1
|
Mohanty S, Lekven AC. Divergent functions of the evolutionarily conserved, yet seemingly dispensable, Wnt target, sp5. Differentiation 2025; 141:100829. [PMID: 39675112 DOI: 10.1016/j.diff.2024.100829] [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: 08/05/2024] [Revised: 11/10/2024] [Accepted: 12/04/2024] [Indexed: 12/17/2024]
Abstract
The activation of sp5 in response to Wnt/β-catenin signaling is observed in many species during axis patterning, neural crest induction, maintenance and differentiation of stem cells. Indeed, the conserved response of sp5 orthologs to Wnt-mediated activation is the basis for this gene commonly being used as a readout for Wnt signaling activity. However, several seemingly conflicting findings regarding the function of sp5 in the context of Wnt signaling cast this gene in an enigmatic light. In this review, we examine current knowledge of sp5 structure and function, its relationship to Wnt signaling in varied contexts, and present perspectives on how progress on this interesting gene can move forward.
Collapse
Affiliation(s)
- Saurav Mohanty
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA
| | - Arne C Lekven
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204-5001, USA.
| |
Collapse
|
2
|
Sun X, Zhou Q, Xiao C, Mao C, Liu Y, Chen G, Song Y. Role of post-translational modifications of Sp1 in cardiovascular diseases. Front Cell Dev Biol 2024; 12:1453901. [PMID: 39252788 PMCID: PMC11381397 DOI: 10.3389/fcell.2024.1453901] [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: 06/24/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Specific protein 1 (Sp1) is pivotal in sustaining baseline transcription as well as modulating cell signaling pathways and transcription factors activity. Through interactions with various proteins, especially transcription factors, Sp1 controls the expression of target genes, influencing numerous biological processes. Numerous studies have confirmed Sp1's significant regulatory role in the pathogenesis of cardiovascular disorders. Post-translational modifications (PTMs) of Sp1, such as phosphorylation, ubiquitination, acetylation, glycosylation, SUMOylation, and S-sulfhydration, can enhance or modify its transcriptional activity and DNA-binding stability. These modifications also regulate Sp1 expression across different cell types. Sp1 is crucial in regulating non-coding gene expression and the activity of proteins in response to pathophysiological stimuli. Understanding Sp1 PTMs advances our knowledge of cell signaling pathways in controlling Sp1 stability during cardiovascular disease onset and progression. It also aids in identifying novel pharmaceutical targets and biomarkers essential for preventing and managing cardiovascular diseases.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Synopsis of the Golden Chamber, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Zhou
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengpu Xiao
- Department of Typhoid, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Caiyun Mao
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Liu
- The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Guozhen Chen
- Department of Pediatrics, Yantai Yuhuangding Hospital, Shandong, China
| | - Yunjia Song
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
3
|
Sun X, Xiao C, Wang X, Wu S, Yang Z, Sui B, Song Y. Role of post-translational modifications of Sp1 in cancer: state of the art. Front Cell Dev Biol 2024; 12:1412461. [PMID: 39228402 PMCID: PMC11368732 DOI: 10.3389/fcell.2024.1412461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/16/2024] [Indexed: 09/05/2024] Open
Abstract
Specific protein 1 (Sp1) is central to regulating transcription factor activity and cell signaling pathways. Sp1 is highly associated with the poor prognosis of various cancers; it is considered a non-oncogene addiction gene. The function of Sp1 is complex and contributes to regulating extensive transcriptional activity, apart from maintaining basal transcription. Sp1 activity and stability are affected by post-translational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, glycosylation, and SUMOylation. These modifications help to determine genetic programs that alter the Sp1 structure in different cells and increase or decrease its transcriptional activity and DNA binding stability in response to pathophysiological stimuli. Investigating the PTMs of Sp1 will contribute to a deeper understanding of the mechanism underlying the cell signaling pathway regulating Sp1 stability and the regulatory mechanism by which Sp1 affects cancer progression. Furthermore, it will facilitate the development of new drug targets and biomarkers, thereby elucidating considerable implications in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Typhoid, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengpu Xiao
- Department of Chinese Internal Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyang Wang
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyu Wu
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhendong Yang
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bowen Sui
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yunjia Song
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Sun L, He X, Kong J, Yu H, Wang Y. Menstrual blood-derived stem cells exosomal miR-let-7 to ameliorate pulmonary fibrosis through inhibiting ferroptosis by Sp3/HDAC2/Nrf2 signaling pathway. Int Immunopharmacol 2024; 126:111316. [PMID: 38056200 DOI: 10.1016/j.intimp.2023.111316] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a serious, lifelong lung disease with high morbidity and high mortality. Menstrual blood-derived stem cells (MenSCs) derived exosomes (MenSCs-Exo) emerge as an attractive tool for the treatment of acute lung injury and fibrosis-related diseases. However, more comprehensive mechanism over how MenSCs derived exosomes exhibits anti-pulmonary fibrosis needs to be elucidated. In this study, TGF-β was used to construct cell fibrosis model, and bleomycin (BLM) was applied to induce lung tissue fibrosis mice model. BLM- and TGF-β1-induced cellular reactive oxygen species (ROS), mitochondrial DNA (mtDNA) damage, and lung epithelial cell apoptosis were alleviated by MenSCs-Exo treatment in vivo and in vitro. Besides, it was found that MenSCs-Exo delivered miR-let-7 into MLE-12 cells/lung epithelial cell and the reduction of miR-let-7 blocked the improvement produced by MenSCs-Exo. Mechanistically, miR-let-7 directly bound to Sp3 and negatively regulated its expression. Sp3 elevation promoted the expression of ferroptosis-related protein and mitochondrial DNA (mtDNA) damage markers via recruiting HDAC2, thereby inactivating keap1/Nrf2 signal cascade, which were confirmed in BLM-induced pulmonary fibrosis mice model under the combination therapy of the MenSCs-Exo and let-7 inhibitor. Collectively, MenSCs derived exosomes could transmit miR-let-7 into MLE-12 cells to inhibit the expression of Sp3, thereby weakening the recruitment effect of Sp3 on HDAC2, lifting the deacetylation restriction of HDAC2 on Nrf2, and enhancing the Nrf2 pathway. These changes further declined ferroptosis and delayed the pathological process of oxidative damage and lung epithelial cell apoptosis in PF.
Collapse
Affiliation(s)
- Lifang Sun
- Department of Tuberculosis, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou 310003, PR China; Department of Tuberculosis, Hangzhou Red Cross Hospital, Hangzhou 310003, PR China
| | - Xinxin He
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 311399, PR China
| | - Jiao Kong
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Huan Yu
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, PR China
| | - Yunguang Wang
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang 310000, PR China.
| |
Collapse
|
5
|
Huang YR, Xie XX, Yang J, Sun XY, Niu XY, Yang CG, Li LJ, Zhang L, Wang D, Liu CY, Hou SJ, Jiang CY, Xu YM, Liu RT. ArhGAP11A mediates amyloid-β generation and neuropathology in an Alzheimer's disease-like mouse model. Cell Rep 2023; 42:112624. [PMID: 37302068 DOI: 10.1016/j.celrep.2023.112624] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Amyloid-β (Aβ) plays an important role in the neuropathology of Alzheimer's disease (AD), but some factors promoting Aβ generation and Aβ oligomer (Aβo) neurotoxicity remain unclear. We here find that the levels of ArhGAP11A, a Ras homology GTPase-activating protein, significantly increase in patients with AD and amyloid precursor protein (APP)/presenilin-1 (PS1) mice. Reducing the ArhGAP11A level in neurons not only inhibits Aβ generation by decreasing the expression of APP, PS1, and β-secretase (BACE1) through the RhoA/ROCK/Erk signaling pathway but also reduces Aβo neurotoxicity by decreasing the expressions of apoptosis-related p53 target genes. In APP/PS1 mice, specific reduction of the ArhGAP11A level in neurons significantly reduces Aβ production and plaque deposition and ameliorates neuronal damage, neuroinflammation, and cognitive deficits. Moreover, Aβos enhance ArhGAP11A expression in neurons by activating E2F1, which thus forms a deleterious cycle. Our results demonstrate that ArhGAP11A may be involved in AD pathogenesis and that decreasing ArhGAP11A expression may be a promising therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Ya-Ru Huang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi-Xiu Xie
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Yang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Henan Medical Key Laboratory of Neurogenetic and Neurodegenerative Disease, Zhengzhou 450052, Henan, China
| | - Xiao-Ying Sun
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Yun Niu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Ningxia University, Yinchuan 750021, Ningxia, China
| | - Cheng-Gang Yang
- Department of BigData, Beijing Medintell Bioinformatic Technology Co., Ltd., Beijing 100081, China; Department of Research and Development, Gu'an Bojian Bio-Technology Co., Ltd., Langfang 065000, Hebei, China
| | - Ling-Jie Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lun Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Dan Wang
- Department of BigData, Beijing Medintell Bioinformatic Technology Co., Ltd., Beijing 100081, China
| | - Chun-Yu Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Shandong Agricultural University, Tai'an 271000, Shandong, China
| | - Sheng-Jie Hou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen-Yang Jiang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yu-Ming Xu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Key Laboratory of Cerebrovascular Disease of Henan Province, Zhengzhou 450052, Henan, China.
| | - Rui-Tian Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
6
|
McAllister JJ, Dahiya S, Berman R, Collins M, Nonnemacher MR, Burdo TH, Wigdahl B. Altered recruitment of Sp isoforms to HIV-1 long terminal repeat between differentiated monoblastic cell lines and primary monocyte-derived macrophages. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2022.971293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) transcription in cells of the monocyte-macrophage lineage is regulated by interactions between the HIV-1 long terminal repeat (LTR) and a variety of host cell and viral proteins. Binding of the Sp family of transcription factors (TFs) to the G/C box array of the LTR governs both basal as well as activated LTR-directed transcriptional activity. The effect of monocytic differentiation on Sp factor binding and transactivation was examined with respect to the HIV-1 LTR. The binding of Sp1, full-length Sp3 and truncated Sp3 to a high affinity HIV-1 Sp element was specifically investigated and results showed that Sp1 binding increased relative to the binding of the sum of full-length and truncated Sp3 binding following chemically-induced monocytic differentiation in monoblastic (U-937, THP-1) and myelomonocytic (HL-60) cells. In addition, Sp binding ratios from PMA-induced cell lines were shown to more closely approximate those derived from primary monocyte-derived macrophages (MDMs) than did ratios derived from uninduced cell lines. The altered Sp binding phenotype associated with changes in the transcriptional activation mediated by the HIV-1 G/C box array. Additionally, analysis of post-translational modifications on Sp1 and Sp3 revealed a loss of phosphorylation on serine and threonine residues with chemically-induced differentiation indicating that the activity of Sp factors is additionally regulated at the level of post-translational modifications (PTMs).
Collapse
|
7
|
Regulation of Viral Restriction by Post-Translational Modifications. Viruses 2021; 13:v13112197. [PMID: 34835003 PMCID: PMC8618861 DOI: 10.3390/v13112197] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/17/2022] Open
Abstract
Intrinsic immunity is orchestrated by a wide range of host cellular proteins called restriction factors. They have the capacity to interfere with viral replication, and most of them are tightly regulated by interferons (IFNs). In addition, their regulation through post-translational modifications (PTMs) constitutes a major mechanism to shape their action positively or negatively. Following viral infection, restriction factor modification can be decisive. Palmitoylation of IFITM3, SUMOylation of MxA, SAMHD1 and TRIM5α or glycosylation of BST2 are some of those PTMs required for their antiviral activity. Nonetheless, for their benefit and by manipulating the PTMs machinery, viruses have evolved sophisticated mechanisms to counteract restriction factors. Indeed, many viral proteins evade restriction activity by inducing their ubiquitination and subsequent degradation. Studies on PTMs and their substrates are essential for the understanding of the antiviral defense mechanisms and provide a global vision of all possible regulations of the immune response at a given time and under specific infection conditions. Our aim was to provide an overview of current knowledge regarding the role of PTMs on restriction factors with an emphasis on their impact on viral replication.
Collapse
|
8
|
Xiu T, Guo Q, Jing FB. Facing Cell Autophagy in Gastric Cancer - What Do We Know so Far? Int J Gen Med 2021; 14:1647-1659. [PMID: 33976565 PMCID: PMC8104978 DOI: 10.2147/ijgm.s298705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/12/2021] [Indexed: 01/17/2023] Open
Abstract
Autophagy is a process by which misfolded proteins and damaged organelles in the lysosomes of tumor cells were degraded reusing decomposed substances and avoiding accumulation of large amounts of harmful substances. Here, the role of autophagy in the development of malignant transformation of gastric tumors, and the underlying mechanisms involved in autophagy formation, and the application of targeted autophagy in the treatment of gastric cancer were summarized.
Collapse
Affiliation(s)
- Ting Xiu
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, People's Republic of China.,Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, 266021, People's Republic of China
| | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, People's Republic of China
| | - Fan-Bo Jing
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, People's Republic of China
| |
Collapse
|
9
|
Sohrabi SS, Sohrabi SM, Rashidipour M, Mohammadi M, Khalili Fard J, Mirzaei Najafgholi H. Identification of common key regulators in rat hepatocyte cell lines under exposure of different pesticides. Gene 2020; 739:144508. [DOI: 10.1016/j.gene.2020.144508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/15/2019] [Accepted: 02/21/2020] [Indexed: 12/15/2022]
|
10
|
Feng Z, Ma J, Hua X. Epigenetic regulation by the menin pathway. Endocr Relat Cancer 2017; 24:T147-T159. [PMID: 28811300 PMCID: PMC5612327 DOI: 10.1530/erc-17-0298] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
There is a trend of increasing prevalence of neuroendocrine tumors (NETs), and the inherited multiple endocrine neoplasia type 1 (MEN1) syndrome serves as a genetic model to investigate how NETs develop and the underlying mechanisms. Menin, encoded by the MEN1 gene, at least partly acts as a scaffold protein by interacting with multiple partners to regulate cellular homeostasis of various endocrine organs. Menin has multiple functions including regulation of several important signaling pathways by controlling gene transcription. Here, we focus on reviewing the recent progress in elucidating the key biochemical role of menin in epigenetic regulation of gene transcription and cell signaling, as well as posttranslational regulation of menin itself. In particular, we will review the progress in studying structural and functional interactions of menin with various histone modifiers and transcription factors such as MLL, PRMT5, SUV39H1 and other transcription factors including c-Myb and JunD. Moreover, the role of menin in regulating cell signaling pathways such as TGF-beta, Wnt and Hedgehog, as well as miRNA biogenesis and processing will be described. Further, the regulation of the MEN1 gene transcription, posttranslational modifications and stability of menin protein will be reviewed. These various modes of regulation by menin as well as regulation of menin by various biological factors broaden the view regarding how menin controls various biological processes in neuroendocrine organ homeostasis.
Collapse
Affiliation(s)
- Zijie Feng
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jian Ma
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- State Key Laboratory of Veterinary BiotechnologyHarbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xianxin Hua
- Department of Cancer BiologyAbramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Chüeh AC, Tse JWT, Dickinson M, Ioannidis P, Jenkins L, Togel L, Tan B, Luk I, Davalos-Salas M, Nightingale R, Thompson MR, Williams BRG, Lessene G, Lee EF, Fairlie WD, Dhillon AS, Mariadason JM. ATF3 Repression of BCL-X L Determines Apoptotic Sensitivity to HDAC Inhibitors across Tumor Types. Clin Cancer Res 2017; 23:5573-5584. [PMID: 28611196 PMCID: PMC5600837 DOI: 10.1158/1078-0432.ccr-17-0466] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/27/2017] [Accepted: 06/09/2017] [Indexed: 12/13/2022]
Abstract
Purpose: Histone deacetylase inhibitors (HDACi) are epigenome-targeting small molecules approved for the treatment of cutaneous T-cell lymphoma and multiple myeloma. They have also demonstrated clinical activity in acute myelogenous leukemia, non-small cell lung cancer, and estrogen receptor-positive breast cancer, and trials are underway assessing their activity in combination regimens including immunotherapy. However, there is currently no clear strategy to reliably predict HDACi sensitivity. In colon cancer cells, apoptotic sensitivity to HDACi is associated with transcriptional induction of multiple immediate-early (IE) genes. Here, we examined whether this transcriptional response predicts HDACi sensitivity across tumor type and investigated the mechanism by which it triggers apoptosis.Experimental Design: Fifty cancer cell lines from diverse tumor types were screened to establish the correlation between apoptotic sensitivity, induction of IE genes, and components of the intrinsic apoptotic pathway.Results: We show that sensitivity to HDACi across tumor types is predicted by induction of the IE genes FOS, JUN, and ATF3, but that only ATF3 is required for HDACi-induced apoptosis. We further demonstrate that the proapoptotic function of ATF3 is mediated through direct transcriptional repression of the prosurvival factor BCL-XL (BCL2L1) These findings provided the rationale for dual inhibition of HDAC and BCL-XL, which we show strongly cooperate to overcome inherent resistance to HDACi across diverse tumor cell types.Conclusions: These findings explain the heterogeneous responses of tumor cells to HDACi-induced apoptosis and suggest a framework for predicting response and expanding their therapeutic use in multiple cancer types. Clin Cancer Res; 23(18); 5573-84. ©2017 AACR.
Collapse
Affiliation(s)
| | - Janson W T Tse
- Ludwig Institute for Cancer Research, Melbourne, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | | | - Paul Ioannidis
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Laura Jenkins
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Lars Togel
- Ludwig Institute for Cancer Research, Melbourne, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - BeeShin Tan
- Ludwig Institute for Cancer Research, Melbourne, Australia
| | - Ian Luk
- Ludwig Institute for Cancer Research, Melbourne, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Mercedes Davalos-Salas
- Ludwig Institute for Cancer Research, Melbourne, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Rebecca Nightingale
- Ludwig Institute for Cancer Research, Melbourne, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Matthew R Thompson
- Hudson Institute of Medical Research and Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria
| | - Bryan R G Williams
- Hudson Institute of Medical Research and Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria
| | | | - Erinna F Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, Victoria, Australia
| | - Walter D Fairlie
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, Victoria, Australia
| | - Amardeep S Dhillon
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - John M Mariadason
- Ludwig Institute for Cancer Research, Melbourne, Australia.
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
12
|
Salazar VS, Ohte S, Capelo LP, Gamer L, Rosen V. Specification of osteoblast cell fate by canonical Wnt signaling requires Bmp2. Development 2016; 143:4352-4367. [PMID: 27802170 DOI: 10.1242/dev.136879] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 10/07/2016] [Indexed: 12/14/2022]
Abstract
Enhanced BMP or canonical Wnt (cWnt) signaling are therapeutic strategies employed to enhance bone formation and fracture repair, but the mechanisms each pathway utilizes to specify cell fate of bone-forming osteoblasts remain poorly understood. Among all BMPs expressed in bone, we find that singular deficiency of Bmp2 blocks the ability of cWnt signaling to specify osteoblasts from limb bud or bone marrow progenitors. When exposed to cWnts, Bmp2-deficient cells fail to progress through the Runx2/Osx1 checkpoint and thus do not upregulate multiple genes controlling mineral metabolism in osteoblasts. Cells lacking Bmp2 after induction of Osx1 differentiate normally in response to cWnts, suggesting that pre-Osx1+ osteoprogenitors are an essential source and a target of BMP2. Our analysis furthermore reveals Grainyhead-like 3 (Grhl3) as a transcription factor in the osteoblast gene regulatory network induced during bone development and bone repair, which acts upstream of Osx1 in a BMP2-dependent manner. The Runx2/Osx1 transition therefore receives crucial regulatory inputs from BMP2 that are not compensated for by cWnt signaling, and this is mediated at least in part by induction and activation of Grhl3.
Collapse
Affiliation(s)
- Valerie S Salazar
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| | - Satoshi Ohte
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA.,Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Luciane P Capelo
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA.,Instituto de Ciência e Tecnologia, Universidade Federal de São Paulo, Rua Talim, 330, São José dos Campos, São Paulo, CEP 12231-280, Brazil
| | - Laura Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
13
|
Chueh AC, Tse JWT, Tögel L, Mariadason JM. Mechanisms of Histone Deacetylase Inhibitor-Regulated Gene Expression in Cancer Cells. Antioxid Redox Signal 2015; 23:66-84. [PMID: 24512308 PMCID: PMC4492771 DOI: 10.1089/ars.2014.5863] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Class I and II histone deacetylase inhibitors (HDACis) are approved for the treatment of cutaneous T-cell lymphoma and are undergoing clinical trials as single agents, and in combination, for other hematological and solid tumors. Understanding their mechanisms of action is essential for their more effective clinical use, and broadening their clinical potential. RECENT ADVANCES HDACi induce extensive transcriptional changes in tumor cells by activating and repressing similar numbers of genes. These transcriptional changes mediate, at least in part, HDACi-mediated growth inhibition, apoptosis, and differentiation. Here, we highlight two fundamental mechanisms by which HDACi regulate gene expression—histone and transcription factor acetylation. We also review the transcriptional responses invoked by HDACi, and compare these effects within and across tumor types. CRITICAL ISSUES The mechanistic basis for how HDACi activate, and in particular repress gene expression, is not well understood. In addition, whether subsets of genes are reproducibly regulated by these agents both within and across tumor types has not been systematically addressed. A detailed understanding of the transcriptional changes elicited by HDACi in various tumor types, and the mechanistic basis for these effects, may provide insights into the specificity of these drugs for transformed cells and specific tumor types. FUTURE DIRECTIONS Understanding the mechanisms by which HDACi regulate gene expression and an appreciation of their transcriptional targets could facilitate the ongoing clinical development of these emerging therapeutics. In particular, this knowledge could inform the design of rational drug combinations involving HDACi, and facilitate the identification of mechanism-based biomarkers of response.
Collapse
Affiliation(s)
- Anderly C Chueh
- Ludwig Institute for Cancer Research , Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne, Australia
| | - Janson W T Tse
- Ludwig Institute for Cancer Research , Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne, Australia
| | - Lars Tögel
- Ludwig Institute for Cancer Research , Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne, Australia
| | - John M Mariadason
- Ludwig Institute for Cancer Research , Olivia Newton John Cancer and Wellness Centre, Austin Health, Melbourne, Australia
| |
Collapse
|
14
|
Haery L, Thompson RC, Gilmore TD. Histone acetyltransferases and histone deacetylases in B- and T-cell development, physiology and malignancy. Genes Cancer 2015; 6:184-213. [PMID: 26124919 PMCID: PMC4482241 DOI: 10.18632/genesandcancer.65] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 05/12/2015] [Indexed: 12/31/2022] Open
Abstract
The development of B and T cells from hematopoietic precursors and the regulation of the functions of these immune cells are complex processes that involve highly regulated signaling pathways and transcriptional control. The signaling pathways and gene expression patterns that give rise to these developmental processes are coordinated, in part, by two opposing classes of broad-based enzymatic regulators: histone acetyltransferases (HATs) and histone deacetylases (HDACs). HATs and HDACs can modulate gene transcription by altering histone acetylation to modify chromatin structure, and by regulating the activity of non-histone substrates, including an array of immune-cell transcription factors. In addition to their role in normal B and T cells, dysregulation of HAT and HDAC activity is associated with a variety of B- and T-cell malignancies. In this review, we describe the roles of HATs and HDACs in normal B- and T-cell physiology, describe mutations and dysregulation of HATs and HDACs that are implicated lymphoma and leukemia, and discuss HAT and HDAC inhibitors that have been explored as treatment options for leukemias and lymphomas.
Collapse
Affiliation(s)
- Leila Haery
- Department of Biology, Boston University, Boston, MA, USA
| | | | | |
Collapse
|
15
|
Tang X, Fu X, Hao B, Zhu F, Xiao S, Xu L, Shen Z. Identification of sumoylated proteins in the silkworm Bombyx mori. Int J Mol Sci 2014; 15:22011-27. [PMID: 25470021 PMCID: PMC4284691 DOI: 10.3390/ijms151222011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 01/20/2023] Open
Abstract
Small ubiquitin-like modifier (SUMO) modification (SUMOylation) is an important and widely used reversible modification system in eukaryotic cells. It regulates various cell processes, including protein targeting, transcriptional regulation, signal transduction, and cell division. To understand its role in the model lepidoptera insect Bombyx mori, a recombinant baculovirus was constructed to express an enhanced green fluorescent protein (eGFP)-SUMO fusion protein along with ubiquitin carrier protein 9 of Bombyx mori (BmUBC9). SUMOylation substrates from Bombyx mori cells infected with this baculovirus were isolated by immunoprecipitation and identified by LC-ESI-MS/MS. A total of 68 candidate SUMOylated proteins were identified, of which 59 proteins were functionally categorized to gene ontology (GO) terms. Analysis of kyoto encyclopedia of genes and genomes (KEGG) pathways showed that 46 of the identified proteins were involved in 76 pathways that mainly play a role in metabolism, spliceosome and ribosome functions, and in RNA transport. Furthermore, SUMOylation of four candidates (polyubiquitin-C-like isoform X1, 3-hydroxyacyl-CoA dehydrogenase, cyclin-related protein FAM58A-like and GTP-binding nuclear protein Ran) were verified by co-immunoprecipitation in Drosophila schneide 2 cells. In addition, 74% of the identified proteins were predicted to have at least one SUMOylation site. The data presented here shed light on the crucial process of protein sumoylation in Bombyx mori.
Collapse
Affiliation(s)
- Xudong Tang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China.
| | - Xuliang Fu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China.
| | - Bifang Hao
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China.
| | - Feng Zhu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China.
| | - Shengyan Xiao
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China.
| | - Li Xu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China.
| | - Zhongyuan Shen
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China.
| |
Collapse
|
16
|
Ethanol-induced transcriptional activation of programmed cell death 4 (Pdcd4) is mediated by GSK-3β signaling in rat cortical neuroblasts. PLoS One 2014; 9:e98080. [PMID: 24837604 PMCID: PMC4024002 DOI: 10.1371/journal.pone.0098080] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/28/2014] [Indexed: 01/20/2023] Open
Abstract
Ingestion of ethanol (ETOH) during pregnancy induces grave abnormalities in developing fetal brain. We have previously reported that ETOH induces programmed cell death 4 (PDCD4), a critical regulator of cell growth, in cultured fetal cerebral cortical neurons (PCNs) and in the cerebral cortex in vivo and affect protein synthesis as observed in Fetal Alcohol Spectrum Disorder (FASD). However, the mechanism which activates PDCD4 in neuronal systems is unclear and understanding this regulation may provide a counteractive strategy to correct the protein synthesis associated developmental changes seen in FASD. The present study investigates the molecular mechanism by which ethanol regulates PDCD4 in cortical neuroblasts, the immediate precursor of neurons. ETOH treatment significantly increased PDCD4 protein and transcript expression in spontaneously immortalized rat brain neuroblasts. Since PDCD4 is regulated at both the post-translational and post-transcriptional level, we assessed ETOH's effect on PDCD4 protein and mRNA stability. Chase experiments demonstrated that ETOH does not significantly impact either PDCD4 protein or mRNA stabilization. PDCD4 promoter-reporter assays confirmed that PDCD4 is transcriptionally regulated by ETOH in neuroblasts. Given a critical role of glycogen synthase kinase 3β (GSK-3β) signaling in regulating protein synthesis and neurotoxic mechanisms, we investigated the involvement of GSK-3β and showed that multifunctional GSK-3β was significantly activated in response to ETOH in neuroblasts. In addition, we found that ETOH-induced activation of PDCD4 was inhibited by pharmacologic blockade of GSK-3β using inhibitors, lithium chloride (LiCl) and SB-216763 or siRNA mediated silencing of GSK-3β. These results suggest that ethanol transcriptionally upregulates PDCD4 by enhancing GSK-3β signaling in cortical neuroblasts. Further, we demonstrate that canonical Wnt-3a/GSK-3β signaling is involved in regulating PDCD4 protein expression. Altogether, we provide evidence that GSK-3β/PDCD4 network may represent a critical modulatory point to manage the protein synthetic anomalies and growth aberrations of neural cells seen in FASD.
Collapse
|
17
|
Hekmatnejad B, Gauthier C, St-Arnaud R. Control of Fiat (factor inhibiting ATF4-mediated transcription) expression by Sp family transcription factors in osteoblasts. J Cell Biochem 2013; 114:1863-70. [PMID: 23463631 DOI: 10.1002/jcb.24528] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 02/21/2013] [Indexed: 12/16/2022]
Abstract
FIAT (factor inhibiting ATF4-mediated transcription) represses Osteocalcin gene transcription and inhibits osteoblast activity by heterodimerizing with ATF4 to prevent it from binding DNA. It thus appears important to identify and characterize the molecular mechanisms that control Fiat gene expression in osteoblasts. In silico sequence analysis identified a canonical GC-box within a 1,400 bp region of the proximal Fiat gene promoter. Electrophoretic mobility shift assays (EMSA) with MC3T3-E1 osteoblastic cells nuclear extracts indicated that the transcription factors Sp1 and Sp3, but not Sp7/OSTERIX, bound this proximal GC-box. Chromatin immunoprecipitation confirmed interaction of the two transcription factors with the Fiat promoter GC-element in living osteoblasts. Transient transfection studies showed that Sp1 dose-dependently activated the expression of a Fiat-luciferase reporter construct while both the long or short isoforms of Sp3 dose-dependently inhibited transcription from the Fiat reporter construct. Transfection of an Sp7/OSTERIX expression vector did not affect expression of the Fiat-luciferase reporter. Co-transfection of increasing amounts of the Sp3 expression vector in the context of maximal Sp1-dependent Fiat-luciferase activation led to dose-dependent repression of the expression of the reporter. Using RNA knockdown, we measured a reduction in steady-state Fiat expression when Sp1 was inhibited, and a reciprocal increase upon Sp3 knockdown. In parallel, treatment of osteoblasts with WP631, which prevents Sp1/DNA interactions, strongly inhibited the expression of Fiat and reduced the occupancy of the Fiat promoter proximal GC-box by Sp1. Taken together, our results suggest an interplay between Sp1 and Sp3 as a mechanism involved in the control of Fiat gene expression in osteoblasts.
Collapse
Affiliation(s)
- Bahareh Hekmatnejad
- Genetics Unit, Shriners Hospitals for Children-Canada, Montreal, Quebec, Canada H3G 1A6
| | | | | |
Collapse
|
18
|
Chen XF, Zhang YW, Xu H, Bu G. Transcriptional regulation and its misregulation in Alzheimer's disease. Mol Brain 2013; 6:44. [PMID: 24144318 PMCID: PMC3854070 DOI: 10.1186/1756-6606-6-44] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 10/15/2013] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder characterized by loss of memory and cognitive function. A key neuropathological event in AD is the accumulation of amyloid-β (Aβ) peptide. The production and clearance of Aβ in the brain are regulated by a large group of genes. The expression levels of these genes must be fine-tuned in the brain to keep Aβ at a balanced amount under physiological condition. Misregulation of AD genes has been found to either increase AD risk or accelerate the disease progression. In recent years, important progress has been made in uncovering the regulatory elements and transcriptional factors that guide the expression of these genes. In this review, we describe the mechanisms of transcriptional regulation for the known AD genes and the misregualtion that leads to AD susceptibility.
Collapse
Affiliation(s)
- Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, 361102 Xiamen, Fujian, People's Republic of China.
| | | | | | | |
Collapse
|
19
|
Güttinger S. Creating parts that allow for rational design: synthetic biology and the problem of context-sensitivity. STUDIES IN HISTORY AND PHILOSOPHY OF BIOLOGICAL AND BIOMEDICAL SCIENCES 2013; 44:199-207. [PMID: 23578488 DOI: 10.1016/j.shpsc.2013.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The parts-based engineering approach in synthetic biology aims to create pre-characterised biological parts that can be used for the rational design of novel functional systems. Given the context-sensitivity of biological entities, a key question synthetic biologists have to address is what properties these parts should have so that they give a predictable output even when they are used in different contexts. In the first part of this paper I will analyse some of the answers that synthetic biologists have given to this question and claim that the focus of these answers on parts and their properties does not allow us to tackle the problem of context-sensitivity. In the second part of the paper, I will argue that we might have to abandon the notions of parts and their properties in order to understand how independence in biology could be achieved. Using Robert Cummins' account of functional analysis, I will then develop the notion of a capacity and its condition space and show how these notions can help to tackle the problem of context-sensitivity in biology.
Collapse
Affiliation(s)
- Stephan Güttinger
- Centre for Philosophy of Natural and Social Science, Lakatos Building, London School of Economics and Political Science, Houghton Street, London WC2A 2AE, United Kingdom.
| |
Collapse
|
20
|
Feng ZJ, Gurung B, Jin GH, Yang XL, Hua XX. SUMO modification of menin. Am J Cancer Res 2013; 3:96-106. [PMID: 23359867 PMCID: PMC3555195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 12/27/2012] [Indexed: 06/01/2023] Open
Abstract
Menin acts as contextual a tumor suppressor and a tumor promoter, partly via epigenetic regulation of gene transcription. While menin is phosphorylated, it remains unclear whether wild type menin has other post-translational modifications. Here, we report that menin is SUMOylated by SUMO1 in vivo and in vitro, and the SUMOylation is reduced by a SUMO protease. Lysine 591 of menin was covalently modified by SUMO1 and K591R mutation in menin blocked SUMOylation of the C-terminal part of menin in transfected cells. Full-length menin with K591 mutation was still SUMOylated in vivo, suggesting the existence of multiple SUMOylation sites. Menin K591R mutant or menin-SUMO fusion protein still retains the ability to regulate cell proliferation and the expression of the examined menin target genes.
Collapse
Affiliation(s)
- Zi-Jie Feng
- Department of Basic Medical Sciences, Medical College, Xiamen University Xiamen, Fujian, China 361005 ; Abramson Family Cancer Research Institute, Department of Cancer Biology, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine 421 Curie Blvd., Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
21
|
Nunes MJ, Moutinho M, Milagre I, Gama MJ, Rodrigues E. Okadaic acid inhibits the trichostatin A-mediated increase of human CYP46A1 neuronal expression in a ERK1/2-Sp3-dependent pathway. J Lipid Res 2012; 53:1910-9. [PMID: 22693257 DOI: 10.1194/jlr.m027680] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The CYP46A1 gene codes for the cholesterol 24-hydroxylase, a cytochrome P450 specifically expressed in neurons and responsible for the majority of cholesterol turnover in the central nervous system. Previously, we have demonstrated the critical participation of Sp transcription factors in the CYP46A1 response to histone deacetylase (HDAC) inhibitors, and in this study we investigated the involvement of intracellular signaling pathways in the trichostatin A (TSA) effect. Our results show that pretreatment of neuroblastoma cells with chemical inhibitors of mitogen-activated kinase kinase (MEK)1 significantly potentiates the TSA-dependent induction of cholesterol 24-hydroxylase, whereas inhibition of protein phosphatases by okadaic acid (OA) or overexpression of MEK1 partially impairs the TSA effect without affecting histone hyperacetylation at the promoter. Immunoblotting revealed that TSA treatment decreases ERK1/2 phosphorylation concomitantly with a decrease in Sp3 binding activity, which are both reversed by pretreatment with OA. Chromatin immunoprecipitation analysis demonstrated that TSA induces the release of p-ERK1/2 from the CYP46A1 proximal promoter, whereas pretreatment with OA restores the co-occupancy of Sp3-ERK1/2 in the same promoter fragments. We demonstrate for the first time the participation of MEK-ERK1/2 signaling pathway in HDAC inhibitor-dependent induction of cytochrome P450 gene expression, underlying the importance of this regulatory signaling mechanism in the control of brain cholesterol elimination.
Collapse
Affiliation(s)
- Maria João Nunes
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), University of Lisbon, 1649-019 Lisbon, Portugal
| | | | | | | | | |
Collapse
|
22
|
Expression characteristics of the SUMOylation genes SUMO-1 and Ubc9 in the developing testis and ovary of Chinese mitten crab, Eriocheir sinensis. Gene 2012; 501:135-43. [DOI: 10.1016/j.gene.2012.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 03/19/2012] [Accepted: 04/05/2012] [Indexed: 11/30/2022]
|
23
|
Kilner J, Waby JS, Chowdry J, Khan AQ, Noirel J, Wright PC, Corfe BM, Evans CA. A proteomic analysis of differential cellular responses to the short-chain fatty acids butyrate, valerate and propionate in colon epithelial cancer cells. MOLECULAR BIOSYSTEMS 2011; 8:1146-56. [PMID: 22075547 DOI: 10.1039/c1mb05219e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The short chain fatty acids (SCFAs) are inhibitors of histone deacetylases (HDACi); they are produced naturally in the colon by fermentation. They affect cellular processes at a molecular and transcriptional level, the mechanisms of which may involve large numbers of proteins and integrated pathways. Butyrate is the most biologically potent of the SCFAs in colon epithelial cells, inhibiting human colon carcinoma cell proliferation and inducing apoptosis in vitro. In order to investigate the hypothesis that propionate and valerate possess unique and independent actions from butyrate, we combined proteomic and cellomic approaches for large-scale comparative analysis. Proteomic evaluation was undertaken using an iTRAQ tandem mass-spectrometry workflow and high-throughput High-content Analysis microscopy (HCA) was applied to generate cellomic information on the cell cycle and the cytoskeletal structure. Our results show that these SCFAs possess specific effects. Butyrate was shown to have more pronounced effects on the keratins and intermediate filaments (IFs); while valerate altered the β-tubulin isotypes' expression and the microtubules (MTs); propionate was involved in both mechanisms, displaying intermediate effects. These data suggest distinct physiological roles for SCFAs in colon epithelial function, offering new possibilities for cancer therapeutics.
Collapse
Affiliation(s)
- Josephine Kilner
- ChELSI Institute, Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Chou CW, Wu MS, Huang WC, Chen CC. HDAC inhibition decreases the expression of EGFR in colorectal cancer cells. PLoS One 2011; 6:e18087. [PMID: 21464950 PMCID: PMC3064594 DOI: 10.1371/journal.pone.0018087] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 02/24/2011] [Indexed: 01/27/2023] Open
Abstract
Epidermal growth factor receptor (EGFR), a receptor tyrosine kinase which
promotes cell proliferation and survival, is abnormally overexpressed in
numerous tumors of epithelial origin, including colorectal cancer (CRC). EGFR
monoclonal antibodies have been shown to increase the median survival and are
approved for the treatment of colorectal cancer. Histone deacetylases (HDACs),
frequently overexpressed in colorectal cancer and several malignancies, are
another attractive targets for cancer therapy. Several inhibitors of HDACs
(HDACi) are developed and exhibit powerful antitumor abilities. In this study,
human colorectal cancer cells treated with HDACi exhibited reduced EGFR
expression, thereby disturbed EGF-induced ERK and Akt phosphorylation. HDACi
also decreased the expression of SGLT1, an active glucose transporter found to
be stabilized by EGFR, and suppressed the glucose uptake of cancer cells. HDACi
suppressed the transcription of EGFR and class I HDACs were proved to be
involved in this event. Chromatin immunoprecipitation analysis showed that HDACi
caused the dissociation of SP1, HDAC3 and CBP from EGFR promoter. Our data
suggested that HDACi could serve as a single agent to block both EGFR and HDAC,
and may bring more benefits to the development of CRC therapy.
Collapse
Affiliation(s)
- Chia-Wei Chou
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|
25
|
Khan AQ, Bury JP, Brown SR, Riley SA, Corfe BM. Keratin 8 expression in colon cancer associates with low faecal butyrate levels. BMC Gastroenterol 2011; 11:2. [PMID: 21219647 PMCID: PMC3027188 DOI: 10.1186/1471-230x-11-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 01/10/2011] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Butyrate has been implicated in the mechanistic basis of the prevention of colorectal cancer by dietary fibre. Numerous in vitro studies have shown that butyrate regulates cell cycle and cell death. More recently we have shown that butyrate also regulates the integrity of the intermediate filament (IF) cytoskeleton in vitro. These and other data suggest a link between the role of diet and the implication of a central role for the keratin 8 (K8) as guardian of the colorectal epithelium. METHODS In this cross-sectional study possible links between butyrate levels, field effects and keratin expression in cancer were addressed directly by analysing how levels of expression of the IF protein K8 in tumours, in adjacent fields and at a distant landmark site may be affected by the level of butyrate in the colon microenvironment. An immunohistochemical scoring protocol for K8 was developed and applied to samples, findings were further tested by immunoblotting. RESULTS Levels of K8 in colorectal tumours are lower in subjects with higher levels of faecal butyrate. Immunoblotting supported this finding.Although there were no significant relationships with butyrate on the non-tumour tissues, there was a consistent trend in all measures of extent or intensity of staining towards a reduction in expression with elevated butyrate, consistent with the inverse association in tumours. CONCLUSIONS The data suggest that butyrate may associate with down-regulation of the expression of K8 in the cancerized colon. If further validated these findings may suggest the chemopreventive value of butyrate is limited to early stage carcinogenesis as low K8 expression is associated with a poor prognosis.
Collapse
Affiliation(s)
- Abdul Q Khan
- Department of Oncology, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2JF, UK
- Department of Gastroenterology, Northern General Hospital, Herries Road, Sheffield, S5 7AU, UK
| | - Jonathan P Bury
- Department of Oncology, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2JF, UK
| | - Steven R Brown
- Department of Gastroenterology, Northern General Hospital, Herries Road, Sheffield, S5 7AU, UK
| | - Stuart A Riley
- Department of Gastroenterology, Northern General Hospital, Herries Road, Sheffield, S5 7AU, UK
| | - Bernard M Corfe
- Department of Oncology, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2JF, UK
| |
Collapse
|
26
|
Kilner J, Corfe BM, Wilkinson SJ. Modelling the microtubule: towards a better understanding of short-chain fatty acid molecular pharmacology. MOLECULAR BIOSYSTEMS 2011; 7:975-83. [DOI: 10.1039/c0mb00281j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
27
|
Hannoun Z, Greenhough S, Jaffray E, Hay RT, Hay DC. Post-translational modification by SUMO. Toxicology 2010; 278:288-93. [PMID: 20674646 DOI: 10.1016/j.tox.2010.07.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 07/06/2010] [Accepted: 07/21/2010] [Indexed: 11/28/2022]
Abstract
Post-translational modifications (PTMs) are chemical alterations to a protein following translation, regulating stability and function. Reversible phosphorylation is an example of an important and well studied PTM involved in a number of cellular processes. SUMOylation is another PTM known to modify a large number of proteins and plays a role in various cellular processes including: cell cycle regulation, gene transcription, differentiation and cellular localisation. Therefore, understanding the role of SUMOylation in cell biology may allow the development of more efficient models, important in streamlining the drug discovery process. This review will focus on protein SUMOylation and its role in stem cell and somatic cell biology.
Collapse
Affiliation(s)
- Zara Hannoun
- Medical Research Council-Centre for Regenerative Medicine, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh EH164SB, UK
| | | | | | | | | |
Collapse
|
28
|
Yiu WH, Yeung TL, Poon JWM, Tsui SKW, Fung KP, Waye MMY. Transcriptional regulation of IER3IP1 gene by tumor necrosis factor-alpha and Sp family proteins. Cell Biochem Funct 2010; 28:31-7. [PMID: 19885854 DOI: 10.1002/cbf.1613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Immediate early response 3 interacting protein 1 (IER3IP1) is an endoplasmic reticulum protein with its potential cellular function involved in cell differentiation and cell death processes. In this report, we investigated the molecular mechanism by which the expression of IER3IP1 gene is regulated by cloning the 5' flanking region of the human IER3IP1 gene for various promoter studies. Deletion analysis was used to identify the basal promoter activity retained at -298/-59 region and mutation analysis proved that Sp1 is a transcriptional activator of this gene expression. As an early response gene, IER3IP1 showed an increase in transcription in response to tumor necrosis factor alpha (TNF-alpha) in a time- and dose-dependent manner. This inducible response to TNF-alpha is mediated by the demonstration of nuclear factor kappaB (NF-kappaB) responsive element on IER3IP1 promoter sequence. From our results, we suggest that IER3IP1 gene is involved in TNF-alpha-mediated cellular response to stressful conditions.
Collapse
Affiliation(s)
- Wai Han Yiu
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, N.T., China
| | | | | | | | | | | |
Collapse
|
29
|
Simmen RCM, Pabona JMP, Velarde MC, Simmons C, Rahal O, Simmen FA. The emerging role of Krüppel-like factors in endocrine-responsive cancers of female reproductive tissues. J Endocrinol 2010; 204:223-31. [PMID: 19833720 PMCID: PMC2971688 DOI: 10.1677/joe-09-0329] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Krüppel-like factors (KLFs), of which there are currently 17 known protein members, belong to the specificity protein (Sp) family of transcription factors and are characterized by the presence of Cys(2)/His(2) zinc finger motifs in their carboxy-terminal domains that confer preferential binding to GC/GT-rich sequences in gene promoter and enhancer regions. While previously regarded to simply function as silencers of Sp1 transactivity, many KLFs are now shown to be relevant to human cancers by their newly identified abilities to mediate crosstalk with signaling pathways involved in the control of cell proliferation, apoptosis, migration, and differentiation. Several KLFs act as tumor suppressors and/or oncogenes under distinct cellular contexts, underscoring their prognostic potential for cancer survival and outcome. Recent studies suggest that a number of KLFs can influence steroid hormone signaling through transcriptional networks involving steroid hormone receptors and members of the nuclear receptor family of transcription factors. Since inappropriate sensitivity or resistance to steroid hormone actions underlies endocrine-related malignancies, we consider the intriguing possibility that dysregulation of expression and/or activity of KLF members is linked to the pathogenesis of endometrial and breast cancers. In this review, we focus on recently described mechanisms of actions of several KLFs (KLF4, KLF5, KLF6, and KLF9) in cancers of the mammary gland and uterus. We suggest that understanding the mode of actions of KLFs and their functional networks may lead to the development of novel therapeutics to improve current prospects for cancer prevention and cure.
Collapse
Affiliation(s)
- R C M Simmen
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72202, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Drake PJM, Griffiths GJ, Shaw L, Benson RP, Corfe BM. Application of High-Content Analysis to the Study of Post-Translational Modifications of the Cytoskeleton. J Proteome Res 2008; 8:28-34. [DOI: 10.1021/pr8006396] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Peter J. M. Drake
- Human Nutrition Unit, School of Medicine, University of Sheffield, Royal Hallamshire Hospital, Sheffield S10 2JF, United Kingdom, and Imagen Biotechnology Ltd., Grafton Street, Manchester M13 9NT, United Kingdom
| | - Gareth J. Griffiths
- Human Nutrition Unit, School of Medicine, University of Sheffield, Royal Hallamshire Hospital, Sheffield S10 2JF, United Kingdom, and Imagen Biotechnology Ltd., Grafton Street, Manchester M13 9NT, United Kingdom
| | - Leila Shaw
- Human Nutrition Unit, School of Medicine, University of Sheffield, Royal Hallamshire Hospital, Sheffield S10 2JF, United Kingdom, and Imagen Biotechnology Ltd., Grafton Street, Manchester M13 9NT, United Kingdom
| | - Rod P. Benson
- Human Nutrition Unit, School of Medicine, University of Sheffield, Royal Hallamshire Hospital, Sheffield S10 2JF, United Kingdom, and Imagen Biotechnology Ltd., Grafton Street, Manchester M13 9NT, United Kingdom
| | - Bernard M. Corfe
- Human Nutrition Unit, School of Medicine, University of Sheffield, Royal Hallamshire Hospital, Sheffield S10 2JF, United Kingdom, and Imagen Biotechnology Ltd., Grafton Street, Manchester M13 9NT, United Kingdom
| |
Collapse
|