [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: 05/07/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Renal fibrosis, a critical factor in the development of chronic kidney disease (CKD), is predominantly initiated by acute kidney injury (AKI) and subsequent maladaptive repair resulting from pharmacological or pathological stimuli. Phosphatase and tensin homolog (PTEN), also known as phosphatase and tensin-associated phosphatase, plays a pivotal role in regulating the physiological behavior of renal tubular epithelial cells, glomeruli, and renal interstitial cells, thereby preserving the homeostasis of renal structure and function. It significantly impacts cell proliferation, apoptosis, fibrosis, and mitochondrial energy metabolism during AKI-to-CKD transition. Despite gradual elucidation of PTEN's involvement in various kidney injuries, its specific role in AKI and maladaptive repair after injury remains unclear. This review endeavors to delineate the multifaceted role of PTEN in renal pathology during AKI and CKD progression along with its underlying mechanisms, emphasizing its influence on oxidative stress, autophagy, non-coding RNA-mediated recruitment and activation of immune cells as well as renal fibrosis. Furthermore, we summarize prospective therapeutic targeting strategies for AKI and CKD-treatment related diseases through modulation of PTEN.
Collapse
Affiliation(s)
- Fangfang Cao
- Division of Nephrology, Mianyang Central Hospital, Mianyang, China
| | - Yuanyuan Li
- Division of Science and Education, Mianyang Central Hospital, Mianyang, China
| | - Ting Peng
- Division of Nephrology, Mianyang Central Hospital, Mianyang, China
| | - Yuanmei Li
- Division of Nephrology, Mianyang Central Hospital, Mianyang, China
| | - Lihua Yang
- Division of Nephrology, Mianyang Central Hospital, Mianyang, China
| | - Lanping Hu
- Hemodialysis Center, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Han Zhang
- Hemodialysis Center, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Jiali Wang
- Division of Nephrology, Mianyang Central Hospital, Mianyang, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, China
| |
Collapse
|
6
|
Ma M, Zhu Y, Xiao C, Li R, Cao X, Kang R, Wang X, Li E. Novel insights into RB1 in prostate cancer lineage plasticity and drug resistance. TUMORI JOURNAL 2024; 110:252-263. [PMID: 38316605 DOI: 10.1177/03008916231225576] [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] [Indexed: 02/07/2024]
Abstract
Prostate cancer is the second most common malignancy among men in the world, posing a serious threat to men's health and lives. RB1 is the first human tumor suppressor gene to be described, and it is closely associated with the development, progression, and suppression of a variety of tumors. It was found that the loss of RB1 is an early event in prostate cancer development and is closely related to prostate cancer development, progression and treatment resistance. This paper reviews the current status of research on the relationship between RB1 and prostate cancer from three aspects: RB1 and prostate cell lineage plasticity; biological behavior; and therapeutic resistance. Providing a novel perspective for developing new therapeutic strategies for RB1-loss prostate cancer.
Collapse
Affiliation(s)
- Min Ma
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yazhi Zhu
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Changkai Xiao
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ruidong Li
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xingyu Cao
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ran Kang
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaolan Wang
- Department of Reproductive Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ermao Li
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Huang Y, Guan Y, Zhang X. METTL3-Mediated Maturation of miR-99a-5p Promotes Cell Migration and Invasion in Oral Squamous Cell Carcinoma by Targeting ZBTB7A. Mol Biotechnol 2024; 66:1942-1953. [PMID: 37498409 DOI: 10.1007/s12033-023-00815-x] [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: 01/06/2023] [Accepted: 06/29/2023] [Indexed: 07/28/2023]
Abstract
METTL3 is an important methyltransferase in N(6)-methyladenosine (m6A) modification. Recently, METTL3 mediates methylation of pri-microRNA (miRNA) to accelerate miRNA maturation, regulating tumor development. This study explored whether METTL3 mediated miR-99a-5p to influence oral squamous cell carcinoma (OSCC) cell metastasis. MiR-99a-5p, ZBTB7A, and MATTL3 expression was measured using quantitative real-time PCR. Biological behaviors were assessed using cell counting kit-8, flow cytometry, Transwell assay, as well as western blot. Luciferase reporter assay evaluated the interaction between miR-99a-5p and ZBTB7A. METTL3-regulated pri-miR-99a-5p processing was determined by RNA binding protein immunoprecipitation (RIP) and methylated RNA immunoprecipitation (MeRIP) assays. The consequences clarified that miR-99a-5p was upregulated in OSCC cells. Downregulation of miR-99a-5p suppressed cellular viability, migration, invasion, and epithelial-mesenchymal transition (EMT), and induced apoptosis. ZBTB7A acted as a miR-99a-5p target and reversed the effects on cellular behaviors induced by miR-99a-5p inhibitor. m6A content and METTL3 expression were increased in OSCC cells. METTL3 promoted the m6A modification of pri-miR-99a-5p and thereby facilitated miR-99a-5p processing. Moreover, knockdown of METTL3 inhibited OSCC metastasis by downregulating miR-99a-5p. Taken together, METTL3 promoted miR-99a-5p maturation in an m6A-dependent manner, which further targets ZBTB7A to accelerate the progression of OSCC. These findings suggest potential targets for OSCC therapy.
Collapse
Affiliation(s)
- Yuhua Huang
- Department of stomatology, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine, 6/F, East Zone, No. 111, Dade Road, Yuexiu District, Guangzhou, 510000, Guangdong, China
| | - Yun Guan
- Department of stomatology, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine, 6/F, East Zone, No. 111, Dade Road, Yuexiu District, Guangzhou, 510000, Guangdong, China
| | - Xing Zhang
- Department of stomatology, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine, 6/F, East Zone, No. 111, Dade Road, Yuexiu District, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
8
|
Zanabria D, Galvez-Nino M, Araujo JM, Alfaro A, Fajardo W, Saravia L, Quispe L, Velazque G, Carbajal J, López MJ, Jimenez S, Montenegro P, Zevallos A, Clavo MDLA, Medina-Pérez P, Cornejo M, Requena M, Aguilar A, Pinto JA. Socioeconomic disparities and the genomic landscape of gastric cancer. Sci Rep 2024; 14:15070. [PMID: 38956258 PMCID: PMC11219810 DOI: 10.1038/s41598-024-65912-6] [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: 02/09/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
The genomic characteristics of Peruvian patients with gastric adenocarcinoma from diverse socioeconomic backgrounds were examined in consideration of the possibility that patients from different socioeconomic backgrounds may be exposed to different risk factors. We conducted a prospective pilot study in two Peruvian cities (Lima and Ica). This study enrolled 15 patients from low socioeconomic status (LSES) and 15 patients from medium/high socioeconomic status (MHSES). The genomic profiling of gastric adenocarcinoma samples was done through the FoundationOne CDx platform. We compared the genomic characteristics and the need for targeted therapy and immunotherapy between LSES and MHSES. The genes with higher rates of alterations were TP53 (73.3% vs. 50.0%, P = 0.2635); CDH1 (26.7% vs. 28.6%, P = 1); CDKN2A (20.0% vs. 28.6%, P = 1); KRAS (33.3% vs. 7.1%, P = 0.1686); ARID1A (20.0% vs. 14.3%, P = 1); MLL2 (13.3% vs. 21.4%, P = 1) and SOX9 (33.3% vs. 0.0%, P = 0.0421) in LSES versus HMSES, respectively. There was no significant difference in tumor mutational burden (P = 0.377) or microsatellite status (P = 1). The LSES group had a higher need for targeted therapy or immunotherapy according to gene involvement and alterations. A significant genomic difference exists among patients with gastric adenocarcinoma of different socioeconomic status, which may result in a different need for targeted therapy and immunotherapy.
Collapse
Affiliation(s)
| | | | - Jhajaira M Araujo
- Centro de Investigación Básica y Traslacional, Auna Ideas, Av. Guardia Civil 571, San Borja, 15036, Lima, Peru
| | - Alejandro Alfaro
- Servicio de Anatomía Patológica, Hospital Nacional Dos de Mayo, Lima, Peru
| | - Williams Fajardo
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima, Peru
| | - Luis Saravia
- Servicio de Emergencia, Hospital Regional de Ica, Ica, Peru
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Filial Ica, Ica, Peru
| | - Lidia Quispe
- Departamento de Patología, Hospital Regional de Ica, Ica, Peru
| | - Gina Velazque
- Servicio de Gastroenterología, Hospital Regional de Ica, Ica, Peru
| | - Junior Carbajal
- Facultad de Ciencias Biológicas, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | - María J López
- Facultad de Ciencias Biológicas, Universidad Nacional San Luis Gonzaga, Ica, Peru
| | - Sergio Jimenez
- Facultad de Ciencias Naturales y Matematicas, Universidad Nacional Federico Villarreal, Lima, Peru
| | | | - Alejandra Zevallos
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima, Peru
| | | | - Paula Medina-Pérez
- Centro de Investigación Básica y Traslacional, Auna Ideas, Av. Guardia Civil 571, San Borja, 15036, Lima, Peru
| | - Melanie Cornejo
- Centro de Investigación Básica y Traslacional, Auna Ideas, Av. Guardia Civil 571, San Borja, 15036, Lima, Peru
| | - María Requena
- Centro de Investigación Básica y Traslacional, Auna Ideas, Av. Guardia Civil 571, San Borja, 15036, Lima, Peru
| | - Alfredo Aguilar
- Centro de Investigación Básica y Traslacional, Auna Ideas, Av. Guardia Civil 571, San Borja, 15036, Lima, Peru
| | - Joseph A Pinto
- Centro de Investigación Básica y Traslacional, Auna Ideas, Av. Guardia Civil 571, San Borja, 15036, Lima, Peru.
| |
Collapse
|
9
|
Tao W, Yu Z, Han JDJ. Single-cell senescence identification reveals senescence heterogeneity, trajectory, and modulators. Cell Metab 2024; 36:1126-1143.e5. [PMID: 38604170 DOI: 10.1016/j.cmet.2024.03.009] [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: 08/18/2023] [Revised: 12/15/2023] [Accepted: 03/13/2024] [Indexed: 04/13/2024]
Abstract
Cellular senescence underlies many aging-related pathologies, but its heterogeneity poses challenges for studying and targeting senescent cells. We present here a machine learning program senescent cell identification (SenCID), which accurately identifies senescent cells in both bulk and single-cell transcriptome. Trained on 602 samples from 52 senescence transcriptome datasets spanning 30 cell types, SenCID identifies six major senescence identities (SIDs). Different SIDs exhibit different senescence baselines, stemness, gene functions, and responses to senolytics. SenCID enables the reconstruction of senescent trajectories under normal aging, chronic diseases, and COVID-19. Additionally, when applied to single-cell Perturb-seq data, SenCID helps reveal a hierarchy of senescence modulators. Overall, SenCID is an essential tool for precise single-cell analysis of cellular senescence, enabling targeted interventions against senescent cells.
Collapse
Affiliation(s)
- Wanyu Tao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, China
| | - Zhengqing Yu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| |
Collapse
|
10
|
Saeinasab M, Atlasi Y, M Matin M. Functional role of lncRNAs in gastrointestinal malignancies: the peculiar case of small nucleolar RNA host gene family. FEBS J 2024; 291:1353-1385. [PMID: 36282516 DOI: 10.1111/febs.16668] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in normal physiology and are often de-regulated in disease states such as cancer. Recently, a class of lncRNAs referred to as the small nucleolar RNA host gene (SNHG) family have emerged as important players in tumourigenesis. Here, we discuss new findings describing the role of SNHGs in gastrointestinal tumours and summarize the three main functions by which these lncRNAs promote carcinogenesis, namely: competing with endogenous RNAs, modulating protein function, and regulating epigenetic marking. Furthermore, we discuss how SNHGs participate in different hallmarks of cancer, and how this class of lncRNAs may serve as potential biomarkers in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, UK
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Iran
| |
Collapse
|
11
|
Jiang J, Wang Y, Sun M, Luo X, Zhang Z, Wang Y, Li S, Hu D, Zhang J, Wu Z, Chen X, Zhang B, Xu X, Wang S, Xu S, Huang W, Xia L. SOX on tumors, a comfort or a constraint? Cell Death Discov 2024; 10:67. [PMID: 38331879 PMCID: PMC10853543 DOI: 10.1038/s41420-024-01834-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
The sex-determining region Y (SRY)-related high-mobility group (HMG) box (SOX) family, composed of 20 transcription factors, is a conserved family with a highly homologous HMG domain. Due to their crucial role in determining cell fate, the dysregulation of SOX family members is closely associated with tumorigenesis, including tumor invasion, metastasis, proliferation, apoptosis, epithelial-mesenchymal transition, stemness and drug resistance. Despite considerable research to investigate the mechanisms and functions of the SOX family, confusion remains regarding aspects such as the role of the SOX family in tumor immune microenvironment (TIME) and contradictory impacts the SOX family exerts on tumors. This review summarizes the physiological function of the SOX family and their multiple roles in tumors, with a focus on the relationship between the SOX family and TIME, aiming to propose their potential role in cancer and promising methods for treatment.
Collapse
Affiliation(s)
- Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xiangyuan Luo
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zerui Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yijun Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Dian Hu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zhangfan Wu
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xiaoping Chen
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China
| | - Bixiang Zhang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Westlake university school of medicine, Hangzhou, 310006, China
| | - Shengjun Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases; Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
12
|
Zhang H, Ouyang C. BTB protein family and human breast cancer: signaling pathways and clinical progress. J Cancer Res Clin Oncol 2023; 149:16213-16229. [PMID: 37682360 DOI: 10.1007/s00432-023-05314-9] [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: 07/11/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Breast cancer is considered the number one killer of women both in China and abroad, and the leading cause of cancer death. It severely affects female health-related quality of life. Broad-complex, tramtrack, bric à brac (BTB) protein family was first discovered in drosophila as early as in 1993 by Godt D and peers, since then, more family members and their critical biological functions were uncovered. Moreover, researchers around the world have recently demonstrated that numerous signaling pathways connect BTB family members and human breast cancer. PURPOSE In this review, we critically discuss these findings regarding the essential mechanisms and functions of the BTB protein family in mediating the organic processes of human breast cancer. Meanwhile, we summarize the signaling pathways the BTB protein family participates in. And we address that BTB proteins regulate the growth, apoptosis, and other behaviors of breast cancer cells. We also point out the future directions for further studies in this field. METHODS The relevant online literatures have been reviewed for this article. CONCLUSION This review could offer an update on novel molecular targets for treating human breast cancer and new insights into BTB protein family research.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China.
| |
Collapse
|
13
|
Aquino AM, Alonso-Costa LG, Santos SAA, Rocha VA, Barbisan LF, Bedrat A, Justulin LA, Flaws JA, Lemos B, Scarano WR. Integrated transcriptome and proteome analysis indicates potential biomarkers of prostate cancer in offspring of pregnant rats exposed to a phthalate mixture during gestation and lactation. CHEMOSPHERE 2023; 341:140020. [PMID: 37690569 DOI: 10.1016/j.chemosphere.2023.140020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/09/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
As the second leading cause of death for cancer among men worldwide, prostate cancer (PCa) prevention and detection remain a critical challenge. One aspect of PCa research is the identification of common environmental agents that may increase the risk of initiation and progression of PCa. Endocrine disrupting chemicals (EDCs) are strong candidates for risk factors, partially because they alter essential pathways for prostate gland development and oncogenesis. Phthalates correspond to a set of commercially used plasticizers that humans are exposed to ubiquitously. Here, we show that maternal exposure to a phthalate mixture interferes with the expression profile of mRNA and proteins in the ventral prostate of offspring and increases the susceptibility to prostate adenocarcinomas in aged animals. The data highlight Ubxn11, Aldoc, Kif5c, Tubb4a, Tubb3, Tubb2, Rab6b and Rab3b as differentially expressed targets in young and adult offspring descendants (PND22 and PND120). These phthalate-induced targets were enriched for pathways such as: dysregulation in post-translational protein modification (PTPM), cell homeostasis, HSP90 chaperone activity, gap junctions, and kinases. In addition, the Kif5c, Tubb3, Tubb2b and Tubb4a targets were enriched for impairment in cell cycle and GTPase activity. Furthermore, these targets showed strong relationships with 12 transcriptional factors (TF), which regulate the phosphorylation of eight protein kinases. The correlation of TF-kinases is associated with alterations in immune system, RAS/ErbB/VEGF/estrogen/HIF-1 signaling pathways, cellular senescence, cell cycle, autophagy, and apoptosis. Downregulation of KIF5C, TUBB3 and RAB6B targets is associated with poor prognosis in patients diagnosed with adenocarcinoma. Collectively, this integrative investigation establishes the post-transcriptional mechanisms in the prostate that are modulated by maternal exposure to phthalate mixture during gestation and lactation.
Collapse
Affiliation(s)
- A M Aquino
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - L G Alonso-Costa
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - S A A Santos
- Cancer Signaling and Epigenetics, Fox Chase Cancer Center, Philadelphia, USA
| | - V A Rocha
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - L F Barbisan
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - A Bedrat
- Harvard T. H. Chan School of Public Health, Department of Environmental Health & Molecular and Integrative Physiological Sciences Program, Boston, Massachussets, USA
| | - L A Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - J A Flaws
- Department of Comparative Biosciences; University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - B Lemos
- Harvard T. H. Chan School of Public Health, Department of Environmental Health & Molecular and Integrative Physiological Sciences Program, Boston, Massachussets, USA
| | - W R Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| |
Collapse
|
14
|
Thomsen MK, Busk M. Pre-Clinical Models to Study Human Prostate Cancer. Cancers (Basel) 2023; 15:4212. [PMID: 37686488 PMCID: PMC10486646 DOI: 10.3390/cancers15174212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Prostate cancer is a common cancer among men and typically progresses slowly for several decades before becoming aggressive and spreading to other organs, leaving few treatment options. While large animals have been studied, the dog's prostate is anatomically similar to humans and has been used to study spontaneous prostate cancer. However, most research currently focuses on the mouse as a model organism due to the ability to genetically modify their prostatic tissues for molecular analysis. One milestone in this research was the identification of the prostate-specific promoter Probasin, which allowed for the prostate-specific expression of transgenes. This has led to the generation of mice with aggressive prostatic tumors through overexpression of the SV40 oncogene. The Probasin promoter is also used to drive Cre expression and has allowed researchers to generate prostate-specific loss-of-function studies. Another landmark moment in the process of modeling prostate cancer in mice was the orthoptic delivery of viral particles. This technology allows the selective overexpression of oncogenes from lentivirus or the use of CRISPR to generate complex loss-of-function studies. These genetically modified models are complemented by classical xenografts of human prostate tumor cells in immune-deficient mice. Overall, pre-clinical models have provided a portfolio of model systems to study and address complex mechanisms in prostate cancer for improved treatment options. This review will focus on the advances in each technique.
Collapse
Affiliation(s)
| | - Morten Busk
- Department of Experimental Clinical Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark;
- Danish Centre for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
15
|
Xu Y, Xu M, Li X, Weng X, Su Z, Zhang M, Tan J, Zeng H, Li X, Nie L, Gong J, Chen N, Chen X, Zhou Q. SOX9 and HMGB3 co-operatively transactivate NANOG and promote prostate cancer progression. Prostate 2023; 83:440-453. [PMID: 36541373 DOI: 10.1002/pros.24476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/03/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The homeodomain-containing transcription factor NANOG is overexpressed in prostate adenocarcinoma (PCa) and predicts poor prognosis. The SOX family transcription factor SOX9, as well as the transcription co-activator HMGB3 of the HMGB family, are also overexpressed and may play pivotal roles in PCa. However, it is unknown whether SOX9 and HMGB3 interact with each other, or if they regulate NANOG gene transcription. METHODS We identified potential SOX9 responsive elements in NANOG promoter, and investigated if SOX9 regulated NANOG transcription in co-operation with HMGB3 by experimental analysis of potential SOX9 binding sites in NANOG promoter, reporter gene transcription assays with or without interference or artificial overexpression of SOX9 and/or HMGB3, and protein-binding assays of SOX9-HMGB3 interaction. Clinicopathologic and prognostic significance of SOX9-HMGB3 overexpression in PCa was analyzed. RESULTS SOX9 activated NANOG gene transcription by preferentially binding to a highly conserved consensus cis-regulatory element (-573 to -568) in NANOG promoter, and promoted the expression of NANOG downstream oncogenic genes. Importantly, HMGB3 functioned as a partner of SOX9 to co-operatively enhance transactivation of NANOG by interacting with SOX9, predominantly via the HMG Box A domain of HMGB3. Overexpression of SOX9 and/or HMGB3 enhanced PCa cell survival and cell migration and were significantly associated with PCa progression. Notably, Cox proportional regression analysis showed that co-overexpression of both SOX9 and HMGB3 was an independent unfavorable prognosticator for both CRPC-free survival (relative risk [RR] = 3.779,95% confidence interval [CI]: 1.159-12.322, p = 0.028) and overall survival (RR = 3.615,95% CI: 1.101-11.876, p = 0.034). CONCLUSIONS These findings showed a novel SOX9/HMGB3/NANOG regulatory mechanism, deregulation of which played important roles in PCa progression.
Collapse
Affiliation(s)
- Yunyi Xu
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Miao Xu
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinglan Li
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Weng
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhengzheng Su
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Junya Tan
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xun Li
- Department of Ophthalmology and Research Laboratory of Ophthalmology and Vision Sciences, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Gong
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ni Chen
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xueqin Chen
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Zhou
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Bae SY, Bergom HE, Day A, Greene JT, Sychev ZE, Larson G, Corey E, Plymate SR, Freedman TS, Hwang JH, Drake JM. ZBTB7A as a novel vulnerability in neuroendocrine prostate cancer. Front Endocrinol (Lausanne) 2023; 14:1093332. [PMID: 37065756 PMCID: PMC10090553 DOI: 10.3389/fendo.2023.1093332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/08/2023] [Indexed: 03/31/2023] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is a highly aggressive subtype of prostate cancer. NEPC is characterized by the loss of androgen receptor (AR) signaling and transdifferentiation toward small-cell neuroendocrine (SCN) phenotypes, which results in resistance to AR-targeted therapy. NEPC resembles other SCN carcinomas clinically, histologically and in gene expression. Here, we leveraged SCN phenotype scores of various cancer cell lines and gene depletion screens from the Cancer Dependency Map (DepMap) to identify vulnerabilities in NEPC. We discovered ZBTB7A, a transcription factor, as a candidate promoting the progression of NEPC. Cancer cells with high SCN phenotype scores showed a strong dependency on RET kinase activity with a high correlation between RET and ZBTB7A dependencies in these cells. Utilizing informatic modeling of whole transcriptome sequencing data from patient samples, we identified distinct gene networking patterns of ZBTB7A in NEPC versus prostate adenocarcinoma. Specifically, we observed a robust association of ZBTB7A with genes promoting cell cycle progression, including apoptosis regulating genes. Silencing ZBTB7A in a NEPC cell line confirmed the dependency on ZBTB7A for cell growth via suppression of the G1/S transition in the cell cycle and induction of apoptosis. Collectively, our results highlight the oncogenic function of ZBTB7A in NEPC and emphasize the value of ZBTB7A as a promising therapeutic strategy for targeting NEPC tumors.
Collapse
Affiliation(s)
- Song Yi Bae
- Department of Pharmacology, University of Minnesota-Twin Cities, Minneapolis, MN, United States
| | - Hannah E. Bergom
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN, United States
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Abderrahman Day
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN, United States
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, United States
| | - Joseph T. Greene
- Department of Pharmacology, University of Minnesota-Twin Cities, Minneapolis, MN, United States
| | - Zoi E. Sychev
- Department of Pharmacology, University of Minnesota-Twin Cities, Minneapolis, MN, United States
| | - Gabrianne Larson
- Department of Pharmacology, University of Minnesota-Twin Cities, Minneapolis, MN, United States
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, United States
| | - Stephen R. Plymate
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, United States
- Geriatric Research, Education, and Clinical Center, Veterans Affairs (VA) Puget Sound Health Care System, Seattle, WA, United States
| | - Tanya S. Freedman
- Department of Pharmacology, University of Minnesota-Twin Cities, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Justin H. Hwang
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN, United States
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
- Department of Urology, University of Washington, Seattle, WA, United States
| | - Justin M. Drake
- Department of Pharmacology, University of Minnesota-Twin Cities, Minneapolis, MN, United States
- Department of Urology, University of Washington, Seattle, WA, United States
- Department of Urology, University of Minnesota-Twin Cities, Minneapolis, MN, United States
| |
Collapse
|
17
|
Zhang Y, Bao S, Wang D, Lu W, Xu S, Zhou W, Wang X, Xu X, Ding X, Zhao S. Downregulation of KLF10 contributes to the regeneration of survived renal tubular cells in cisplatin-induced acute kidney injury via ZBTB7A-KLF10-PTEN axis. Cell Death Discov 2023; 9:82. [PMID: 36878898 PMCID: PMC9988960 DOI: 10.1038/s41420-023-01381-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
Acute kidney injury (AKI) is a common clinical dysfunction with complicated pathophysiology and limited therapeutic methods. Renal tubular injury and the following regeneration process play a vital role in the course of AKI, but the underlining molecular mechanism remains unclear. In this study, network-based analysis of online transcriptional data of human kidney found that KLF10 was closely related to renal function, tubular injury and regeneration in various renal diseases. Three classical mouse models confirmed the downregulation of KLF10 in AKI and its correlation with tubular regeneration and AKI outcome. The 3D renal tubular model in vitro and fluorescent visualization system of cellular proliferation were constructed to show that KLF10 declined in survived cells but increased during tubular formation or conquering proliferative impediment. Furthermore, overexpression of KLF10 significantly inhibited, whereas knockdown of KLF10 extremely promoted the capacity of proliferation, injury repairing and lumen-formation of renal tubular cells. In mechanism, PTEN/AKT pathway were validated as the downstream of KLF10 and participated in its regulation of tubular regeneration. By adopting proteomic mass spectrum and dual-luciferase reporter assay, ZBTB7A were found to be the upstream transcription factor of KLF10. Our findings suggest that downregulation of KLF10 positively contributed to tubular regeneration in cisplatin induced acute kidney injury via ZBTB7A-KLF10-PTEN axis, which gives insight into the novel therapeutic and diagnostical target of AKI.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyu Bao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daxi Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sujuan Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiran Zhou
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Medical Center of Kidney Disease, Shanghai, China. .,Kidney and Dialysis Institute of Shanghai, Shanghai, China. .,Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Medical Center of Kidney Disease, Shanghai, China. .,Kidney and Dialysis Institute of Shanghai, Shanghai, China. .,Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China.
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Medical Center of Kidney Disease, Shanghai, China. .,Kidney and Dialysis Institute of Shanghai, Shanghai, China. .,Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China.
| |
Collapse
|
18
|
Ren R, Horton JR, Chen Q, Yang J, Liu B, Huang Y, Blumenthal RM, Zhang X, Cheng X. Structural basis for transcription factor ZBTB7A recognition of DNA and effects of ZBTB7A somatic mutations that occur in human acute myeloid leukemia. J Biol Chem 2023; 299:102885. [PMID: 36626981 PMCID: PMC9932118 DOI: 10.1016/j.jbc.2023.102885] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/09/2023] Open
Abstract
ZBTB7A belongs to a small family of transcription factors having three members in humans (7A, 7B, and 7C). They share a BTB/POZ protein interaction domain at the amino end and a zinc-finger DNA-binding domain at the carboxyl end. They control the transcription of a wide range of genes, having varied functions in hematopoiesis, oncogenesis, and metabolism (in particular glycolysis). ZBTB7A-binding profiles at gene promoters contain a consensus G(a/c)CCC motif, followed by a CCCC sequence in some instances. Structural and mutational investigations suggest that DNA-specific contacts with the four-finger tandem array of ZBTB7A are formed sequentially, initiated from ZF1-ZF2 binding to G(a/c)CCC before spreading to ZF3-ZF4, which bind the DNA backbone and the 3' CCCC sequence, respectively. Here, we studied some mutations found in t(8;21)-positive acute myeloid leukemia patients that occur within the ZBTB7A DNA-binding domain. We determined that these mutations generally impair ZBTB7A DNA binding, with the most severe disruptions resulting from mutations in ZF1 and ZF2, and the least from a frameshift mutation in ZF3 that results in partial mislocalization. Information provided here on ZBTB7A-DNA interactions is likely applicable to ZBTB7B/C, which have overlapping functions with ZBTB7A in controlling primary metabolism.
Collapse
Affiliation(s)
- Ren Ren
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John R Horton
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qin Chen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jie Yang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bin Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, Texas, USA
| | - Robert M Blumenthal
- Department of Medical Microbiology and Immunology, and Program in Bioinformatics, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Xing Zhang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Xiaodong Cheng
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
19
|
Microcribriform Adenocarcinoma of Salivary Glands: A Unique Tumor Entity Characterized by an SS18::ZBTB7A Fusion. Am J Surg Pathol 2023; 47:194-201. [PMID: 36221318 DOI: 10.1097/pas.0000000000001980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The landscape of salivary gland carcinomas is ever-changing, with a growing list of new tumors and newly elucidated variants of well-known tumor entities. The routine use of next-generation sequencing has been instrumental in identifying novel fusions and tumor entities, which has helped bring the classification to a more objective and evidenced-based model. However, morphology remains critical in assessing the validity of these novel molecular findings, and most importantly, in assessing which of these findings will have an impact on the prognosis and treatment decisions for patients. The recognition of microsecretory adenocarcinoma (MSA) as a distinct low-grade malignancy of salivary glands, underpinned by MEF2C::SS18 , and a single possibly related case of SS18::ZBTB7A , recently expanded this growing list of distinctive tumors. It was not until now, however, that the morphology of the latter case was known to be unique and reproducible. The authors have now seen 4 of these distinctive tumors that show a combination of distinctive oncocytic cells forming compact glandular growth as well as amphophilic cells forming tubular growth, and suggest the appellation "microcribriform adenocarcinoma" (MCA). So far, these tumors appear to preferentially occur in nonoral sites (2 parotid, 1 submandibular gland, and 1 bronchial seromucous glands). By immunohistochemistry, they express S100 and SOX-10 with focal outer myoepithelial cells marked by circumferential p63, p40, and smooth muscle actin staining around some of the nests and tubules. The tumors show infiltrative growth within a hyalinized and myxoid stroma. Cytologically, they appear generally low grade, similar to MSA. The morphologic and molecular uniformity of these 4 microcribriform adenocarcinoma cases warrants their recognition, and while related to MSA, they are sufficiently different to be classified as a distinct tumor. So far, in limited follow-up, these tumors appear to be relatively indolent.
Collapse
|
20
|
Long X, Liu X, Deng T, Chen J, Lan J, Zhang S, Zhou M, Guo D, Zhou J. LARP6 suppresses colorectal cancer progression through ZNF267/SGMS2-mediated imbalance of sphingomyelin synthesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:33. [PMID: 36691044 PMCID: PMC9872320 DOI: 10.1186/s13046-023-02605-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND With increasing incidence and mortality, colorectal cancer (CRC) seriously endangers human health. LARP6, a member of La-related protein (LARP) family, is a RNA binding protein and probably associates with CRC progression, but its specific roles and mechanisms in CRC still remain unknown. METHOD Quantitative real-time PCR (qPCR), western blot, and immunohistochemistry were employed to examine LARP6 expression in CRC tissues. Using the stable LARP6 overexpression or interference CRC cell lines, the effect of LARP6 on CRC progression were evaluated. High-throughput RNA immunoprecipitation sequencing (RIP-seq) and a series of relevant experiments were conducted to explain how LARP6 functions. SPSS software was used for statistical analysis. RESULT In this study, we found that LARP6 expression is downregulated in CRC and correlates with patients' overall survival and relapse-free survival. Furthermore, altered LARP6 expression influences CRC cells invasion and metastasis. Mechanically, we discovered that LARP6 bind ZNF267 mRNA and regulated its stability and translation. LARP6 inhibited expression of SGMS2, a downstream target of ZNF267, resulting in ceramide and sphingomyelin imbalance in CRC cells. Interestingly, LARP6 also enhances autophagy activity of CRC cells, and the effect was at least partially determined by the inhibition of SGMS2-mediated sphingomyelin synthesis. CONCLUSION Our study showed how LARP6/ZNF267/SGMS2 axis influence CRC progression, which contributes to further understanding of the molecular mechanisms underlying CRC development.
Collapse
Affiliation(s)
- Xiaoli Long
- grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China ,grid.284723.80000 0000 8877 7471Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Xunhua Liu
- grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China ,grid.284723.80000 0000 8877 7471Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Ting Deng
- Department of Pathology, YunFu People’s Hospital, Yunfu, 527300 China
| | - Jianxiong Chen
- grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China ,grid.284723.80000 0000 8877 7471Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Jiawen Lan
- grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China ,grid.284723.80000 0000 8877 7471Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Sijing Zhang
- grid.284723.80000 0000 8877 7471Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Miao Zhou
- grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Dan Guo
- grid.284723.80000 0000 8877 7471Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Jun Zhou
- grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China ,grid.284723.80000 0000 8877 7471Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China ,Department of Pathology, YunFu People’s Hospital, Yunfu, 527300 China
| |
Collapse
|
21
|
Jeong JH, Park SH, Kim H, Nam HY, Kim SH, Jeong M, Kong MJ, Son J, Jeong JE, Song JH, Kim SW, Choi KC. ZBTB7A suppresses glioblastoma tumorigenesis through the transcriptional repression of EPB41L5. Exp Mol Med 2023; 55:43-54. [PMID: 36596853 PMCID: PMC9898510 DOI: 10.1038/s12276-022-00908-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma multiforme (GBM), the most aggressive and malignant glioma, has a poor prognosis. Although patients with GBM are treated with surgery, chemotherapy, and radiation therapy, GBM is highly resistant to treatment, making it difficult and expensive to treat. In this study, we analyzed the Gene Expression Profiling Interactive Analysis dataset, the Cancer Genome Atlas dataset, and Gene Expression Omnibus array data. ZBTB7A (also called FBI1/POKEMON/LRF) was found to be highly expressed in low-grade glioma but significantly downregulated in patients with GBM. ZBTB7A is a transcription factor that plays an important role in many developmental stages, including cell proliferation. The activation of epithelial-mesenchymal transition (EMT) is a key process in cancer progression and metastasis. Erythrocyte membrane protein band 4.1 like 5 (EPB41L5) is an essential protein for EMT progression and metastasis in various types of cancer. We found that ZBTB7A depletion in U87 cells induced GBM progression and metastasis. Based on RNA sequencing data, ZBTB7A directly binds to the promoter of the EPB41L5 gene, reducing its expression and inhibiting GBM progression. We demonstrated that ZBTB7A dramatically inhibits GBM tumor growth through transcriptional repression of EPB41L5. Thus, both ZBTB7A and EPB41L5 may be potential biomarkers and novel therapeutic targets for GBM treatment. Overall, we discovered the role of a novel tumor suppressor that directly inhibits GBM progression (ZBTB7A) and identified EPB41L5 as a therapeutic target protein for patients with GBM.
Collapse
Affiliation(s)
- Ji-Hoon Jeong
- grid.413967.e0000 0001 0842 2126Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Ho Park
- grid.413967.e0000 0001 0842 2126Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyunhee Kim
- grid.413967.e0000 0001 0842 2126Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hae Yun Nam
- grid.413967.e0000 0001 0842 2126Department of Biochemistry and Molecular Biology, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Hak Kim
- grid.14005.300000 0001 0356 9399Department of Animal Science, Chonnam National University, Gwangju, Korea
| | - Minseok Jeong
- grid.413967.e0000 0001 0842 2126Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min-Jeong Kong
- grid.413967.e0000 0001 0842 2126Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jihyun Son
- grid.413967.e0000 0001 0842 2126Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Eun Jeong
- grid.413967.e0000 0001 0842 2126Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Hye Song
- grid.418974.70000 0001 0573 0246Korea Food Research Institute, Wanju-gun, 55365 Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Kyung-Chul Choi
- Department of Biomedical Sciences, AAMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
22
|
Adamiecki R, Hryniewicz-Jankowska A, Ortiz MA, Li X, Porter-Hansen BA, Nsouli I, Bratslavsky G, Kotula L. In Vivo Models for Prostate Cancer Research. Cancers (Basel) 2022; 14:5321. [PMID: 36358740 PMCID: PMC9654339 DOI: 10.3390/cancers14215321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/28/2022] Open
Abstract
In 2022, prostate cancer (PCa) is estimated to be the most commonly diagnosed cancer in men in the United States-almost 270,000 American men are estimated to be diagnosed with PCa in 2022. This review compares and contrasts in vivo models of PCa with regards to the altered genes, signaling pathways, and stages of tumor progression associated with each model. The main type of model included in this review are genetically engineered mouse models, which include conditional and constitutive knockout model. 2D cell lines, 3D organoids and spheroids, xenografts and allografts, and patient derived models are also included. The major applications, advantages and disadvantages, and ease of use and cost are unique to each type of model, but they all make it easier to translate the tumor progression that is seen in the mouse prostate to the human prostate. Although both human and mouse prostates are androgen-dependent, the fact that the native, genetically unaltered prostate in mice cannot give rise to carcinoma is an especially critical component of PCa models. Thanks to the similarities between the mouse and human genome, our knowledge of PCa has been expanded, and will continue to do so, through models of PCa.
Collapse
Affiliation(s)
- Robert Adamiecki
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
- Department of Urology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
| | - Anita Hryniewicz-Jankowska
- Department of Urology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Maria A. Ortiz
- Department of Urology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
| | - Xiang Li
- Department of Urology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
| | - Baylee A. Porter-Hansen
- Department of Urology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
| | - Imad Nsouli
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
| | - Gennady Bratslavsky
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
- Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
| | - Leszek Kotula
- Department of Urology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
- Upstate Cancer Center, SUNY Upstate Medical University, 750 East Adams Str., Syracuse, NY 13010, USA
| |
Collapse
|
23
|
Zhu X, Trimarco JD, Williams CA, Barrera A, Reddy TE, Heaton NS. ZBTB7A promotes virus-host homeostasis during human coronavirus 229E infection. Cell Rep 2022; 41:111540. [PMID: 36243002 PMCID: PMC9533670 DOI: 10.1016/j.celrep.2022.111540] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/24/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022] Open
Abstract
The cellular fate after infection with human coronaviruses (HCoVs) is typically death. Previous data suggest, however, that the transcriptional state of an individual cell may sometimes allow additional outcomes of infection. Here, to probe the range of interactions a permissive cell type can have with a HCoV, we perform a CRISPR activation screen with HCoV-229E. The screen identified the transcription factor ZBTB7A, which strongly promotes cell survival after infection. Rather than suppressing viral infection, ZBTB7A upregulation allows the virus to induce a persistent infection and homeostatic state with the cell. We also find that control of oxidative stress is a primary driver of cellular survival during HCoV-229E infection. These data illustrate that, in addition to the nature of the infecting virus and the type of cell that it encounters, the cellular gene expression profile prior to infection can affect the eventual fate.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Joseph D. Trimarco
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Courtney A. Williams
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA,Center for Genomic and Computational Biology, Duke University, Durham, NC, USA,Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Alejandro Barrera
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA,Center for Genomic and Computational Biology, Duke University, Durham, NC, USA,Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Timothy E. Reddy
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA,Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA,Center for Genomic and Computational Biology, Duke University, Durham, NC, USA,Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA,Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA,Corresponding author
| |
Collapse
|
24
|
Xu X, Shobuike T, Shiraki M, Kamohara A, Hirata H, Murayama M, Mawatari D, Ueno M, Morimoto T, Kukita T, Mawatari M, Kukita A. Leukemia/lymphoma-related factor (LRF) or osteoclast zinc finger protein (OCZF) overexpression promotes osteoclast survival by increasing Bcl-xl mRNA: A novel regulatory mechanism mediated by the RNA binding protein SAM68. J Transl Med 2022; 102:1000-1010. [PMID: 36775415 DOI: 10.1038/s41374-022-00792-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/08/2022] Open
Abstract
RANKL induces NFATc1, a key transcriptional factor to induce osteoclast-specific genes such as cathepsin K, whereas transcriptional control of osteoclast survival is not fully understood. Leukemia/lymphoma-related factor (LRF) in mouse and osteoclast zinc finger protein (OCZF) in rat are zinc finger and BTB domain-containing protein (zBTB) family of transcriptional regulators, and are critical regulators of hematopoiesis. We have previously shown that differentiation and survival were enhanced in osteoclasts from OCZF-Transgenic (Tg) mice. In the present study, we show a possible mechanism of osteoclast survival regulated by LRF/OCZF and the role of OCZF overexpression in pathological bone loss. In the in vitro cultures, LRF was highly colocalized with NFATc1 in cells of early stage in osteoclastogenesis, but only LRF expression persisted after differentiation into mature osteoclasts. LRF expression was further enhanced in resorbing osteoclasts formed on dentin slices. Osteoclast survival inhibitor such as alendronate, a bisphosphonate reduced LRF expression. Micro CT evaluation revealed that femurs of OCZF-Tg mice showed significantly lower bone volume compared to that of WT mice. Furthermore, OCZF overexpression markedly promoted bone loss in ovariectomy-induced osteolytic mouse model. The expression of anti-apoptotic Bcl-xl mRNA, which is formed by alternative splicing, was enhanced in the cultures in which osteoclasts are formed from OCZF-Tg mice. In contrast, the expression of pro-apoptotic Bcl-xs mRNA was lost in the culture derived from OCZF-Tg mice. We found that the expression levels of RNA binding splicing regulator, Src substrate associated in mitosis of 68 kDa (Sam68) protein were markedly decreased in OCZF-Tg mice-derived osteoclasts. In addition, shRNA-mediated knockdown of Sam68 expression increased the expression of Bcl-xl mRNA, suggesting that SAM68 regulates the expression of Bcl-xl. These results indicate that OCZF overexpression reduces protein levels of Sam68, thereby promotes osteoclast survival, and suggest that LRF/OCZF is a promising target for regulating pathological bone loss.
Collapse
Affiliation(s)
- Xianghe Xu
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
- Department of Molecular Cell Biology & Oral Anatomy, Faculty of Dentistry, Kyushu University, Fukuoka, Japan
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Takeo Shobuike
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Makoto Shiraki
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Asana Kamohara
- Department of Oral & Maxillofacial Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Daisuke Mawatari
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Tadatsugu Morimoto
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology & Oral Anatomy, Faculty of Dentistry, Kyushu University, Fukuoka, Japan
| | - Masaaki Mawatari
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Akiko Kukita
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.
- Research Center of Arthroplasty, Faculty of Medicine, Saga University, Saga, Japan.
| |
Collapse
|
25
|
Sardar D, Chen HC, Reyes A, Varadharajan S, Jain A, Mohila C, Curry R, Lozzi B, Rajendran K, Cervantes A, Yu K, Jalali A, Rao G, Mack SC, Deneen B. Sox9 directs divergent epigenomic states in brain tumor subtypes. Proc Natl Acad Sci U S A 2022; 119:e2202015119. [PMID: 35858326 PMCID: PMC9303974 DOI: 10.1073/pnas.2202015119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/31/2022] [Indexed: 01/17/2023] Open
Abstract
Epigenetic dysregulation is a universal feature of cancer that results in altered patterns of gene expression that drive malignancy. Brain tumors exhibit subtype-specific epigenetic alterations; however, the molecular mechanisms responsible for these diverse epigenetic states remain unclear. Here, we show that the developmental transcription factor Sox9 differentially regulates epigenomic states in high-grade glioma (HGG) and ependymoma (EPN). Using our autochthonous mouse models, we found that Sox9 suppresses HGG growth and expands associated H3K27ac states, while promoting ZFTA-RELA (ZRFUS) EPN growth and diminishing H3K27ac states. These contrasting roles for Sox9 correspond with protein interactions with histone deacetylating complexes in HGG and an association with the ZRFUS oncofusion in EPN. Mechanistic studies revealed extensive Sox9 and ZRFUS promoter co-occupancy, indicating functional synergy in promoting EPN tumorigenesis. Together, our studies demonstrate how epigenomic states are differentially regulated in distinct subtypes of brain tumors, while revealing divergent roles for Sox9 in HGG and EPN tumorigenesis.
Collapse
Affiliation(s)
- Debosmita Sardar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
| | - Hsiao-Chi Chen
- Cancer Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030
| | - Amanda Reyes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Department of Biomedical Engineering, University of Houston, Houston, TX 77004
| | - Srinidhi Varadharajan
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030
| | - Carrie Mohila
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Rachel Curry
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Cancer Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Genetics and Genomics Program, Baylor College of Medicine, Houston, TX 77030
| | - Kavitha Rajendran
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
| | - Alexis Cervantes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
| | - Kwanha Yu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
| | - Ali Jalali
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030
| | - Stephen C. Mack
- Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Cancer Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
26
|
Liu J, Chou Z, Li C, Huang K, Wang X, Li X, Han C, Al-Danakh A, Li X, Song X. ZBTB7A, a miR-144-3p targeted gene, accelerates bladder cancer progression via downregulating HIC1 expression. Cancer Cell Int 2022; 22:179. [PMID: 35501800 PMCID: PMC9063087 DOI: 10.1186/s12935-022-02596-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/18/2022] [Indexed: 12/24/2022] Open
Abstract
Background Zinc finger and BTB domain-containing 7A (ZBTB7A) is a member of the POK family of transcription factors that plays an oncogenic or tumor-suppressive role in different cancers depending on the type and genetic context of cancer. However, the function and molecular mechanism of ZBTB7A in bladder cancer (BC) remain elusive. Methods The role of ZBTB7A in bladder cancer was detected by colony formation, transwell, and tumor formation assays. The expression levels of ZBTB7A, HIC1, and miR-144-3p were analyzed by qRT-PCR and Western blot. Bioinformatics analysis and a dual-luciferase reporter assay were used to assess the effect of ZBTB7A on the promoter activity of HIC1. Results The present study revealed that knockdown of ZBTB7A suppressed BC cell growth and migration, as indicated by an approximately 50% reduction in the number of colonies and an approximately 70% reduction in the number of migrated cells. Loss of ZBTB7A inhibited tumor growth in vivo, resulting in a 75% decrease in tumor volume and an 80% decrease in tumor weight. Further mechanistic studies revealed that ZBTB7A bound to the hypermethylated in cancer 1 (HIC1) promoter and downregulated HIC1 expression, accelerating the malignant behavior of BC. Increased expression of ZBTB7A in BC tissues was negatively corrected with the expression of HIC1. Moreover, ZBTB7A was a target of miR-144-3p, which decreased ZBTB7A expression in BC. Conclusion Our data demonstrate that ZBTB7A, a targeted gene of miR-144-3p, promoted tumorigenesis of BC through downregulating HIC1 expression. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02596-w.
Collapse
Affiliation(s)
- Junqiang Liu
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhiyuan Chou
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Chun Li
- Central Laboratory, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Kai Huang
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xuejian Wang
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiunan Li
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Chuanchun Han
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Abdullah Al-Danakh
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaodong Li
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Xishuang Song
- Department of Urology of First Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
27
|
Carrasco-Garcia E, Lopez L, Moncho-Amor V, Carazo F, Aldaz P, Collado M, Bell D, Gaafar A, Karamitopoulou E, Tzankov A, Hidalgo M, Rubio Á, Serrano M, Lawrie CH, Lovell-Badge R, Matheu A. SOX9 Triggers Different Epithelial to Mesenchymal Transition States to Promote Pancreatic Cancer Progression. Cancers (Basel) 2022; 14:cancers14040916. [PMID: 35205666 PMCID: PMC8870732 DOI: 10.3390/cancers14040916] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Pancreatic cancers are lethal types of cancer. A majority of patients progress to an advanced and metastatic disease, which remains a major clinical problem. Therefore, it is crucial to identify critical regulators to help predict the disease progression and to develop more efficacious therapeutic approaches. In this work we found that an increased expression of the developmental factor SOX9 is associated with metastasis, a poor prognosis and resistance to therapy in pancreatic ductal adenocarcinoma patients and in cell cultures. We also found that this effect is at least in part due to the ability of SOX9 to regulate the activity of stem cell factors, such as BMI1, in addition to those involved in EMT and metastasis. Abstract Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers mainly due to spatial obstacles to complete resection, early metastasis and therapy resistance. The molecular events accompanying PDAC progression remain poorly understood. SOX9 is required for maintaining the pancreatic ductal identity and it is involved in the initiation of pancreatic cancer. In addition, SOX9 is a transcription factor linked to stem cell activity and is commonly overexpressed in solid cancers. It cooperates with Snail/Slug to induce epithelial-mesenchymal transition (EMT) during neural development and in diseases such as organ fibrosis or different types of cancer. Methods: We investigated the roles of SOX9 in pancreatic tumor cell plasticity, metastatic dissemination and chemoresistance using pancreatic cancer cell lines as well as mouse embryo fibroblasts. In addition, we characterized the clinical relevance of SOX9 in pancreatic cancer using human biopsies. Results: Gain- and loss-of-function of SOX9 in PDAC cells revealed that high levels of SOX9 increased migration and invasion, and promoted EMT and metastatic dissemination, whilst SOX9 silencing resulted in metastasis inhibition, along with a phenotypic reversion to epithelial features and loss of stemness potential. In both contexts, EMT factors were not altered. Moreover, high levels of SOX9 promoted resistance to gemcitabine. In contrast, overexpression of SOX9 was sufficient to promote metastatic potential in K-Ras transformed MEFs, triggering EMT associated with Snail/Slug activity. In clinical samples, SOX9 expression was analyzed in 198 PDAC cases by immunohistochemistry and in 53 patient derived xenografts (PDXs). SOX9 was overexpressed in primary adenocarcinomas and particularly in metastases. Notably, SOX9 expression correlated with high vimentin and low E-cadherin expression. Conclusions: Our results indicate that SOX9 facilitates PDAC progression and metastasis by triggering stemness and EMT.
Collapse
Affiliation(s)
- Estefania Carrasco-Garcia
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (L.L.); (V.M.-A.); (P.A.)
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
- Correspondence: (E.C.-G.); (A.M.); Tel.: +34-943-006073 (E.C.-G. & A.M.); Fax: +34-943-006250 (E.C.-G. & A.M.)
| | - Lidia Lopez
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (L.L.); (V.M.-A.); (P.A.)
| | - Veronica Moncho-Amor
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (L.L.); (V.M.-A.); (P.A.)
- The Francis Crick Institute, London NW1 1AT, UK; (D.B.); (R.L.-B.)
| | - Fernando Carazo
- School of Engineering, University of Navarra, 20009 San Sebastian, Spain; (F.C.); (Á.R.)
| | - Paula Aldaz
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (L.L.); (V.M.-A.); (P.A.)
| | - Manuel Collado
- Health Research Institute of Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), 15706 Santiago de Compostela, Spain;
| | - Donald Bell
- The Francis Crick Institute, London NW1 1AT, UK; (D.B.); (R.L.-B.)
| | - Ayman Gaafar
- Department of Pathology, Cruces University Hospital, 48903 Barakaldo, Spain;
| | | | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, 4056 Basel, Switzerland;
| | - Manuel Hidalgo
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain;
- New York-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY 10065, USA
| | - Ángel Rubio
- School of Engineering, University of Navarra, 20009 San Sebastian, Spain; (F.C.); (Á.R.)
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain;
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Charles H. Lawrie
- Molecular Oncology Group, Biodonostia Institute, 20014 San Sebastian, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | | | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain; (L.L.); (V.M.-A.); (P.A.)
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- Correspondence: (E.C.-G.); (A.M.); Tel.: +34-943-006073 (E.C.-G. & A.M.); Fax: +34-943-006250 (E.C.-G. & A.M.)
| |
Collapse
|
28
|
von der Lippe C, Tveten K, Prescott TE, Holla ØL, Busk ØL, Burke KB, Sansbury FH, Baptista J, Fry AE, Lim D, Jolles S, Evans J, Osio D, Macmillan C, Bruno I, Faltera F, Climent S, Urreitzi R, Hoenicka J, Palau F, Cohen ASA, Engleman K, Zhou D, Amudhavalli SM, Jeanne M, Bonnet-Brilhault F, Lévy J, Drunat S, Derive N, Haug MG, Thorstensen WM. Heterozygous variants in ZBTB7A cause a neurodevelopmental disorder associated with symptomatic overgrowth of pharyngeal lymphoid tissue, macrocephaly, and elevated fetal hemoglobin. Am J Med Genet A 2021; 188:272-282. [PMID: 34515416 DOI: 10.1002/ajmg.a.62492] [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: 06/19/2021] [Revised: 08/14/2021] [Accepted: 08/21/2021] [Indexed: 11/11/2022]
Abstract
By clinical whole exome sequencing, we identified 12 individuals with ages 3 to 37 years, including three individuals from the same family, with a consistent phenotype of intellectual disability (ID), macrocephaly, and overgrowth of adenoid tissue. All 12 individuals harbored a rare heterozygous variant in ZBTB7A which encodes the transcription factor Zinc finger and BTB-domain containing protein 7A, known to play a role in lympho- and hematopoiesis. ID was generally mild. Fetal hemoglobin (HbF) fraction was elevated 2.2%-11.2% (reference value <2% in individuals > 6 months) in four of the five individuals for whom results were available. Ten of twelve individuals had undergone surgery at least once for lymphoid hypertrophy limited to the pharynx. In the most severely affected individual (individual 1), airway obstruction resulted in 17 surgical procedures before the age of 13 years. Sleep apnea was present in 8 of 10 individuals. In the nine unrelated individuals, ZBTB7A variants were novel and de novo. The six frameshift/nonsense and four missense variants were spread throughout the gene. This is the first report of a cohort of individuals with this novel syndromic neurodevelopmental disorder.
Collapse
Affiliation(s)
| | - Kristian Tveten
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Trine E Prescott
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Øystein L Holla
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Øyvind L Busk
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Katherine B Burke
- All Wales Medical Genomics Service, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Francis H Sansbury
- All Wales Medical Genomics Service, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Júlia Baptista
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK.,Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Andrew E Fry
- All Wales Medical Genomics Service, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK.,Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Derek Lim
- Clinical Genetics, West Midlands Regional Genetics Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Jennifer Evans
- Department of Paediatrics, University Hospital of Wales, Cardiff, UK
| | - Deborah Osio
- Clinical Genetics, West Midlands Regional Genetics Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Carol Macmillan
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Irene Bruno
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Flavio Faltera
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Salvador Climent
- Pediatrics Service, Hospital General d'Ontinyent, Ontinyent, Spain
| | - Roser Urreitzi
- Department of Clinical Biochemistry and CIBERER, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Janet Hoenicka
- Laboratory of Neurogenetics and Molecular Medicine - IPER and CIBERER, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Francesc Palau
- Laboratory of Neurogenetics and Molecular Medicine - IPER and CIBERER, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Department of Genetic Medicine - IPER, Hospital Sant Joan de Déu, Barcelona, Spain.,Hospital Clínic and Division of Pediatrics, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Ana S A Cohen
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, Missouri, USA.,Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri, USA.,Kansas City School of Medicine, University of Missouri, Kansas City, Missouri, USA
| | - Kendra Engleman
- Kansas City School of Medicine, University of Missouri, Kansas City, Missouri, USA.,Division of Clinical Genetics, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Dihong Zhou
- Kansas City School of Medicine, University of Missouri, Kansas City, Missouri, USA.,Division of Clinical Genetics, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Shivarajan M Amudhavalli
- Kansas City School of Medicine, University of Missouri, Kansas City, Missouri, USA.,Division of Clinical Genetics, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Médéric Jeanne
- Service de Génétique, Centre Hospitalier Régional Universitaire, Tours, France.,UMR1253, iBrain, University of Tours, INSERM, Tours, France.,Excellence Center in Autism and Neurodevelopmental Disorders, Centre Hospitalier Régional Universitaire, Tours, France
| | - Frédérique Bonnet-Brilhault
- UMR1253, iBrain, University of Tours, INSERM, Tours, France.,Excellence Center in Autism and Neurodevelopmental Disorders, Centre Hospitalier Régional Universitaire, Tours, France
| | - Jonathan Lévy
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France.,Laboratoire de Biologie Médicale Multisites SeqOIA, Paris, France
| | - Séverine Drunat
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France.,Laboratoire de Biologie Médicale Multisites SeqOIA, Paris, France
| | - Nicolas Derive
- Laboratoire de Biologie Médicale Multisites SeqOIA, Paris, France
| | - Marte G Haug
- Department of Medical Genetics, St. Olavs University Hospital, Trondheim, Norway
| | - Wenche M Thorstensen
- Department of Otolaryngology, Head and Neck Surgery, St. Olavs University Hospital, Trondheim, Norway.,Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
29
|
Singh AK, Verma S, Kushwaha PP, Prajapati KS, Shuaib M, Kumar S, Gupta S. Role of ZBTB7A zinc finger in tumorigenesis and metastasis. Mol Biol Rep 2021; 48:4703-4719. [PMID: 34014468 DOI: 10.1007/s11033-021-06405-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/08/2021] [Indexed: 02/08/2023]
Abstract
The zinc finger and BTB (broad-complex, tramtrack and bric a brac) domain containing protein 7A (ZBTB7A) is a pleiotropic transcription factor that plays an important role in various stages of cell proliferation, differentiation, and other developmental processes. ZBTB7A is a member of the POK family that directly and specifically binds to short DNA recognition sites located near their target genes thereby acting as transcriptional activator or repressor. ZBTB7A overexpression has been associated with tumorigenesis and metastasis in various human cancer types, including breast, prostate, lung, ovarian, and colon cancer. However in some instances downregulation of ZBTB7A results in tumor progression, suggesting its role as a tumor suppressor. ZBTB7A is involved with complicated regulatory networks which include protein-protein and protein-nucleic acid interactions. ZBTB7A involvement in cancer progression and metastasis is perhaps enabled through the regulation of various signaling pathways depending on the type and genetic context of cancer. The association of ZBTB7A with other proteins affects cancer aggressiveness, therapeutic resistance and clinical outcome. This review focuses on the involvement of ZBTB7A in various signaling pathways and its role in cancer progression. We will also review the literature on ZBTB7A and cancer which could be potentially explored for its therapeutic implications.
Collapse
Affiliation(s)
- Atul Kumar Singh
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Shiv Verma
- Department of Urology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- Department of Urology, The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Prem Prakash Kushwaha
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Kumari Sunita Prajapati
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Mohd Shuaib
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Shashank Kumar
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India.
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
- Department of Urology, The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
30
|
Yan M, Sun S, Xu K, Huang X, Dou L, Pang J, Tang W, Shen T, Li J. Cardiac Aging: From Basic Research to Therapeutics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9570325. [PMID: 33777324 PMCID: PMC7969106 DOI: 10.1155/2021/9570325] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 02/07/2021] [Accepted: 02/11/2021] [Indexed: 01/12/2023]
Abstract
With research progress on longevity, we have gradually recognized that cardiac aging causes changes in heart structure and function, including progressive myocardial remodeling, left ventricular hypertrophy, and decreases in systolic and diastolic function. Elucidating the regulatory mechanisms of cardiac aging is a great challenge for biologists and physicians worldwide. In this review, we discuss several key molecular mechanisms of cardiac aging and possible prevention and treatment methods developed in recent years. Insights into the process and mechanism of cardiac aging are necessary to protect against age-related diseases, extend lifespan, and reduce the increasing burden of cardiovascular disease in elderly individuals. We believe that research on cardiac aging is entering a new era of unique significance for the progress of clinical medicine and social welfare.
Collapse
Affiliation(s)
- Mingjing Yan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
- Peking University Fifth School of Clinical Medicine, Beijing 100730, China
| | - Shenghui Sun
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Kun Xu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Lin Dou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Pang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
- Peking University Fifth School of Clinical Medicine, Beijing 100730, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
- Peking University Fifth School of Clinical Medicine, Beijing 100730, China
| |
Collapse
|
31
|
Pang B, Quan F, Ping Y, Hu J, Lan Y, Pang L. Dissecting the Invasion-Associated Long Non-coding RNAs Using Single-Cell RNA-Seq Data of Glioblastoma. Front Genet 2021; 11:633455. [PMID: 33505440 PMCID: PMC7831882 DOI: 10.3389/fgene.2020.633455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/14/2020] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is characterized by rapid and lethal infiltration of brain tissue, which is the primary cause of treatment failure and deaths for GBM. Therefore, understanding the molecular mechanisms of tumor cell invasion is crucial for the treatment of GBM. In this study, we dissected the single-cell RNA-seq data of 3345 cells from four patients and identified dysregulated genes including long non-coding RNAs (lncRNAs), which were involved in the development and progression of GBM. Based on co-expression network analysis, we identified a module (M1) that significantly overlapped with the largest number of dysregulated genes and was confirmed to be associated with GBM invasion by integrating EMT signature, experiment-validated invasive marker and pseudotime trajectory analysis. Further, we denoted invasion-associated lncRNAs which showed significant correlations with M1 and revealed their gradually increased expression levels along the tumor cell invasion trajectory, such as VIM-AS1, WWTR1-AS1, and NEAT1. We also observed the contribution of higher expression of these lncRNAs to poorer survival of GBM patients. These results were mostly recaptured in another validation data of 7930 single cells from 28 GBM patients. Our findings identified lncRNAs that played critical roles in regulating or controlling cell invasion and migration of GBM and provided new insights into the molecular mechanisms underlying GBM invasion as well as potential targets for the treatment of GBM.
Collapse
Affiliation(s)
- Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fei Quan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jing Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lin Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
32
|
To JC, Chiu AP, Tschida BR, Lo LH, Chiu CH, Li XX, Kuka TP, Linden MA, Amin K, Chan WC, Bell JB, Moriarity BS, Largaespada DA, Keng VW. ZBTB20 regulates WNT/CTNNB1 signalling pathway by suppressing PPARG during hepatocellular carcinoma tumourigenesis. JHEP Rep 2020; 3:100223. [PMID: 33604532 PMCID: PMC7873381 DOI: 10.1016/j.jhepr.2020.100223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 01/19/2023] Open
Abstract
Background & Aims Zinc finger and BTB domain containing 20 (ZBTB20) has been implicated as a potential oncogene in liver cancer. However, knockout studies have shown it to be a transcriptional repressor of the alpha-foetoprotein (Afp) gene in adult liver, and reduced levels of ZBTB20 allow for upregulation of AFP with increased tumour severity in certain cases of hepatocellular carcinoma (HCC). As there are many discrepancies in the literature regarding its role in liver tumourigenesis, the aim of this study was to elucidate the role of ZBTB20 in HCC tumourigenesis. Methods A reverse genetic study using the Sleeping Beauty (SB) transposon system in mice was performed to elucidate the role of ZBTB20 in HCC tumourigenesis. In vitro ZBTB20 gain- and loss-of-function experiments were used to assess the relationship amongst ZBTB20, peroxisome proliferator activated receptor gamma (PPARG) and catenin beta 1 (CTNNB1). Results Transgenic overexpression of ZBTB20 in hepatocytes and in the context of transformation related protein (T r p53) inactivation induced hepatic hypertrophy, activation of WNT/CTNNB1 signalling, and development of liver tumours. In vitro overexpression and knockout experiments using CRISPR/Cas9 demonstrated the important role for ZBTB20 in downregulating PPARG, resulting in activation of the WNT/CTNNB1 signalling pathway and its downstream effectors in HCC tumourigenesis. Conclusions These findings demonstrate a novel interaction between ZBTB20 and PPARG, which leads to activation of the WNT/CTNNB1 signalling pathway in HCC tumourigenesis. Lay summary ZBTB20 has been implicated as a potential oncogene in liver cancer. Herein, we uncover its important role in liver cancer development. We show that it interacts with PPARG to upregulate the WNT/CTNNB1 signalling pathway, leading to tumourigenesis.
Collapse
Key Words
- AFP, alpha-foetoprotein
- BTB/POZ, broad complex
- CTNNB1
- CTNNB1, catenin beta 1
- Fah, fumarylacetoacetate hydrolase
- GSK3B, glycogen synthase kinase 3 beta
- HCC, hepatocellular carcinoma
- HHL, immortalized human hepatic cell line
- Hepatocellular carcinoma
- IF, immunofluorescence
- NTBC, 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione
- OFP, orange fluorescent protein
- PHI, post-hydrodynamic injection
- POK, POZ and Kruppel
- PPARG
- PPARG, peroxisome proliferator activated receptor gamma
- Reverse genetic screen
- SB, Sleeping Beauty
- Sleeping Beauty
- ZBTB20
- ZBTB20, zinc finger and BTB domain containing 20
- qPCR, quantitative RT-PCR
- tramtrack, bric a brac/poxvirus and zinc finger
Collapse
Affiliation(s)
- Jeffrey C To
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Amy P Chiu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Barbara R Tschida
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Lilian H Lo
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Cynthia H Chiu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Xiao-Xiao Li
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Timothy P Kuka
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, MN, 55455, USA.,College of Natural Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Michael A Linden
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Khalid Amin
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wing-Cheung Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Jason B Bell
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Branden S Moriarity
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - David A Largaespada
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Vincent W Keng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR.,State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| |
Collapse
|
33
|
Gupta S, Singh AK, Prajapati KS, Kushwaha PP, Shuaib M, Kumar S. Emerging role of ZBTB7A as an oncogenic driver and transcriptional repressor. Cancer Lett 2020; 483:22-34. [PMID: 32348807 DOI: 10.1016/j.canlet.2020.04.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 02/08/2023]
Abstract
ZBTB7A is a member of the POK family of transcription factors that possesses a POZ-domain at the N-terminus and Krüppel-like zinc-finger at the c-terminus. ZBTB7A was initially isolated as a protein that binds to the inducer of the short transcript of HIV-1 virus TAT gene promoter. The protein forms a homodimer through protein-protein interaction via the N-terminus POZ-domains. ZBTB7A typically binds to the DNA elements through its zinc-finger domains and represses transcription both by modification of the chromatin organization and through the direct recruitment of transcription factors to gene regulatory regions. ZBTB7A is involved in several fundamental biological processes including cell proliferation, differentiation, and development. It also participates in hematopoiesis, adipogenesis, chondrogenesis, cellular metabolism and alternative splicing of BCLXL, DNA repair, development of oligodendrocytes, osteoclast and unfolded protein response. Aberrant ZBTB7A expression promotes oncogenic transformation and tumor progression, but also maintains a tumor suppressive role depending on the type and genetic context of cancer. In this comprehensive review we provide information about the structure, function, targets, and regulators of ZBTB7A and its role as an oncogenic driver and transcriptional repressor in various human diseases.
Collapse
Affiliation(s)
- Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, OH 44106, USA; The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA; Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA; Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| | - Atul Kumar Singh
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, India
| | - Kumari Sunita Prajapati
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, India
| | - Prem Prakash Kushwaha
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, India
| | - Mohd Shuaib
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, India
| | - Shashank Kumar
- Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, India.
| |
Collapse
|
34
|
Computational Identification of Tumor Suppressor Genes Based on Gene Expression Profiles in Normal and Cancerous Gastrointestinal Tissues. JOURNAL OF ONCOLOGY 2020; 2020:2503790. [PMID: 32774369 PMCID: PMC7396062 DOI: 10.1155/2020/2503790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022]
Abstract
Cancer prevails in various gastrointestinal (GI) organs, such as esophagus, stomach, and colon. However, the small intestine has an extremely low cancer risk. It is interesting to investigate the molecular cues that could explain the significant difference in cancer incidence rates among different GI tissues. Using several large-scale normal and cancer tissue genomics datasets, we compared the gene expression profiling between small intestine and other GI tissues and between GI cancers and normal tissues. We identified 17 tumor suppressor genes (TSGs) which showed significantly higher expression levels in small intestine than in other GI tissues and significantly lower expression levels in GI cancers than in normal tissues. These TSGs were mainly involved in metabolism, immune, and cell growth signaling-associated pathways. Many TSGs had a positive expression correlation with survival prognosis in various cancers, confirming their tumor suppressive function. We demonstrated that the downregulation of many TSGs was associated with their hypermethylation in cancer. Moreover, we showed that the expression of many TSGs inversely correlated with tumor purity and positively correlated with antitumor immune response in various cancers, suggesting that these TSGs may exert their tumor suppressive function by promoting antitumor immunity. Furthermore, we identified a transcriptional regulatory network of the TSGs and their master transcriptional regulators (MTRs). Many of MTRs have been recognized as tumor suppressors, such as HNF4A, ZBTB7A, p53, and RUNX3. The TSGs could provide new molecular cues associated with tumorigenesis and tumor development and have potential clinical implications for cancer diagnosis, prognosis, and treatment.
Collapse
|
35
|
Hao Y, Xi J, Peng Y, Bian B, Hao G, Xi Y, Zhang Z. Circular RNA Circ_0016760 Modulates Non-Small-Cell Lung Cancer Growth Through the miR-577/ZBTB7A Axis. Cancer Manag Res 2020; 12:5561-5574. [PMID: 32753969 PMCID: PMC7354959 DOI: 10.2147/cmar.s243675] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 05/21/2020] [Indexed: 12/24/2022] Open
Abstract
Background Patients with locally advanced or metastatic non-small-cell lung cancer (NSCLC) have a poor prognosis. Circular RNA circ_0016760 (circ_0016760) is associated with the development of NSCLC. At present, the role and regulatory mechanism of circ_0016760 in NSCLC have not been well explained. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was executed to detect the expression of circ_0016760, miR-577, and Zinc finger and BTB domain containing 7A (ZBTB7A) mRNA in NSCLC tissues and cells. The colony formation, migration, invasion, and extracellular acidification rate (ECAR) of NSCLC cells were determined through colony formation, transwell, or ECAR assays. The relationship between circ_0016760 or ZBTB7A and miR-577 was analyzed via dual-luciferase reporter and RNA pull-down or RNA immunoprecipitation (RIP) assays. Protein level of ZBTB7A was evaluated with Western blot analysis. Xenograft assay was conducted to confirm the role of circ_0016760 in vivo. Results Circ_0016760 and ZBTB7A were upregulated and miR-577 was downregulated in NSCLC tissues and cells. Circ_0016760 exhaustion curbed the colony formation, migration, invasion, and ECAR of NSCLC cells in vitro and impeded tumor growth in vivo. Mechanically, circ_0016760 modulated ZBTB7A expression via sponging miR-577 in NSCLC cells. MiR-577 downregulation abolished the repressive effects of circ_0016760 silencing on colony formation, migration, invasion, and ECAR of NSCLC cells. Also, ZBTB7A upregulation overturned the repressive impacts of miR-577 elevation on colony formation, migration, invasion, and ECAR of NSCLC cells. Conclusion Circ_0016760 silencing impeded NSCLC advancement through regulation of the miR-577/ZBTB7A axis.
Collapse
Affiliation(s)
- Yanhong Hao
- Department of Cardiothoracic Surgery, The First Hospital of Yulin, Yulin City, Shanxi Province, People's Republic of China
| | - Junfeng Xi
- Department of Cardiothoracic Surgery, The First Hospital of Yulin, Yulin City, Shanxi Province, People's Republic of China
| | - Yancai Peng
- Department of Cardiothoracic Surgery, The First Hospital of Yulin, Yulin City, Shanxi Province, People's Republic of China
| | - Burong Bian
- Department of Oncology, The First Hospital of Yulin, Yulin City, Shanxi Province, People's Republic of China
| | - Guangjun Hao
- Department of Oncology, The First Hospital of Yulin, Yulin City, Shanxi Province, People's Republic of China
| | - Yunfeng Xi
- Department of Dermatological Surgery, The First Hospital of Yulin, Yulin City, Shanxi Province, People's Republic of China
| | - Zhibin Zhang
- Department of Cardiothoracic Surgery, The First Hospital of Yulin, Yulin City, Shanxi Province, People's Republic of China
| |
Collapse
|
36
|
Integrated transcriptomic and genomic analysis improves prediction of complete remission and survival in elderly patients with acute myeloid leukemia. Blood Cancer J 2020; 10:67. [PMID: 32527994 PMCID: PMC7289793 DOI: 10.1038/s41408-020-0332-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 01/18/2023] Open
Abstract
Relevant molecular tools for treatment stratification of patients ≥65 years with acute myeloid leukemia (AML) are lacking. We combined clinical data with targeted DNA- and full RNA-sequencing of 182 intensively and palliatively treated patients to predict complete remission (CR) and survival in AML patients ≥65 years. Intensively treated patients with NPM1 and IDH2R172 mutations had longer overall survival (OS), whereas mutated TP53 conferred lower CR rates and shorter OS. FLT3-ITD and TP53 mutations predicted worse OS in palliatively treated patients. Gene expression levels most predictive of CR were combined with somatic mutations for an integrated risk stratification that we externally validated using the beatAML cohort. We defined a high-risk group with a CR rate of 20% in patients with mutated TP53, compared to 97% CR in low-risk patients defined by high expression of ZBTB7A and EEPD1 without TP53 mutations. Patients without these criteria had a CR rate of 54% (intermediate risk). The difference in CR rates translated into significant OS differences that outperformed ELN stratification for OS prediction. The results suggest that an integrated molecular risk stratification can improve prediction of CR and OS and could be used to guide treatment in elderly AML patients.
Collapse
|
37
|
Yu S, Zhou C, Cao S, He J, Cai B, Wu K, Qin Y, Huang X, Xiao L, Ye J, Xu S, Xie W, Kuang J, Chu S, Guo J, Liu H, Pang W, Guo L, Zeng M, Wang X, Luo R, Li C, Zhao G, Wang B, Wu L, Chen J, Liu J, Pei D. BMP4 resets mouse epiblast stem cells to naive pluripotency through ZBTB7A/B-mediated chromatin remodelling. Nat Cell Biol 2020; 22:651-662. [PMID: 32393886 DOI: 10.1038/s41556-020-0516-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 04/03/2020] [Indexed: 01/09/2023]
Abstract
BMP4 regulates a plethora of developmental processes, including the dorsal-ventral axis and neural patterning. Here, we report that BMP4 reconfigures the nuclear architecture during the primed-to-naive transition (PNT). We first established a BMP4-driven PNT and show that BMP4 orchestrates the chromatin accessibility dynamics during PNT. Among the loci opened early by BMP4, we identified Zbtb7a and Zbtb7b (Zbtb7a/b) as targets that drive PNT. ZBTB7A/B in turn facilitate the opening of naive pluripotent chromatin loci and the activation of nearby genes. Mechanistically, ZBTB7A not only binds to chromatin loci near to the genes that are activated, but also strategically occupies those that are silenced, consistent with a role of BMP4 in both activating and suppressing gene expression during PNT at the chromatin level. Our results reveal a previously unknown function of BMP4 in regulating nuclear architecture and link its targets ZBTB7A/B to chromatin remodelling and pluripotent fate control.
Collapse
Affiliation(s)
- Shengyong Yu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Chunhua Zhou
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Shangtao Cao
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jiangping He
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Baomei Cai
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Kaixin Wu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Yue Qin
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xingnan Huang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Branch of the Supercomputing Center, Chinese Academy of Sciences, Guangzhou, China
| | - Lizhan Xiao
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Jing Ye
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Shuyang Xu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Wenxiu Xie
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Junqi Kuang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Shilong Chu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jing Guo
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - He Liu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Wei Pang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Lin Guo
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Mengying Zeng
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xiaoshan Wang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,Guangzhou Branch of the Supercomputing Center, Chinese Academy of Sciences, Guangzhou, China
| | - Rongping Luo
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Chen Li
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Guoqing Zhao
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of the Chinese Academy of Sciences, Beijing, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,Laboratory of Regenerative Biology, Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Bo Wang
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Linlin Wu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jiekai Chen
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,Guangzhou Branch of the Supercomputing Center, Chinese Academy of Sciences, Guangzhou, China.,Laboratory of Regenerative Biology, Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
| | - Jing Liu
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China. .,Guangzhou Branch of the Supercomputing Center, Chinese Academy of Sciences, Guangzhou, China. .,Laboratory of Regenerative Biology, Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China.
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Center for Cell Fate and Lineage, Division of Basic Research and International Corporation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China. .,Guangzhou Branch of the Supercomputing Center, Chinese Academy of Sciences, Guangzhou, China. .,Laboratory of Regenerative Biology, Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Lee YR, Pandolfi PP. PTEN Mouse Models of Cancer Initiation and Progression. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a037283. [PMID: 31570383 DOI: 10.1101/cshperspect.a037283] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is one of the most frequently mutated, deleted, and functionally inactivated tumor suppressor genes in human cancer. PTEN is found mutated both somatically and in the germline of patients with PTEN hamartoma tumor syndrome (PHTS). PTEN encodes a dual lipid and protein phosphatase that dephosphorylates the lipid phosphatidylinositol-3,4,5-trisphosphate (PIP3), in turn negatively regulating the oncogenic PI3K-AKT pathway, a key proto-oncogenic player in cancer development and progression. Because of importance of PTEN in tumorigenesis, a large number of sophisticated genetically engineered mouse models (GEMMs) has been designed to elucidate the underlying mechanisms by which the "PTEN pathway" promotes tumorigenesis, while simultaneously providing a well-tailored system for the identification of novel therapies and offering platforms for new drug discoveries. This review summarizes the major cancer mouse models through which the PTEN pathway has been genetically deconstructed, and outlines the rapid development of GEMMs toward more detailed functional and tissue-specific analysis.
Collapse
Affiliation(s)
- Yu-Ru Lee
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
39
|
Yeh LY, Yang CC, Wu HL, Kao SY, Liu CJ, Chen YF, Lin SC, Chang KW. The miR-372-ZBTB7A Oncogenic Axis Suppresses TRAIL-R2 Associated Drug Sensitivity in Oral Carcinoma. Front Oncol 2020; 10:47. [PMID: 32083004 PMCID: PMC7005910 DOI: 10.3389/fonc.2020.00047] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/10/2020] [Indexed: 01/31/2023] Open
Abstract
miR-372 has been shown a potent oncogenic miRNA in the pathogenesis of oral squamous cell carcinoma (OSCC). The zinc finger and BTB domain containing 7A protein (ZBTB7A) is a transcriptional regulator that is involved in a great diversity of physiological and oncogenic regulation. However, the modulation of ZBTB7A in OSCC remains unclear. Tissue analysis identifies a reverse correlation in expression between miR-372 and ZBTB7A in OSCC tumors. When OSCC cells have stable knockdown of ZBTB7A, their oncogenic potential and drug resistance is increased. By way of contrast, such an increase is attenuated by expression of ZBTB7A. Screening and validation confirms that ZBTB7A is able to modulate expression of the death receptors TRAIL-R1, TRAIL-R2, Fas and p53 phosphorylated at serine-15. In addition, ZBTB7A transactivates TRAIL-R2, which sensitizes cells to cisplatin-induced apoptosis. The ZBTB7A-TRAIL-R2 cascade is involved in both the extrinsic and intrinsic cisplatin-induced pathways of apoptosis. Database analysis indicates that the expression level of and the copy status of ZBTB7A and TRAIL-R2 are important survival predictors for head and neck cancers. Collectively, this study indicates the importance of the miR-372-ZBTB7A-TRAIL-R2 axis in mediating OSCC pathogenesis and in controlling OSCC drug resistance. Therefore, silencing miR-372 and/or upregulating ZBTB7A would seem to be promising strategies for enhancing the sensitivity of OSCC to cisplatin therapy.
Collapse
Affiliation(s)
- Li-Yin Yeh
- Department of Dentistry, School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Chieh Yang
- Department of Dentistry, School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsiao-Li Wu
- Department of Dentistry, School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Shou-Yen Kao
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chung-Ji Liu
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Fen Chen
- Department of Dentistry, School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Shu-Chun Lin
- Department of Dentistry, School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Department of Dentistry, School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
40
|
Abstract
Long non-coding RNAs (lncRNAs) are regulators of cellular machinery that are commonly dysregulated in genitourinary malignancies. Accordingly, the investigation of lncRNAs is improving our understanding of genitourinary cancers, from development to progression and dissemination. lncRNAs are involved in major oncogenic events in genitourinary malignancies, including androgen receptor (AR) signalling in prostate cancer, hypoxia-inducible factor (HIF) pathway activation in renal cell carcinoma and invasiveness in bladder cancer, as well as multiple other proliferation and survival mechanisms. In line with their putative oncogenic roles, new lncRNA-based classifications are emerging as potent predictors of prognosis. In clinical practice, detection of oncogenic lncRNAs in serum or urine might enable early cancer detection, and lncRNAs might also be promising therapeutic targets for patients with genitourinary cancer. Furthermore, as predictors of sensitivity to anticancer treatments, lncRNAs could be integrated into future precision medicine strategies. Overall, lncRNAs are promising new candidates for molecular studies and for discovery of innovative biomarkers and are putative therapeutic targets in genitourinary oncology.
Collapse
|
41
|
Zhou JP, Ren YD, Xu QY, Song Y, Zhou F, Chen MY, Liu JJ, Chen LG, Pan JS. Obesity-Induced Upregulation of ZBTB7A Promotes Lipid Accumulation through SREBP1. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4087928. [PMID: 31998789 PMCID: PMC6973189 DOI: 10.1155/2020/4087928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/07/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) is among the most common chronic liver diseases. However, the pathogenesis of NAFLD is not still unclear. This study aims at evaluating the role of zinc finger and BTB domain-containing 7A (ZBTB7A) in NAFLD. METHODS Western blotting, real-time reverse transcription PCR (RT-PCR), and immunohistochemistry were submitted to evaluate the level of ZBTB7A in the high fatty diet- (HFD-) induced NAFLD mouse model. In vitro, the expression of ZBTB7A was assessed in oleic acid- (OA-) induced HepG2 cells with western blotting and RT-PCR. The luciferase reporter assay was used to estimate the effect of ZBTB7A on the SREBP1 and NF-κB, and the ChIP assay was subjected to evaluate the direct binding to the SREBP1 promoter. Oil Red staining was used to detect lipid accumulation, and the ELISA was used to verify the levels of TG, T-CHO, and MDA. ZBTB7A was knocked down with siRNA, and RT-PCR was performed to analyze the lipogenesis-, fatty acid transporter-, and oxidation metabolism-related genes expression. The levels of ZBTB7A in primary hepatocyte, Kupffer, and hepatic stellate cells (HSCs) were tested by RT-PCR. RESULTS The upregulation of ZBTB7A expression was assessed in NAFLD mice, and ZBTB7A expression was positively correlated with TNFα, IL-6, TG, T-CHO, and MDA. ZBTB7A was highly expressed in the hepatocytes. In vitro, OA-induced ZBTB7A expression and ZBTB7A expression were closely associated with SREBP1c. ZBTB7A could activate the promoter activity of SREBP1 and activate NF-κB activity. Interestingly, the direct binding of ZBTB7A in the SREBP1 promoter was acquired in HepG2 cells. Inhibition of ZBTB7A expression could attenuate OA-induced lipid accumulation, inhibit the expression of the lipogenesis-related genes and fatty acid transporter genes, and promote the expression of oxidation metabolism-related genes. CONCLUSION ZBTB7A plays a significant role in the development process of NAFLD, and obesity-induced upregulation of ZBTB7A promotes lipid accumulation through activation of SREBP1 and NF-κB. ZBTB7A may be a potential novel target for the therapy of NAFLD.
Collapse
Affiliation(s)
- Jing-ping Zhou
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Yan-dan Ren
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Qin-yu Xu
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Yang Song
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Fei Zhou
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Mei-Ya Chen
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Jing-jing Liu
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Li-Gang Chen
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| | - Jin-Shui Pan
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361000, China
| |
Collapse
|
42
|
Zhao C, Li G, Li J. Non-coding RNAs and Cardiac Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:247-258. [PMID: 32285416 DOI: 10.1007/978-981-15-1671-9_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aging is an important risk factor for cardiovascular diseases. Aging increasing the morbidity and mortality in cardiovascular disease patients. With the society is aging rapidly in the world, medical burden of aging-related cardiovascular diseases increasing drastically. Hence, it is urgent to explore the underlying mechanism and treatment of cardiac aging. Noncoding RNAs (ncRNAs, including microRNAs, long noncoding RNAs and circular RNAs) have been reported to be involved in many pathological processes, including cell proliferation, cell death differentiation, hypertrophy and aging in wide variety of cells and tissues. In this chapter, we will summarize the physiology and molecular mechanisms of cardiac aging. Then, the recent research advances of ncRNAs in cardiac aging will be provided. The lessons learned from ncRNAs and cardiac aging studies would bring new insights into the regulatory mechanisms ncRNAs as well as treatment of aging-related cardiovascular diseases.
Collapse
Affiliation(s)
- Cuimei Zhao
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guoping Li
- Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jin Li
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.
| |
Collapse
|
43
|
Constantinou C, Spella M, Chondrou V, Patrinos GP, Papachatzopoulou A, Sgourou A. The multi-faceted functioning portrait of LRF/ZBTB7A. Hum Genomics 2019; 13:66. [PMID: 31823818 PMCID: PMC6905007 DOI: 10.1186/s40246-019-0252-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
Transcription factors (TFs) consisting of zinc fingers combined with BTB (for broad-complex, tram-track, and bric-a-brac) domain (ZBTB) are a highly conserved protein family that comprises a multifunctional and heterogeneous group of TFs, mainly modulating cell developmental events and cell fate. LRF/ZBTB7A, in particular, is reported to be implicated in a wide variety of physiological and cancer-related cell events. These physiological processes include regulation of erythrocyte maturation, B/T cell differentiation, adipogenesis, and thymic insulin expression affecting consequently insulin self-tolerance. In cancer, LRF/ZBTB7A has been reported to act either as oncogenic or as oncosuppressive factor by affecting specific cell processes (proliferation, apoptosis, invasion, migration, metastasis, etc) in opposed ways, depending on cancer type and molecular interactions. The molecular mechanisms via which LRF/ZBTB7A is known to exert either physiological or cancer-related cellular effects include chromatin organization and remodeling, regulation of the Notch signaling axis, cellular response to DNA damage stimulus, epigenetic-dependent regulation of transcription, regulation of the expression and activity of NF-κB and p53, and regulation of aerobic glycolysis and oxidative phosphorylation (Warburg effect). It is a pleiotropic TF, and thus, alterations to its expression status become detrimental for cell survival. This review summarizes its implication in different cellular activities and the commonly invoked molecular mechanisms triggered by LRF/ZBTB7A’s orchestrated action.
Collapse
Affiliation(s)
- Caterina Constantinou
- Biology laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece.,Laboratory of Pharmacology, Department of Medicine, University of Patras, Patras, Greece
| | - Magda Spella
- Biology laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece.,Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Medical Faculty, University of Patras, Patras, Greece
| | - Vasiliki Chondrou
- Biology laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece.,Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE.,Zayed Center of Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | | | - Argyro Sgourou
- Biology laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece.
| |
Collapse
|
44
|
Fan X, Bjerke GA, Riemondy K, Wang L, Yi R. A basal-enriched microRNA is required for prostate tumorigenesis in a Pten knockout mouse model. Mol Carcinog 2019; 58:2241-2253. [PMID: 31512783 PMCID: PMC7791532 DOI: 10.1002/mc.23112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) play important roles in prostate cancer development. However, it remains unclear how individual miRNAs contribute to the initiation and progression of prostate cancer. Here we show that a basal layer-enriched miRNA is required for prostate tumorigenesis. We identify miR-205 as the most highly expressed miRNA and enriched in the basal cells of the prostate. Although miR-205 is not required for normal prostate development and homeostasis, genetic deletion of miR-205 in a Pten null tumor model significantly compromises tumor progression and does not promote metastasis. In Pten null basal cells, loss of miR-205 attenuates pAkt levels and promotes cellular senescence. Furthermore, although overexpression of miR-205 in prostate cancer cells with luminal phenotypes inhibits cell growth in both human and mouse, miR-205 has a minimal effect on the growth of a normal human prostate cell line. Taken together, we have provided genetic evidence for a requirement of miR-205 in the progression of Pten null-induced prostate cancer.
Collapse
Affiliation(s)
- Xiying Fan
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado
| | - Glen A Bjerke
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado
| | - Kent Riemondy
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado
| | - Li Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado
| | - Rui Yi
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado
| |
Collapse
|
45
|
Xiao X, Shen Y, Yin L, He J, Ni X, Luo G, Chen X, Zhu W, Zhong J, Liu J, Peng X, Zu X. Knockdown of ZBTB7A inhibits cell proliferation of breast cancer through regulating the ubiquitination of estrogen receptor alpha. Life Sci 2019; 239:117042. [PMID: 31715186 DOI: 10.1016/j.lfs.2019.117042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/27/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022]
Abstract
AIMS ZBTB7A, a transcriptional repressor, accelerates the breast cancer progression. Over 70% of breast cancer samples are identified as ER-α positive. Due to the function of ZBTB7A in ER-α positive breast cancer incompletely known, we aimed to determine the role of ZBTB7A in ER-α positive cancer and explore the underlying mechanisms. MAIN METHODS In this study, the correlation between ZBTB7A and ER-α was confirmed by tissue microarray-based and TCGA database. Then, we explore if ZBTB7A maintains ER-α's level via targeting ER-α's expression or degradation. Finally, we examined the effect of ZBTB7A on the proliferation of breast cancer cells. KEY FINDINGS We further confirmed that ZBTB7A shows a significant positive correlation with ER-α in clinical breast cancer samples by tissue microarray-based analysis. Mechanically, we identified that the inhibition of ZBTB7A could upregulate E3 ligase TRIM25 leading to enhancement of ER-α ubiquitination and proteasomal degradation, which could partly explain the correlation between ZBTB7A and ER-α. Besides, we uncovered that ZBTB7A could also transcriptionally increase the expression of ER-α via indirectly binding to the region +146 to +461 bp downstream of the transcription start site of ESR1 (ERpro315) in breast cancer cells. Furthermore, ZBTB7A is found to stimulate the expression of ER-α's downstream genes, and promote the growth of estrogen receptor alpha (ER-α)-positive breast cancer cells. SIGNIFICANCE Our data revealed the novel mechanisms through which ZBTB7A manipulates ER-α level and might provide a new avenue for endocrine therapy in breast cancer.
Collapse
Affiliation(s)
- Xiao Xiao
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China; Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yingying Shen
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Liyang Yin
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Jun He
- Department of Spine Surgery, The Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan, China
| | - Xiaoyu Ni
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Gang Luo
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiguang Chen
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Wenbo Zhu
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Jing Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Jianghua Liu
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiuda Peng
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| | - Xuyu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| |
Collapse
|
46
|
Han D, Chen S, Han W, Gao S, Owiredu JN, Li M, Balk SP, He HH, Cai C. ZBTB7A Mediates the Transcriptional Repression Activity of the Androgen Receptor in Prostate Cancer. Cancer Res 2019; 79:5260-5271. [PMID: 31444154 PMCID: PMC6801099 DOI: 10.1158/0008-5472.can-19-0815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/09/2019] [Accepted: 08/20/2019] [Indexed: 01/15/2023]
Abstract
Loss of expression of context-specific tumor suppressors is a critical event that facilitates the development of prostate cancer. Zinc finger and BTB domain containing transcriptional repressors, such as ZBTB7A and ZBTB16, have been recently identified as tumor suppressors that play important roles in preventing prostate cancer progression. In this study, we used combined ChIP-seq and RNA-seq analyses of prostate cancer cells to identify direct ZBTB7A-repressed genes, which are enriched for transcriptional targets of E2F, and identified that the androgen receptor (AR) played a critical role in the transcriptional suppression of these E2F targets. AR recruitment of the retinoblastoma protein (Rb) was required to strengthen the E2F-Rb transcriptional repression complex. In addition, ZBTB7A was rapidly recruited to the E2F-Rb binding sites by AR and negatively regulated the transcriptional activity of E2F1 on DNA replication genes. Finally, ZBTB7A suppressed the growth of castration-resistant prostate cancer (CRPC) in vitro and in vivo, and overexpression of ZBTB7A acted in synergy with high-dose testosterone treatment to effectively prevent the recurrence of CRPC. Overall, this study provides novel molecular insights of the role of ZBTB7A in CRPC cells and demonstrates globally its critical role in mediating the transcriptional repression activity of AR. SIGNIFICANCE: ZBTB7A is recruited to the E2F-Rb binding sites by AR and negatively regulates the transcriptional activity of E2F1 on DNA replication genes.
Collapse
Affiliation(s)
- Dong Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Sujun Chen
- Princess Margaret Cancer Center/University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Wanting Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Shuai Gao
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Jude N Owiredu
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Muqing Li
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
| | - Steven P Balk
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Housheng Hansen He
- Princess Margaret Cancer Center/University Health Network, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts.
| |
Collapse
|
47
|
Yu C, Hu K, Nguyen D, Wang ZA. From genomics to functions: preclinical mouse models for understanding oncogenic pathways in prostate cancer. Am J Cancer Res 2019; 9:2079-2102. [PMID: 31720076 PMCID: PMC6834478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/10/2019] [Indexed: 06/10/2023] Open
Abstract
Next-generation sequencing has revealed numerous genomic alterations that induce aberrant signaling activities in prostate cancer (PCa). Among them are pathways affecting multiple cancer types, including the PI3K/AKT/mTOR, p53, Rb, Ras/Raf/MAPK, Myc, FGF, and Wnt signaling pathways, as well as ones that are prominent in PCa, including alterations in genes of AR signaling, the ETS family, NKX3.1, and SPOP. Cross talk among the oncogenic pathways can confer PCa resistance to therapy, particularly in advanced tumors, which are castration-resistant or show neuroendocrine features. Various experimental models, such as cancer cell lines, animal models, and patient-derived xenografts and organoids have been utilized to dissect PCa progression mechanisms. Here, we review the current preclinical mouse models for studying the most commonly altered pathways in PCa, with an emphasis on their interplays. We highlight the power of genetically engineered mouse models (GEMMs) in translating genomic discoveries into understanding of the functions of these oncogenic events in vivo. Developing and analyzing PCa mouse models will undoubtedly continue to offer new insights into tumor biology and guide novel rationalized therapy.
Collapse
Affiliation(s)
- Chuan Yu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Kevin Hu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Daniel Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| |
Collapse
|
48
|
Molloy ME, Lewinska M, Williamson AK, Nguyen TT, Kuser-Abali G, Gong L, Yan J, Little JB, Pandolfi PP, Yuan ZM. ZBTB7A governs estrogen receptor alpha expression in breast cancer. J Mol Cell Biol 2019; 10:273-284. [PMID: 30265334 DOI: 10.1093/jmcb/mjy020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/19/2018] [Indexed: 01/30/2023] Open
Abstract
ZBTB7A, a member of the POZ/BTB and Krüppel (POK) family of transcription factors, has been shown to have a context-dependent role in cancer development and progression. The role of ZBTB7A in estrogen receptor alpha (ERα)-positive breast cancer is largely unknown. Approximately 70% of breast cancers are classified as ERα-positive. ERα carries out the biological effects of estrogen and its expression level dictates response to endocrine therapies and prognosis for breast cancer patients. In this study, we find that ZBTB7A transcriptionally regulates ERα expression in ERα-positive breast cancer cell lines by binding to the ESR1 promoter leading to increased transcription of ERα. Inhibition of ZBTB7A in ERα-positive cells results in decreased estrogen responsiveness as demonstrated by diminished estrogen-response element-driven luciferase reporter activity, induction of estrogen target genes, and estrogen-stimulated growth. We also report that ERα potentiates ZBTB7A expression via a post-translational mechanism, suggesting the presence of a positive feedback loop between ZBTB7A and ERα, conferring sensitivity to estrogen in breast cancer. Clinically, we find that ZBTB7A and ERα are often co-expressed in breast cancers and that high ZBTB7A expression correlates with improved overall and relapse-free survival for breast cancer patients. Importantly, high ZBTB7A expression predicts a more favorable outcome for patients treated with endocrine therapies. Together, these findings demonstrate that ZBTB7A contributes to the transcriptional program maintaining ERα expression and potentially an endocrine therapy-responsive phenotype in breast cancer.
Collapse
Affiliation(s)
- Mary Ellen Molloy
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Monika Lewinska
- Biotech Research and Innovation Centre, Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amanda K Williamson
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Thanh Thao Nguyen
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gamze Kuser-Abali
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Gong
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jiawei Yan
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - John B Little
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Pier Paolo Pandolfi
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhi-Min Yuan
- Department of Environmental Health, John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
49
|
Guarnerio J, Zhang Y, Cheloni G, Panella R, Mae Katon J, Simpson M, Matsumoto A, Papa A, Loretelli C, Petri A, Kauppinen S, Garbutt C, Nielsen GP, Deshpande V, Castillo-Martin M, Cordon-Cardo C, Dimitrios S, Clohessy JG, Batish M, Pandolfi PP. Intragenic antagonistic roles of protein and circRNA in tumorigenesis. Cell Res 2019; 29:628-640. [PMID: 31209250 PMCID: PMC6796857 DOI: 10.1038/s41422-019-0192-1] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 05/23/2019] [Indexed: 01/28/2023] Open
Abstract
circRNAs arise from back splicing events during mRNA processing, and when deregulated can play an active role in cancer. Here we characterize a new circRNA (circPOK) encoded by the Zbtb7a gene (also kown as POKEMON, LRF) in the context of mesenchymal tumor progression. circPOK functions as a non-coding proto-oncogenic RNA independently and antithetically to its linear transcript counterpart, which acts as a tumor suppressor by encoding the Pokemon transcription factor. We find that circPOK regulates pro-proliferative and pro-angiogenic factors by co-activation of the ILF2/3 complex. Importantly, the expression of Pokemon protein and circRNA is aberrantly uncoupled in cancer through differential post-transcriptional regulation. Thus, we identify a novel type of genetic unit, the iRegulon, that yields biochemically distinct RNA products, circular and linear, with diverse and antithetical functions. Our findings further expand the cellular repertoire towards the control of normal biological outputs, while aberrant expression of such components may underlie disease pathogenesis including cancer.
Collapse
Affiliation(s)
- Jlenia Guarnerio
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
- Cedars-Sinai Medical Center, Department of Radiation Oncology, Samuel Oschin Comprehensive Cancer Center, Los Angeles, CA, 90048, USA
| | - Yang Zhang
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Giulia Cheloni
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Riccardo Panella
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jesse Mae Katon
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Mark Simpson
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, Newark, NJ, 07103, USA
| | - Akinobu Matsumoto
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Antonella Papa
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Cristian Loretelli
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Andreas Petri
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark; Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Sakari Kauppinen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark; Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Cassandra Garbutt
- MGH Center for Sarcoma and Connective Tissue Oncology, Department of Orthopedic Surgery, New York, USA
| | | | - Vikram Deshpande
- MGH Center for Sarcoma and Connective Tissue Oncology, Department of Pathology, New York, USA
| | - Mireia Castillo-Martin
- Department of Pathology, Mount Sinai School of Medicine, The Mount Sinai Medical Center, New York, NY, 10029, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Mount Sinai School of Medicine, The Mount Sinai Medical Center, New York, NY, 10029, USA
| | - Spentzos Dimitrios
- MGH Center for Sarcoma and Connective Tissue Oncology, Department of Orthopedic Surgery, New York, USA
| | - John G Clohessy
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Mona Batish
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, Newark, NJ, 07103, USA
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
50
|
Testa U, Castelli G, Pelosi E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E82. [PMID: 31366128 PMCID: PMC6789661 DOI: 10.3390/medicines6030082] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent nonskin cancer and second most common cause of cancer-related deaths in man. Prostate cancer is a clinically heterogeneous disease with many patients exhibiting an aggressive disease with progression, metastasis, and other patients showing an indolent disease with low tendency to progression. Three stages of development of human prostate tumors have been identified: intraepithelial neoplasia, adenocarcinoma androgen-dependent, and adenocarcinoma androgen-independent or castration-resistant. Advances in molecular technologies have provided a very rapid progress in our understanding of the genomic events responsible for the initial development and progression of prostate cancer. These studies have shown that prostate cancer genome displays a relatively low mutation rate compared with other cancers and few chromosomal loss or gains. The ensemble of these molecular studies has led to suggest the existence of two main molecular groups of prostate cancers: one characterized by the presence of ERG rearrangements (~50% of prostate cancers harbor recurrent gene fusions involving ETS transcription factors, fusing the 5' untranslated region of the androgen-regulated gene TMPRSS2 to nearly the coding sequence of the ETS family transcription factor ERG) and features of chemoplexy (complex gene rearrangements developing from a coordinated and simultaneous molecular event), and a second one characterized by the absence of ERG rearrangements and by the frequent mutations in the E3 ubiquitin ligase adapter SPOP and/or deletion of CDH1, a chromatin remodeling factor, and interchromosomal rearrangements and SPOP mutations are early events during prostate cancer development. During disease progression, genomic and epigenomic abnormalities accrued and converged on prostate cancer pathways, leading to a highly heterogeneous transcriptomic landscape, characterized by a hyperactive androgen receptor signaling axis.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|