1
|
Izquierdo-Pujol J, Puertas MC, Martinez-Picado J, Morón-López S. Targeting Viral Transcription for HIV Cure Strategies. Microorganisms 2024; 12:752. [PMID: 38674696 PMCID: PMC11052381 DOI: 10.3390/microorganisms12040752] [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: 03/21/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Combination antiretroviral therapy (ART) suppresses viral replication to undetectable levels, reduces mortality and morbidity, and improves the quality of life of people living with HIV (PWH). However, ART cannot cure HIV infection because it is unable to eliminate latently infected cells. HIV latency may be regulated by different HIV transcription mechanisms, such as blocks to initiation, elongation, and post-transcriptional processes. Several latency-reversing (LRA) and -promoting agents (LPA) have been investigated in clinical trials aiming to eliminate or reduce the HIV reservoir. However, none of these trials has shown a conclusive impact on the HIV reservoir. Here, we review the cellular and viral factors that regulate HIV-1 transcription, the potential pharmacological targets and genetic and epigenetic editing techniques that have been or might be evaluated to disrupt HIV-1 latency, the role of miRNA in post-transcriptional regulation of HIV-1, and the differences between the mechanisms regulating HIV-1 and HIV-2 expression.
Collapse
Affiliation(s)
- Jon Izquierdo-Pujol
- IrsiCaixa, 08916 Badalona, Spain; (J.I.-P.); (M.C.P.); (J.M.-P.)
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Maria C. Puertas
- IrsiCaixa, 08916 Badalona, Spain; (J.I.-P.); (M.C.P.); (J.M.-P.)
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- CIBERINFEC, 28029 Madrid, Spain
| | - Javier Martinez-Picado
- IrsiCaixa, 08916 Badalona, Spain; (J.I.-P.); (M.C.P.); (J.M.-P.)
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- CIBERINFEC, 28029 Madrid, Spain
- Department of Infectious Diseases and Immunity, School of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Sara Morón-López
- IrsiCaixa, 08916 Badalona, Spain; (J.I.-P.); (M.C.P.); (J.M.-P.)
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
- CIBERINFEC, 28029 Madrid, Spain
| |
Collapse
|
2
|
Malbeteau L, Pham HT, Eve L, Stallcup MR, Poulard C, Le Romancer M. How Protein Methylation Regulates Steroid Receptor Function. Endocr Rev 2022; 43:160-197. [PMID: 33955470 PMCID: PMC8755998 DOI: 10.1210/endrev/bnab014] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Indexed: 02/06/2023]
Abstract
Steroid receptors (SRs) are members of the nuclear hormonal receptor family, many of which are transcription factors regulated by ligand binding. SRs regulate various human physiological functions essential for maintenance of vital biological pathways, including development, reproduction, and metabolic homeostasis. In addition, aberrant expression of SRs or dysregulation of their signaling has been observed in a wide variety of pathologies. SR activity is tightly and finely controlled by post-translational modifications (PTMs) targeting the receptors and/or their coregulators. Whereas major attention has been focused on phosphorylation, growing evidence shows that methylation is also an important regulator of SRs. Interestingly, the protein methyltransferases depositing methyl marks are involved in many functions, from development to adult life. They have also been associated with pathologies such as inflammation, as well as cardiovascular and neuronal disorders, and cancer. This article provides an overview of SR methylation/demethylation events, along with their functional effects and biological consequences. An in-depth understanding of the landscape of these methylation events could provide new information on SR regulation in physiology, as well as promising perspectives for the development of new therapeutic strategies, illustrated by the specific inhibitors of protein methyltransferases that are currently available.
Collapse
Affiliation(s)
- Lucie Malbeteau
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Ha Thuy Pham
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Louisane Eve
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Michael R Stallcup
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Coralie Poulard
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Muriel Le Romancer
- Université de Lyon, F-69000 Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| |
Collapse
|
3
|
Tseng MH, Huang SM, Konrad M, Huang JL, Shaw SW, Tian YC, Chueh HY, Fan WL, Wu TW, Ding JJ, Chiang MC, Lin SH. Effect of Hydrocortisone on Angiotensinogen ( AGT) Mutation-Causing Autosomal Recessive Renal Tubular Dysgenesis. Cells 2021; 10:cells10040782. [PMID: 33916187 PMCID: PMC8065467 DOI: 10.3390/cells10040782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
We has identified a founder homozygous E3_E4 del: 2870 bp deletion + 9 bp insertion in AGT gene encoding angiotensinogen responsible for autosomal recessive renal tubular dysgenesis (ARRTD) with nearly-fatal outcome. High-dose hydrocortisone therapy successfully rescued one patient with an increased serum Angiotensinogen (AGT), Ang I, and Ang II levels. The pathogenesis of ARRTD caused by this AGT mutation and the potential therapeutic effect of hydrocortisone were examined by in vitro functional studies. The expression of this truncated AGT protein was relatively low with a dose-dependent manner. This truncated mutation diminished the interaction between mutant AGT and renin. The truncated AGT also altered the glucocorticoid receptor (GR)-dependent transactivation, indicating that AGT may affect the development of proximal convoluted tubule by alteration of glucocorticoid-dependent transactivation. In hepatocytes, hydrocortisone increased the AGT level by accentuating the stability of mutant AGT and increasing its binding with renin. Therefore, hydrocortisone may exert the therapeutic effect through the enhanced stability and interaction with renin of truncated AGT in patients carrying this AGT mutation.
Collapse
Affiliation(s)
- Min-Hua Tseng
- Division of Nephrology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 330, Taiwan;
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan;
| | - Martin Konrad
- Department of General Pediatrics, University Children’s Hospital Münster, 481 Münster, Germany;
| | - Jing-Long Huang
- Division of Pediatric Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 330, Taiwan;
| | - Steven W. Shaw
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital and Chang Gung University, Taipei 114, Taiwan;
| | - Ya-Chung Tian
- Division of Nephrology, Department of Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 330, Taiwan;
| | - Ho-Yen Chueh
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 330, Taiwan;
| | - Wen-Lang Fan
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Tai-Wei Wu
- Fetal and Neonatal Institute, Division of Neonatology Children’s Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 900, USA;
| | - Jhao-Jhuang Ding
- Department of Pediatrics, Tri-Service General Hospital, Taipei 114, Taiwan;
| | - Ming-Chou Chiang
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 330, Taiwan;
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, Taipei 114, Taiwan
- Correspondence: ; Tel.: +886-2-87927213; Fax: +886-2-87927134
| |
Collapse
|
4
|
Sanchez Caballero L, Gorgogietas V, Arroyo MN, Igoillo-Esteve M. Molecular mechanisms of β-cell dysfunction and death in monogenic forms of diabetes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:139-256. [PMID: 33832649 DOI: 10.1016/bs.ircmb.2021.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monogenetic forms of diabetes represent 1%-5% of all diabetes cases and are caused by mutations in a single gene. These mutations, that affect genes involved in pancreatic β-cell development, function and survival, or insulin regulation, may be dominant or recessive, inherited or de novo. Most patients with monogenic diabetes are very commonly misdiagnosed as having type 1 or type 2 diabetes. The severity of their symptoms depends on the nature of the mutation, the function of the affected gene and, in some cases, the influence of additional genetic or environmental factors that modulate severity and penetrance. In some patients, diabetes is accompanied by other syndromic features such as deafness, blindness, microcephaly, liver and intestinal defects, among others. The age of diabetes onset may also vary from neonatal until early adulthood manifestations. Since the different mutations result in diverse clinical presentations, patients usually need different treatments that range from just diet and exercise, to the requirement of exogenous insulin or other hypoglycemic drugs, e.g., sulfonylureas or glucagon-like peptide 1 analogs to control their glycemia. As a consequence, awareness and correct diagnosis are crucial for the proper management and treatment of monogenic diabetes patients. In this chapter, we describe mutations causing different monogenic forms of diabetes associated with inadequate pancreas development or impaired β-cell function and survival, and discuss the molecular mechanisms involved in β-cell demise.
Collapse
Affiliation(s)
- Laura Sanchez Caballero
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Vyron Gorgogietas
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Maria Nicol Arroyo
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/.
| |
Collapse
|
5
|
A functional screen identifies transcriptional networks that regulate HIV-1 and HIV-2. Proc Natl Acad Sci U S A 2021; 118:2012835118. [PMID: 33836568 DOI: 10.1073/pnas.2012835118] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The molecular networks involved in the regulation of HIV replication, transcription, and latency remain incompletely defined. To expand our understanding of these networks, we performed an unbiased high-throughput yeast one-hybrid screen, which identified 42 human transcription factors and 85 total protein-DNA interactions with HIV-1 and HIV-2 long terminal repeats. We investigated a subset of these transcription factors for transcriptional activity in cell-based models of infection. KLF2 and KLF3 repressed HIV-1 and HIV-2 transcription in CD4+ T cells, whereas PLAGL1 activated transcription of HIV-2 through direct protein-DNA interactions. Using computational modeling with interacting proteins, we leveraged the results from our screen to identify putative pathways that define intrinsic transcriptional networks. Overall, we used a high-throughput functional screen, computational modeling, and biochemical assays to identify and confirm several candidate transcription factors and biochemical processes that influence HIV-1 and HIV-2 transcription and latency.
Collapse
|
6
|
Chung LH, Liu ST, Huang SM, Salter DM, Lee HS, Hsu YJ. High phosphate induces skeletal muscle atrophy and suppresses myogenic differentiation by increasing oxidative stress and activating Nrf2 signaling. Aging (Albany NY) 2020; 12:21446-21468. [PMID: 33136552 PMCID: PMC7695395 DOI: 10.18632/aging.103896] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022]
Abstract
Skeletal muscle wasting represents both a common phenotype of aging and a feature of pathological conditions such as chronic kidney disease (CKD). Although both clinical data and genetic experiments in mice suggest that hyperphosphatemia accelerates muscle wasting, the underlying mechanism remains unclear. Here, we showed that inorganic phosphate (Pi) dose-dependently decreases myotube size, fusion index, and myogenin expression in mouse C2C12 skeletal muscle cells. These changes were accompanied by increases in reactive oxygen species (ROS) production and Nrf2 and p62 expression, and reductions in mitochondrial membrane potential (MMP) and Keap1 expression. Inhibition of Pi entry, cytosolic ROS production, or Nrf2 activation reversed the effects of high Pi on Nrf2, p62, and myogenin expression. Overexpression of Nrf2 respectively increased and decreased the promoter activity of p62-Luc and myogenin-Luc reporters. Analysis of nuclear extracts from gastrocnemius muscles from mice fed a high-Pi (2% Pi) diet showed increased Nrf2 phosphorylation in sham-operated and 5/6 nephrectomized (CKD) mice, and both increased p62 phosphorylation and decreased myogenin expression in CKD mice. These data suggest that high Pi suppresses myogenic differentiation in vitro and promotes muscle atrophy in vivo through oxidative stress-mediated protein degradation and both canonical (ROS-mediated) and non-canonical (p62-mediated) activation of Nrf2 signaling.
Collapse
Affiliation(s)
- Lin-Huei Chung
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Yuan Rung Hospital, Changhua, Taiwan
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ming Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Donald M Salter
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Herng-Sheng Lee
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yu-Juei Hsu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
7
|
Wu TM, Liu ST, Chen SY, Chen GS, Wu CC, Huang SM. Mechanisms and Applications of the Anti-cancer Effect of Pharmacological Ascorbic Acid in Cervical Cancer Cells. Front Oncol 2020; 10:1483. [PMID: 33014789 PMCID: PMC7507989 DOI: 10.3389/fonc.2020.01483] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, L-ascorbic acid (L-AA), or vitamin C, has been attracting attention as a potential anticancer drug that mediates hydrogen peroxide-induced oxidation and ten-eleven translocation 2-catalyzed DNA demethylation. However, the precise mechanism by which L-AA acts remains unclear. We examined the cytotoxic effects of L-AA or sodium ascorbate in human cervical carcinoma cells by assessing cell viability, expression of cell cycle-related mRNAs and proteins, and mitochondrial functions, and by performing flow cytometric analyses of cell cycle profiles, apoptosis, cell proliferation, and production of reactive oxygen species (ROS). We later tested the effects of ascorbates in combination with two first-line chemotherapeutic drugs, cisplatin, and doxorubicin. At pharmacological concentrations (1–10 mM), L-AA increased ROS levels; decreased levels of several cell cycle-related proteins, including p53, p21, cyclin D1, and phosphorylated histone 3 at serine residue 10; induced DNA damage, as indicated by changes in γH2A.x; decreased levels of the anti-oxidative transcription factor Nrf2; and increased levels of catalase, superoxide dismutase 1, and endoplasmic reticulum stress-related indicators, such as the p-eIF2α/eIF2α ratio and CHOP levels. L-AA also promoted cell proliferation and induced apoptosis and mitochondrial dysfunction. Finally, L-AA increased the susceptibility of HeLa cells to cisplatin and doxorubicin. These findings provide insight into how the adjustment of the cellular ROS status through L-ascorbate (L-AA or sodium ascorbate) administration could potentially synergistically enhance the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Tsai-Ming Wu
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan
| | - Ssu-Yu Chen
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan
| | - Gunng-Shinng Chen
- Department of Dentistry of Tri-service General Hospital, School of Dentistry, National Defense Medical Center, Taipei City, Taiwan
| | - Chia-Chun Wu
- Department of Orthopaedic Surgery, Tri-service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan
| |
Collapse
|
8
|
Gene expression profiling identifies the role of Zac1 in cervical cancer metastasis. Sci Rep 2020; 10:11837. [PMID: 32678267 PMCID: PMC7367306 DOI: 10.1038/s41598-020-68835-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/06/2020] [Indexed: 12/22/2022] Open
Abstract
The zinc-finger protein which regulates apoptosis and cell cycle arrest 1 (Zac1), encoded by Plagl1 gene, is a seven-zinc-finger containing transcription factor belonging to the imprinted genome and is expressed in diverse types of embryonic and adult human tissues. Zac1 is postulated to be a tumor suppressor by inducing cell cycle arrest and apoptosis through interacting and modulating transcriptional activity of p53 as it was named. Correspondingly, the reduction or loss of Zac1 expression is associated with the incidence and progression of several human tumors, including cervical cancer, breast cancer, ovarian cancer, pituitary tumors, and basal cell carcinoma, implying the rationality of utilizing Zac1 expression as novel a biomarker for the evaluation of cervical cancer prognosis. However, to date, it has not been elucidated whether Zac1 expression is related to the prognosis of patients in clinical cervical cancer tumor samples. To address the questions outlined above, we report here a comprehensive investigation of Zac1 expression in biopsies of clinical cervical carcinoma. By analyzing Zac1 expression in various gene expression profiling of cervical cancer databases, we show the association between high Zac1 expression and poor prognosis of cervical cancer. Functional enrichment analysis showed that high Zac1 expression was associated with epithelial-mesenchymal transition (EMT), which was further observed in clinical characteristics and metastatic carcinoma samples using immunohistochemical staining. Correspondingly, hypomethylation of CpG island on Zac1 promoter was observed in samples with high Zac1 expression in cervical carcinoma. Finally, overexpression of Zac1 in a variety of cervical cancer cell lines increase their mesenchymal biomarker expression and migration, strengthening the correlation between cervical cancers with high Zac1 expression and metastasis in clinical. In summary, this research firstly revealed that identifying Zac1 expression or the methylation status of CpG site on Zac1 promoter may provide us with novel indicators for the evaluation of cervical cancer metastasis.
Collapse
|
9
|
Stallcup MR, Poulard C. Gene-Specific Actions of Transcriptional Coregulators Facilitate Physiological Plasticity: Evidence for a Physiological Coregulator Code. Trends Biochem Sci 2020; 45:497-510. [PMID: 32413325 DOI: 10.1016/j.tibs.2020.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/24/2020] [Accepted: 02/10/2020] [Indexed: 01/14/2023]
Abstract
The actions of transcriptional coregulators are highly gene-specific, that is, each coregulator is required only for a subset of the genes regulated by a specific transcription factor. These coregulator-specific gene subsets often represent selected physiological responses among multiple pathways targeted by a transcription factor. Regulating the activity of a coregulator via post-translational modifications would thus affect only a subset of the transcription factor's physiological actions. Using the context of transcriptional regulation by steroid hormone receptors, this review focuses on gene-specific actions of coregulators and evidence linking individual coregulators with specific physiological pathways. Such evidence suggests that there is a 'physiological coregulator code', which represents a fertile area for future research with important clinical implications.
Collapse
Affiliation(s)
- Michael R Stallcup
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA90089-9176, USA.
| | - Coralie Poulard
- Université de Lyon, F-69000 Lyon, France; Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France; CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| |
Collapse
|
10
|
Kuo CL, Hsieh Li SM, Liang SY, Liu ST, Huang LC, Wang WM, Yen LC, Huang SM. The antitumor properties of metformin and phenformin reflect their ability to inhibit the actions of differentiated embryo chondrocyte 1. Cancer Manag Res 2019; 11:6567-6579. [PMID: 31410055 PMCID: PMC6643064 DOI: 10.2147/cmar.s210637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Background Differentiated embryo chondrocyte 1 (DEC1) is a helix-loop-helix transcription factor that directly binds to the class B E-box in target genes. DEC1 exerts both pro-survival and pro-apoptotic effects in a cell- and tissue-dependent manner. Its actions play role the progression of cancer remains unclear. Methods We first examined the functional roles of DEC1 using the transient promoter reporter assay. Then, the knockdown of DEC1 expression was performed with the short hairpin RNA strategy in HeLa and A2058 cancer cell lines to check the cell cycle and mitochondrial function profile using the flow cytometry and Seahorse assays. We later clarified the role of DEC1 in the tumorigenesis using the colony formation, anchorage-independent growth assay, and cellular proliferation analysis. Results In the present study, we tested two guanide-containing drugs, metformin and phenformin, and found that both exhibit cytotoxicity against HeLa cervical carcinoma and A2058 melanoma cells. This effect was mediated, at least in part, through activation of the AMPK pathway; degradation of important cellular proteins, such as DEC1 and p53; and suppression of mitochondrial function, colony formation, and anchorage-independent cell proliferation. Our results further suggest that the cytotoxicity of metformin and phenformin reflect the impact of the repressive actions of DEC1 on gene expression, including DEC1 itself. This in turn suppresses both anchorage-independent growth and cell proliferation. Conclusion These findings provide several lines of evidence suggesting that DEC1 activity contributes to tumorigenicity and that the antitumor properties of biguanides reflect their ability to inhibit DEC1 functions.
Collapse
Affiliation(s)
- Chun-Lin Kuo
- Department of Orthopaedic Surgery, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | - Shu-Man Hsieh Li
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Shu-Yi Liang
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Li-Chun Huang
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Wei-Ming Wang
- Department of Dermatology, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | - Li-Chen Yen
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China.,Department of Microbiology and Immunology, National Defense Medical Center, Taiwan, Republic of China
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| |
Collapse
|
11
|
Adnani L, Dixit R, Chen X, Balakrishnan A, Modi H, Touahri Y, Logan C, Schuurmans C. Plag1 and Plagl2 have overlapping and distinct functions in telencephalic development. Biol Open 2018; 7:bio.038661. [PMID: 30361413 PMCID: PMC6262857 DOI: 10.1242/bio.038661] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The Plag gene family has three members; Plagl1/Zac1, which is a tumor suppressor gene, and Plag1 and Plagl2, which are proto-oncogenes. All three genes are known to be expressed in embryonic neural progenitors, and Zac1 regulates proliferation, neuronal differentiation and migration in the developing neocortex. Here we examined the functions of Plag1 and Plagl2 in neocortical development. We first attempted, and were unable to generate, E12.5 Plag1;Plagl2 double mutants, indicating that at least one Plag1 or Plagl2 gene copy is required for embryonic survival. We therefore focused on single mutants, revealing a telencephalic patterning defect in E12.5 Plagl2 mutants and a proliferation/differentiation defect in Plag1 mutant neocortices. Specifically, the ventral pallium, a dorsal telencephalic territory, expands into the ventral telencephalon in Plagl2 mutants. In contrast, Plag1 mutants develop normal regional territories, but neocortical progenitors proliferate less and instead produce more neurons. Finally, in gain-of-function studies, both Plag1 and Plagl2 reduce neurogenesis and increase BrdU-uptake, indicative of enhanced proliferation, but while Plagl2 effects on proliferation are more immediate, Plag1 effects are delayed. Taken together, we found that the Plag proto-oncogenes genes are essential regulators of neocortical development and although Plag1 and Plagl2 functions are similar, they do not entirely overlap. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lata Adnani
- Sunnybrook Research Institute, Biological Sciences, Room S1-16A, 2075 Bayview Ave, Toronto, ON, Canada M4N 3M5.,Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Rajiv Dixit
- Sunnybrook Research Institute, Biological Sciences, Room S1-16A, 2075 Bayview Ave, Toronto, ON, Canada M4N 3M5.,Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Xingyu Chen
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Anjali Balakrishnan
- Sunnybrook Research Institute, Biological Sciences, Room S1-16A, 2075 Bayview Ave, Toronto, ON, Canada M4N 3M5.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Harshil Modi
- Sunnybrook Research Institute, Biological Sciences, Room S1-16A, 2075 Bayview Ave, Toronto, ON, Canada M4N 3M5
| | - Yacine Touahri
- Sunnybrook Research Institute, Biological Sciences, Room S1-16A, 2075 Bayview Ave, Toronto, ON, Canada M4N 3M5
| | - Cairine Logan
- Department of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences, Room S1-16A, 2075 Bayview Ave, Toronto, ON, Canada M4N 3M5 .,Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
12
|
Mackeh R, Marr AK, Fadda A, Kino T. C2H2-Type Zinc Finger Proteins: Evolutionarily Old and New Partners of the Nuclear Hormone Receptors. NUCLEAR RECEPTOR SIGNALING 2018; 15:1550762918801071. [PMID: 30718982 PMCID: PMC6348741 DOI: 10.1177/1550762918801071] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/02/2017] [Indexed: 12/21/2022]
Abstract
Nuclear hormone receptors (NRs) are evolutionarily conserved ligand-dependent
transcription factors. They are essential for human life, mediating the actions
of lipophilic molecules, such as steroid hormones and metabolites of fatty acid,
cholesterol, and external toxic compounds. The C2H2-type zinc finger proteins
(ZNFs) form the largest family of the transcription factors in humans and are
characterized by multiple, tandemly arranged zinc fingers. Many of the C2H2-type
ZNFs are conserved throughout evolution, suggesting their involvement in
preserved biological activities, such as general transcriptional regulation and
development/differentiation of organs/tissues observed in the early embryonic
phase. However, some C2H2-type ZNFs, such as those with the Krüppel-associated
box (KRAB) domain, appeared relatively late in evolution and have significantly
increased family members in mammals including humans, possibly modulating their
complicated transcriptional network and/or supporting the morphological
development/functions specific to them. Such evolutional characteristics of the
C2H2-type ZNFs indicate that these molecules influence the NR functions
conserved through evolution, whereas some also adjust them to meet with specific
needs of higher organisms. We review the interaction between NRs and C2H2-type
ZNFs by focusing on some of the latter molecules.
Collapse
|
13
|
Kuo CL, Liu ST, Chang YL, Wu CC, Huang SM. Zac1 regulates IL-11 expression in osteoarthritis. Oncotarget 2018; 9:32478-32495. [PMID: 30197757 PMCID: PMC6126702 DOI: 10.18632/oncotarget.25980] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/29/2018] [Indexed: 01/24/2023] Open
Abstract
Interleukin (IL)-11, a member of the IL-6 family of cytokines, exerts pleiotropic effects under normal and various disease conditions. We assessed IL-11 expression regulation and the IL-11/IL-6 ratio in osteoarthritis (OA) to better guide clinical therapeutic decision-making. Our findings suggest that Zac1, a zinc finger protein that regulates apoptosis and cell cycle arrest, is a transcription factor regulating IL-11 expression. Zac1 overexpression or knockdown respectively induced or suppressed IL-11 expression in HeLa cells. Zac1 acted synergistically with AP-1, human papillomavirus E2, and hypoxia inducible factor 1 alpha (HIF1α). IL-11 expression under various conditions, including hypoxia or treatment with phorbol 12-myristate 13-acetate or copper sulfate. Recombinant IL-11-induced phosphorylation of signal transducer and activator of transcription 3 at tyrosine 705 was reduced in a dose-dependent manner in HeLa cells. Cross-talk between Zac1, IL-11, p53, and suppressor of cytokine signaling 3 was differentially affected by copper sulfate, digoxin, and caffeine. Finally, aggressive vs. conventional treatment of OA patients was primarily determined by IL-6 levels. However, we suggest that OA patients with higher IL-11 levels may respond well to conventional treatments, even in the presence of high IL-6.
Collapse
Affiliation(s)
- Chun-Lin Kuo
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taiwan, Republic of China
- Department of Orthopaedic Surgery, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| | - Chia-Chun Wu
- Department of Orthopaedic Surgery, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | - Shih-Ming Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taiwan, Republic of China
- Department of Biochemistry, National Defense Medical Center, Taiwan, Republic of China
| |
Collapse
|
14
|
Fortunati N, Guaraldi F, Zunino V, Penner F, D'Angelo V, Zenga F, Pecori Giraldi F, Catalano MG, Arvat E. Effects of environmental pollutants on signaling pathways in rat pituitary GH3 adenoma cells. ENVIRONMENTAL RESEARCH 2017; 158:660-668. [PMID: 28732322 DOI: 10.1016/j.envres.2017.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/26/2017] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
An increased rate of acromegaly was reported in industrialized areas, suggesting an involvement of environmental pollutants in the pathogenesis and behavior of GH-secreting pituitary adenomas. Based on these premises, the aim of the study was to evaluate the effects of some widely diffused pollutants (i.e. benzene, BZ; bis(2-ethylhexyl) phthalate, DEHP and polychlorinated biphenyls, PCB) on growth hormone secretion, the somatostatin and estrogenic pathways, viability and proliferation of rat GH-producing pituitary adenoma (GH3) cells. All the pollutants induced a statistically significant increase in GH secretion and interfered with cell signaling. They all modulated the expression of SSTR2 and ZAC1, involved in the somatostatin signaling, and the expression of the transcription factor FOXA1, involved in the estrogen receptor signaling. Moreover, all the pollutants increased the expression of the CYP1A1, suggesting AHR pathway activation. None of the pollutants impacted on cell proliferation or viability. Present data demonstrate that exposure to different pollutants, used at in vivo relevant concentrations, plays an important role in the behavior of GH3 pituitary adenoma cells, by increasing GH secretion and modulating several cellular signaling pathways. These observations support a possible influence of different pollutants in vivo on the GH-adenoma aggressiveness and biological behavior.
Collapse
Affiliation(s)
- Nicoletta Fortunati
- Division of Oncological Endocrinology, Città della Salute e della Scienza University Hospital, I-10126 Turin, Italy
| | - Federica Guaraldi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, Città della Salute e della Scienza University Hospital, I-10126 Turin, Italy
| | - Valentina Zunino
- Division of Oncological Endocrinology, Città della Salute e della Scienza University Hospital, I-10126 Turin, Italy
| | - Federica Penner
- Division of Neurosurgery, Città della Salute e della Scienza University Hospital, I-10126 Turin, Italy
| | - Valentina D'Angelo
- Division of Oncological Endocrinology, Città della Salute e della Scienza University Hospital, I-10126 Turin, Italy
| | - Francesco Zenga
- Division of Neurosurgery, Città della Salute e della Scienza University Hospital, I-10126 Turin, Italy
| | - Francesca Pecori Giraldi
- Neuroendocrinology Research Laboratory, Istituto Auxologico Italiano IRCCS, Cusano Milanino, (MI), Italy and Department of Clinical Sciences and Community Health, University of Milan, I-20149 Milan, Italy
| | | | - Emanuela Arvat
- Division of Oncological Endocrinology, Città della Salute e della Scienza University Hospital, I-10126 Turin, Italy; Department of Medical Sciences, University of Turin, I-10126 Turin, Italy.
| |
Collapse
|
15
|
Chu YW, Liu ST, Yang YL, Huang SM, Wang WM. The cytotoxic mechanism of epigallocatechin gallate on proliferative HaCaT keratinocytes. J Biomed Sci 2017; 24:55. [PMID: 28810862 PMCID: PMC5556358 DOI: 10.1186/s12929-017-0363-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/09/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epigallocatechin gallate (EGCG) is the major ingredient of sinecatechins ointment, approved for the treatment of external genital and perianal warts. However, the molecular mechanism for EGCG's effect on warts resulting from the human papillomavirus (HPV) infection of keratinocytes is not well understood. HPV may survive in proliferative keratinocytes and may be involved in cell cycle regulation and progression. The objective of this study was to investigate the mechanism underlying EGCG's treatment on external genital warts of HPV infection through the cultured keratinocyte cells from the HaCaT cell line. METHODS MTT and flow cytometry assays were used to measure cell viability and the cell cycle profile, with and without EGCG treatment, for HaCaT keratinocyte cells cultured in a calcium-free medium and 1.8 mM calcium which induced proliferative and differentiated keratinocytes, respectively, for 24 h. The expression levels of cytotoxic proteins and factors were evaluated with the RT-PCR and western blotting analysis. RESULTS EGCG influenced the proliferation stage but not the differentiation stage of keratinocytes. We suggest that apoptosis and autophagy might be the possible mechanism for the EGCG's effect on the proliferative HaCaT cells. Furthermore, we found that EGCG reduced the protein levels of cyclin D1 and Zac1 (a zinc-finger protein which regulates apoptosis and cell cycle arrest 1) dose-dependently in proliferative as compared to differentiated keratinocytes. It also induced the expression of p21 and DEC1 (differentiated embryo-chondrocyte expressed gene 1), and promoted G1 arrest of cell cycle in proliferative keratinocytes. CONCLUSIONS These results help clarify the mechanisms of EGCG treatment of HPV-infected keratinocytes and may contribute to new targets, such as Zac1 and DEC1 for external genital and perianal warts.
Collapse
Affiliation(s)
- Yu-Wen Chu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, 114, Taiwan, Republic of China.,Department of Pharmacy, Taichung Veterans General Hospital, Taichung, 407, Taiwan, Republic of China
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taipei, 114, Taiwan, Republic of China
| | - Ya-Lan Yang
- Department of Biochemistry, National Defense Medical Center, Taipei, 114, Taiwan, Republic of China
| | - Shih-Ming Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, 114, Taiwan, Republic of China. .,Department of Biochemistry, National Defense Medical Center, Taipei, 114, Taiwan, Republic of China.
| | - Wei-Ming Wang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, 114, Taiwan, Republic of China. .,Department of Dermatology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan, Republic of China.
| |
Collapse
|
16
|
Poulard C, Bittencourt D, Wu DY, Hu Y, Gerke DS, Stallcup MR. A post-translational modification switch controls coactivator function of histone methyltransferases G9a and GLP. EMBO Rep 2017; 18:1442-1459. [PMID: 28615290 DOI: 10.15252/embr.201744060] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/10/2017] [Accepted: 05/16/2017] [Indexed: 11/09/2022] Open
Abstract
Like many transcription regulators, histone methyltransferases G9a and G9a-like protein (GLP) can act gene-specifically as coregulators, but mechanisms controlling this specificity are mostly unknown. We show that adjacent post-translational methylation and phosphorylation regulate binding of G9a and GLP to heterochromatin protein 1 gamma (HP1γ), formation of a ternary complex with the glucocorticoid receptor (GR) on chromatin, and function of G9a and GLP as coactivators for a subset of GR target genes. HP1γ is recruited by G9a and GLP to GR binding sites associated with genes that require G9a, GLP, and HP1γ for glucocorticoid-stimulated transcription. At the physiological level, G9a and GLP coactivator function is required for glucocorticoid activation of genes that repress cell migration in A549 lung cancer cells. Thus, regulated methylation and phosphorylation serve as a switch controlling G9a and GLP coactivator function, suggesting that this mechanism may be a general paradigm for directing specific transcription factor and coregulator actions on different genes.
Collapse
Affiliation(s)
- Coralie Poulard
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Danielle Bittencourt
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Dai-Ying Wu
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Yixin Hu
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Daniel S Gerke
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Michael R Stallcup
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Chen SJ, Kao YH, Jing L, Chuang YP, Wu WL, Liu ST, Huang SM, Lai JH, Ho LJ, Tsai MC, Lin CS. Epigallocatechin-3-gallate Reduces Scavenger Receptor A Expression and Foam Cell Formation in Human Macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3141-3150. [PMID: 28367625 DOI: 10.1021/acs.jafc.6b05832] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Foam cells are formed when macrophages imbibe low-density lipoprotein (LDL) through scavenger receptors. Here we examined how epigallocatechin-3-gallate (EGCG) influences foam cell formation. We found that EGCG dose-dependently reduced oxidized LDL (oxLDL) uptake in THP-1 (10 μM, 20.0 ± 0.50, p < 0.05) and primary macrophages (134.6 ± 15.6, p < 0.05) and reduced intracellular cholesterol content in these cells, respectively (10 μM, 32.6 ± 0.14, p < 0.05; 31.7 ± 1.26, p < 0.05). EGCG treatment decreased scavenger receptor A expression, but not the expression of CD36 or of reverse cholesterol transporters. Moreover, EGCG stimulated translocation of the p50 and p65 subunits of NF-κB and enhanced NF-κB DNA-binding activity, thus suppressing SR-A promoter activity. EGCG's suppression of SR-A expression was blocked by the NF-κB inhibitor Bay. The present findings suggest that EGCG regulates NF-κB activity and thus suppresses SR-A expression, oxLDL uptake, and foam cell formation.
Collapse
Affiliation(s)
- Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center , Taipei, Taiwan, R.O.C
- Graduate Institute of Injury Prevention and Control, College of Public Health and Nutrition, Taipei Medical University , Taipei, Taiwan, R.O.C
| | - Yung-Hsi Kao
- Department of Life Sciences, National Central University , Jhongli, Taoyuan, Taiwan, R.O.C
| | - Li Jing
- Department of Emergency Medicine, The University of Illinois Hospital & Health Sciences System , Chicago, Illinois, United States
| | - Yi-Ping Chuang
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center , Taipei, Taiwan, R.O.C
| | - Wan-Lin Wu
- Department of Cell Biology and Neuroscience, College of Natural and Agricultural Sciences, University of California-Riverside , Riverside, California, United States
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center , Taipei, Taiwan, R.O.C
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center , Taipei, Taiwan, R.O.C
| | - Jenn-Haung Lai
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital , Tao-Yuan, Taiwan, R.O.C
| | - Ling-Jun Ho
- Institute of Cellular and System Medicine, National Health Research Institute , Zhunan, Taiwan, R.O.C
| | - Min-Chien Tsai
- Department of Physiology, National Defense Medical Center , Taipei, Taiwan, R.O.C
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center , Taipei, Taiwan, R.O.C
| |
Collapse
|
18
|
Vega-Benedetti AF, Saucedo C, Zavattari P, Vanni R, Zugaza JL, Parada LA. PLAGL1: an important player in diverse pathological processes. J Appl Genet 2016; 58:71-78. [PMID: 27311313 DOI: 10.1007/s13353-016-0355-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/02/2016] [Accepted: 06/02/2016] [Indexed: 12/23/2022]
Abstract
The PLAGL1 gene encodes a homonymous zinc finger protein that promotes cell cycle arrest and apoptosis through multiple pathways. The protein has been implicated in metabolic, genetic, and neoplastic illnesses, but the molecular mechanisms by which the protein PLAGL1 participates in such diverse processes remains to be elucidated. In this review, we focus mainly on the molecular biology of PLAGL1 and the relevance of its abnormalities to several pathological processes.
Collapse
Affiliation(s)
- Ana F Vega-Benedetti
- Institute of Experimental Pathology, UNSa-CONICET, Ave. Bolivia 5150, 4400, Salta, Argentina
| | - Cinthia Saucedo
- Institute of Experimental Pathology, UNSa-CONICET, Ave. Bolivia 5150, 4400, Salta, Argentina
| | - Patrizia Zavattari
- Biochemistry, Biology and Genetics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato SP 8, Km 0.700, 09042, Monserrato, Cagliari, Italy
| | - Roberta Vanni
- Biochemistry, Biology and Genetics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato SP 8, Km 0.700, 09042, Monserrato, Cagliari, Italy
| | - José L Zugaza
- IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.,Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, Building 205, Zamudio, Spain.,Department of Genetics, Physic Anthropology and Animal Physiology, Faculty of Medicine and Dentistry, University of the Basque Country, Barrio Sarriena s/n, 48940, Leioa, Spain
| | - Luis Antonio Parada
- Institute of Experimental Pathology, UNSa-CONICET, Ave. Bolivia 5150, 4400, Salta, Argentina.
| |
Collapse
|
19
|
Abstract
Imprinted genes are dosage sensitive, and their dysregulated expression is linked to disorders of growth and proliferation, including fetal and postnatal growth restriction. Common sequelae of growth disorders include neurodevelopmental defects, some of which are indirectly related to placental insufficiency. However, several growth-associated imprinted genes are also expressed in the embryonic CNS, in which their aberrant expression may more directly affect neurodevelopment. To test whether growth-associated genes influence neural lineage progression, we focused on the maternally imprinted gene Zac1. In humans, either loss or gain of ZAC1 expression is associated with reduced growth rates and intellectual disability. To test whether increased Zac1 expression directly perturbs neurodevelopment, we misexpressed Zac1 in murine neocortical progenitors. The effects were striking: Zac1 delayed the transition of apical radial glial cells to basal intermediate neuronal progenitors and postponed their subsequent differentiation into neurons. Zac1 misexpression also blocked neuronal migration, with Zac1-overexpressing neurons pausing more frequently and forming fewer neurite branches during the period when locomoting neurons undergo dynamic morphological transitions. Similar, albeit less striking, neuronal migration and morphological defects were observed on Zac1 knockdown, indicating that Zac1 levels must be regulated precisely. Finally, Zac1 controlled neuronal migration by regulating Pac1 transcription, a receptor for the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP). Pac1 and Zac1 loss- and gain-of-function presented as phenocopies, and overexpression of Pac1 rescued the Zac1 knockdown neuronal migration phenotype. Thus, dysregulated Zac1 expression has striking consequences on neocortical development, suggesting that misexpression of this transcription factor in the brain in certain growth disorders may contribute to neurocognitive deficits. Significance statement: Altered expression of imprinted genes is linked to cognitive dysfunction and neuropsychological disorders, such as Angelman and Prader-Willi syndromes, and autism spectrum disorder. Mouse models have also revealed the importance of imprinting for brain development, with chimeras generated with parthenogenetic (two maternal chromosomes) or androgenetic (two paternal chromosomes) cells displaying altered brain sizes and cellular defects. Despite these striking phenotypes, only a handful of imprinted genes are known or suspected to regulate brain development (e.g., Dlk1, Peg3, Ube3a, necdin, and Grb10). Herein we show that the maternally imprinted gene Zac1 is a critical regulator of neocortical development. Our studies are relevant because loss of 6q24 maternal imprinting in humans results in elevated ZAC1 expression, which has been associated with neurocognitive defects.
Collapse
|
20
|
Noriega-Reyes MY, Rivas-Torres MA, Oñate-Ocaña LF, Vallés AJ, Baranda-Avila N, Langley E. Novel role for PINX1 as a coregulator of nuclear hormone receptors. Mol Cell Endocrinol 2015; 414:9-18. [PMID: 26187699 DOI: 10.1016/j.mce.2015.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 11/20/2022]
Abstract
Estrogen receptor alpha (ERα) has an established role in breast cancer biology. Transcriptional activation by ERα is a multistep process influenced by coactivator and corepressor proteins. This work shows that Pin2 interacting protein 1 (PINX1) interacts with the N-terminal domain of ERα and functions as a corepressor of ERα. Furthermore, it represses both AF-1 and AF-2 transcriptional activities. Chromatin immunoprecipitation assays verified that the interaction between ERα and PINX1 occurs on E2 regulated promoters and enhanced expression of PINX1 deregulates the expression of a number of genes that have a role in cell growth and proliferation in breast cancer. PINX1 overexpression decreases estrogen mediated proliferation of breast cancer cell lines, while its depletion shows the opposite effect. Taken together, these data show a novel molecular mechanism for PINX1 as an attenuator of estrogen receptor activity in breast cancer cell lines, furthering its role as a tumor suppressor gene in breast cancer.
Collapse
Affiliation(s)
- Maria Yamilet Noriega-Reyes
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de Mexico. D.F., Mexico
| | - Miguel Angel Rivas-Torres
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de Mexico. D.F., Mexico
| | - Luis Fernando Oñate-Ocaña
- Departamento de Investigación Clínica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico
| | - Albert Jordan Vallés
- Institut de Biología Molecular de Barcelona (IBMB-CSIC) Parc Científic de Barcelona, Barcelona, Cataluña, España
| | - Noemi Baranda-Avila
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico
| | - Elizabeth Langley
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico.
| |
Collapse
|
21
|
Kowalczyk AE, Krazinski BE, Godlewski J, Kiewisz J, Kwiatkowski P, Sliwinska-Jewsiewicka A, Kiezun J, Wierzbicki PM, Bodek G, Sulik M, Kmiec Z. Altered expression of the PLAGL1 (ZAC1/LOT1) gene in colorectal cancer: Correlations to the clinicopathological parameters. Int J Oncol 2015; 47:951-62. [PMID: 26134521 DOI: 10.3892/ijo.2015.3067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/02/2015] [Indexed: 11/05/2022] Open
Abstract
Pleomorphic adenoma gene-like 1 gene (PLAGL1) encodes a zinc-finger nuclear transcription factor which promotes apoptosis and cell cycle arrest. Loss or downregulation of its expression has been observed in various human neoplasms. This study compared PLAGL1 expression in colorectal cancer (CRC) tissue and colon mucosa of healthy subjects at the mRNA and protein levels, and estimated its prognostic value. The PLAGL1 mRNA levels were also determined in CRC cell lines. We collected paired tumor tissue and unchanged mucosa of the large intestine from 121 CRC patients as well as 72 colon biopsies of healthy subjects obtained during screening colonoscopy. PLAGL1 mRNA levels were determined by quantitative PCR, while PLAGL1 protein expression was estimated by western blotting and immunohistochemistry. PLAGL1 mRNA level in tumor tissue was ~2-fold lower than in samples of corresponding unchanged tissues and biopsies of healthy colon mucosa. Downregulated expression of PLAGL1 mRNA was also observed in all tested CRC cell lines. Although the average content of PLAGL1 protein did not differ significantly between tumor and unchanged tissues of CRC patients or colon mucosa of healthy individuals, the decreased PLAGL1 protein levels in tumor specimens correlated with lymph node involvement, the presence of metastases and higher TNM disease stage. The PLAGL1 expression level did not correlate significantly with patient overall survival; however, the hazard ratio for patients whose tumor tissues showed reduced PLAGL1 immunohistochemical staining was twice higher than in patients with increased PLAGL1 immunoreactivity. In conclusion, these results suggest that dysregulation of PLAGL1 expression may be involved to some extent in the progression of CRC, but the so far collected patient survival data do not confirm applicability of the PLAGL1 expression level as a prognostic factor in CRC.
Collapse
Affiliation(s)
- Anna E Kowalczyk
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Bartlomiej E Krazinski
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Janusz Godlewski
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Jolanta Kiewisz
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Przemyslaw Kwiatkowski
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Agnieszka Sliwinska-Jewsiewicka
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Jacek Kiezun
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Piotr M Wierzbicki
- Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Gabriel Bodek
- In Vitro and Cell Biotechnology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-243 Olsztyn, Poland
| | - Marian Sulik
- Department of Pathomorphology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-561 Olsztyn, Poland
| | - Zbigniew Kmiec
- Department of Human Histology and Embryology, Faculty of Medical Sciences, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| |
Collapse
|
22
|
Daniel G, Schmidt-Edelkraut U, Spengler D, Hoffmann A. Imprinted Zac1 in neural stem cells. World J Stem Cells 2015; 7:300-314. [PMID: 25815116 PMCID: PMC4369488 DOI: 10.4252/wjsc.v7.i2.300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/24/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Neural stem cells (NSCs) and imprinted genes play an important role in brain development. On historical grounds, these two determinants have been largely studied independently of each other. Recent evidence suggests, however, that NSCs can reset select genomic imprints to prevent precocious depletion of the stem cell reservoir. Moreover, imprinted genes like the transcriptional regulator Zac1 can fine tune neuronal vs astroglial differentiation of NSCs. Zac1 binds in a sequence-specific manner to pro-neuronal and imprinted genes to confer transcriptional regulation and furthermore coregulates members of the p53-family in NSCs. At the genome scale, Zac1 is a central hub of an imprinted gene network comprising genes with an important role for NSC quiescence, proliferation and differentiation. Overall, transcriptional, epigenomic, and genomic mechanisms seem to coordinate the functional relationships of NSCs and imprinted genes from development to maturation, and possibly aging.
Collapse
|
23
|
Ansems M, Søndergaard JN, Sieuwerts AM, Looman MWG, Smid M, de Graaf AMA, de Weerd V, Zuidscherwoude M, Foekens JA, Martens JWM, Adema GJ. DC-SCRIPT is a novel regulator of the tumor suppressor gene CDKN2B and induces cell cycle arrest in ERα-positive breast cancer cells. Breast Cancer Res Treat 2015; 149:693-703. [PMID: 25663546 PMCID: PMC4326655 DOI: 10.1007/s10549-015-3281-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/18/2015] [Indexed: 01/07/2023]
Abstract
Breast cancer is one of the most common causes of cancer-related deaths in women. The estrogen receptor (ERα) is well known for having growth promoting effects in breast cancer. Recently, we have identified DC-SCRIPT (ZNF366) as a co-suppressor of ERα and as a strong and independent prognostic marker in ESR1 (ERα gene)-positive breast cancer patients. In this study, we further investigated the molecular mechanism on how DC-SCRIPT inhibits breast cancer cell growth. DC-SCRIPT mRNA levels from 190 primary ESR1-positive breast tumors were related to global gene expression, followed by gene ontology and pathway analysis. The effect of DC-SCRIPT on breast cancer cell growth and cell cycle arrest was investigated using novel DC-SCRIPT-inducible MCF7 breast cancer cell lines. Genome-wide expression profiling of DC-SCRIPT-expressing MCF7 cells was performed to investigate the effect of DC-SCRIPT on cell cycle-related gene expression. Findings were validated by real-time PCR in a cohort of 1,132 ESR1-positive breast cancer patients. In the primary ESR1-positive breast tumors, DC-SCRIPT expression negatively correlated with several cell cycle gene ontologies and pathways. DC-SCRIPT expression strongly reduced breast cancer cell growth in vitro, breast tumor growth in vivo, and induced cell cycle arrest. In addition, in the presence of DC-SCRIPT, multiple cell cycles related genes were differentially expressed including the tumor suppressor gene CDKN2B. Moreover, in 1,132 primary ESR1-positive breast tumors, DC-SCRIPT expression also correlated with CDKN2B expression. Collectively, these data show that DC-SCRIPT acts as a novel regulator of CDKN2B and induces cell cycle arrest in ESR1-positive breast cancer cells.
Collapse
Affiliation(s)
- Marleen Ansems
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chan JYH, Chen YC, Liu ST, Chou WY, Ho CL, Huang SM. Characterization of a new mouse p53 variant: loss-of-function and gain-of-function. J Biomed Sci 2014; 21:40. [PMID: 24884657 PMCID: PMC4022406 DOI: 10.1186/1423-0127-21-40] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 05/05/2014] [Indexed: 11/17/2022] Open
Abstract
Background p53 is a major tumor suppressor that is inactivated in over 50% of human cancer types through either mutation or inactivating interactions with viral or cellular proteins. The uncertainties around the link between p53 status, therapeutic response, and outcome in cancer suggest that additional factors may be involved. p53 isoforms that are generated via the alternative splicing pathway may be promising candidates for further investigation. Result In this study, we report one new p53 protein with two internally deleted regions, resulting in one deleted amino acid fragment (from amino acid residues 42 to 89) and one reading frame-shift region (from amino acid residues 90-120) compared to wild-type p53. The functional status of the new p53 protein, which has a defect in its proline-rich and N-terminal DNA-binding domains, was characterized as possessing an intact conformation, exhibiting no transactivation activity, exerting a dominant-negative effect and an interacting with a coactivator with an arginine methyltransferase activity. Conclusion Taken together, our findings provide valuable information about the structure and function of p53 for the regulation of transactivation activity and cellular protein-protein interactions. Furthermore, natural p53 isoforms will help us understand the functional roles of the p53 family and potential therapeutics for p53-dependent cancers.
Collapse
Affiliation(s)
| | | | | | - Wei-Yuan Chou
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan.
| | | | | |
Collapse
|
25
|
Mirowska A, Sledzinski T, Smolenski RT, Swierczynski J. Down-regulation of Zac1 gene expression in rat white adipose tissue by androgens. J Steroid Biochem Mol Biol 2014; 140:63-70. [PMID: 24316431 DOI: 10.1016/j.jsbmb.2013.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 11/22/2013] [Accepted: 11/24/2013] [Indexed: 12/12/2022]
Abstract
ZAC1 is a zinc-finger protein transcription factor, a transcriptional cofactor for nuclear receptors, and a co-activator of nuclear receptors, which interacts with multiple signaling pathways affecting apoptosis, cell cycle arrest, and metabolism. Some data suggest that ZAC1 regulates the expression of genes associated with function of adipose tissue. Since there is no information about the levels of Zac1 gene expression in white adipose tissue (WAT), and the expression of several genes associated with metabolic function of WAT is significantly lower in male than female animals, we have examined: (a) the relative ZAC1 mRNA levels in some organs/tissues, including three main depots of WAT, in 3-month-old male rats; (b) the relative ZAC1 mRNA levels in WAT of male and female rats; (c) the effect of orchidectomy and orchidectomy with concomitant testosterone treatment on ZAC1 mRNA and protein levels; (d) the effect of ovariectomy and ovariectomy with concomitant 17β-estradiol treatment on ZAC1 mRNA levels; (e) the effect of dihydrotestosterone on ZAC1 mRNA levels in isolated adipocytes. Our results indicate that: (a) ZAC1 mRNA levels are relatively high in WAT in comparison with other organs/tissues; (b) ZAC1 mRNA levels in subcutaneous WAT are approximately 2-fold lower than in epididymal and retroperitoneal adipose tissue; (c) ZAC1 mRNA levels in WAT of adult female rats are approximately 2-fold higher than in male rats; (d) testosterone is inversely related to ZAC1 mRNA and protein levels in WAT of male rats; and (e) dihydrotestosterone decreases the ZAC1 mRNA levels in adipocytes in dose dependent manner. In conclusion, Zac1 gene is highly expressed in white adipose tissue of adult rats. Androgens could play an important role in down-regulation of the ZAC1 mRNA and protein levels in rats.
Collapse
Affiliation(s)
- Agnieszka Mirowska
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | | | | |
Collapse
|
26
|
Abstract
Imprinted genes play a critical role in brain development and mental health, although the underlying molecular and cellular mechanisms remain incompletely understood. The family of basic helix-loop-helix (bHLH) proteins directs the proliferation, differentiation, and specification of distinct neuronal progenitor populations. Here, we identified the bHLH factor gene Tcf4 as a direct target gene of Zac1/Plagl1, a maternally imprinted transcriptional regulator, during early neurogenesis. Zac1 and Tcf4 expression levels concomitantly increased during neuronal progenitor differentiation; moreover, Zac1 interacts with two cis-regulatory elements in the Tcf4 gene locus, and these elements together confer synergistic activation of the Tcf4 gene. Tcf4 upregulation enhances the expression of the cyclin-dependent kinase inhibitor gene p57(Kip2), a paternally imprinted Tcf4 target gene, and increases the number of cells in G1 phase. Overall, we show that Zac1 controls cell cycle arrest function in neuronal progenitors through induction of p57(Kip2) via Tcf4 and provide evidence for cooperation between imprinted genes and a bHLH factor in early neurodevelopment.
Collapse
|
27
|
Hisanaga-Oishi Y, Nishiwaki-Ueda Y, Nojima K, Ueda H. Analysis of the expression of candidate genes for type 1 diabetes susceptibility in T cells. Endocr J 2014; 61:577-88. [PMID: 24705559 DOI: 10.1507/endocrj.ej14-0002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Type 1 diabetes is characterized by T-cell-mediated autoimmune destruction of pancreatic β-cells. Currently, approximately 50 type 1 diabetes susceptibility genes or chromosomal regions have been identified. However, the functions of type 1 diabetes susceptibility genes in T cells are elusive. In this study, we evaluated the correlation between type 1 diabetes susceptibility genes and T-cell signaling. The expression levels of 22 candidate type 1 diabetes susceptibility genes in T cells from nonobese diabetic (NOD), control C57BL/6 (B6), and NOD-control F1 hybrid mice were analyzed in response to 2 key immunoregulatory cytokines: interleukin-2 (IL-2) and transforming growth factor β (TGF-β). Exogenous gene expression studies were also performed in EL4 and Jurkat E6.1 T-cell lines. Significant differences in the expression of Clec16a, Dlk1, Il2, Ptpn22, Rnls, and Zac1 (also known as Plagl1) were observed in T cells derived from the 3 strains of mice, and TGF-β differentially influenced the expression of Ctla4, Foxp3, Il2, Ptpn22, Sh2b3, and Zac1. We found that TGF-β induced Zac1 expression in both primary T cells and EL4 cells and that exogenous expression of Zac1 and ZAC1 in T-cell lines altered the expression of Il2 and DLK1, respectively. The results of our study indicate the possibility that additional genetic pathways underlying type 1 diabetes susceptibility, including those involving Clec16a, Dlk1, Rnls, Sh2b3, and Zac1 under IL-2 and TGF-β signaling in T cells, may be shared between human and NOD mice.
Collapse
Affiliation(s)
- Yuko Hisanaga-Oishi
- Department of Molecular Endocrinology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | | | | | | |
Collapse
|
28
|
Dual roles for lysine 490 of promyelocytic leukemia protein in the transactivation of glucocorticoid receptor-interacting protein 1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1799-810. [PMID: 23542129 DOI: 10.1016/j.bbamcr.2013.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 03/12/2013] [Accepted: 03/18/2013] [Indexed: 01/20/2023]
Abstract
Glucocorticoid receptor-interacting protein 1 (GRIP1), a p160 family nuclear receptor co-activator protein, has three activation domains that recruit at least three secondary co-activators: CBP/p300, co-activator-associated arginine methyltransferase 1, and coiled-coil co-activator, which exhibits histone acetyltransferase and/or arginine methyltransferase activities. The regulatory mechanisms underlying the co-activation functions of GRIP1, which associates with promyelocytic leukemia protein (PML) in PML-nuclear bodies, are not well-understood. This study showed that PML specifically and dramatically enhanced the C-terminal transactivation activity of GRIP1 by directly binding to GRIP1 but only when it was sumoylated. Most of the transactivation activity resided in the N-terminal PML regions that are conserved among isoforms. Three N-terminal sumoylation residues (Lys 65, 160, and 490) exhibited differential roles in the regulation of GRIP1 activity, and the sumoylation of Lys 490 acted as the primary nuclear localization signal of PML. While GRIP1 transactivation was stimulated to a similar degree by PML (K490R), located in the nucleus, and wild-type PML, PML (K490D) and the C-truncated mutant PML1-489 both displayed an epinuclear localization and were mostly inactive in stimulating GRIP. Based on these data, nuclear foci, nuclear localization, and the sumoylation status of Lys 490 were not essential for the enhancement of GRIP1 activity by PML, but the charge status of Lys 490 was important for subcellular localization of PML and cross-talk between its N- and C-terminal regions to modulate transcriptional activation. Taken together, these results provide insight into the regulatory mechanisms of PML that control the functional activities of GRIP1.
Collapse
|
29
|
Iglesias-Platas I, Court F, Camprubi C, Sparago A, Guillaumet-Adkins A, Martin-Trujillo A, Riccio A, Moore GE, Monk D. Imprinting at the PLAGL1 domain is contained within a 70-kb CTCF/cohesin-mediated non-allelic chromatin loop. Nucleic Acids Res 2013; 41:2171-9. [PMID: 23295672 PMCID: PMC3575839 DOI: 10.1093/nar/gks1355] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/04/2012] [Accepted: 12/06/2012] [Indexed: 12/22/2022] Open
Abstract
Paternal duplications of chromosome 6q24, a region that contains the imprinted PLAGL1 and HYMAI transcripts, are associated with transient neonatal diabetes mellitus. A common feature of imprinted genes is that they tend to cluster together, presumably as a result of sharing common cis-acting regulatory elements. To determine the extent of this imprinted cluster in human and mouse, we have undertaken a systematic analysis of allelic expression and DNA methylation of the genes mapping within an ∼1.4-Mb region flanking PLAGL1/Plagl1. We confirm that all nine neighbouring genes are biallelically expressed in both species. In human we identify two novel paternally expressed PLAGL1 coding transcripts that originate from unique promoter regions. Chromatin immunoprecipitation for CTCF and the cohesin subunits RAD21 and SMC3 reveals evolutionarily conserved binding sites within unmethylated regions ∼5 kb downstream of the PLAGL1 differentially methylated region and within the PLAGL1 3' untranslated region (UTR). Higher-order chromatin looping occurs between these regions in both expressing and non-expressing tissues, forming a non-allelic chromatin loop around the PLAGL1/Plagl1 gene. In placenta and brain tissues, we identify an additional interaction between the PLAGL1 P3/P4 promoters and the unmethylated element downstream of the PLAGL1 differentially methylated region that we propose facilitates imprinted expression of these alternative isoforms.
Collapse
Affiliation(s)
- Isabel Iglesias-Platas
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - Franck Court
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - Cristina Camprubi
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - Angela Sparago
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - Amy Guillaumet-Adkins
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - Alex Martin-Trujillo
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - Andrea Riccio
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - Gudrun E. Moore
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| | - David Monk
- Servicio de Neonatología, Hospital Sant Joan de Déu (HSJD), Fundació Sant Joan de Déu, 08950 Barcelona, Spain, Imprinting and Cancer Group, Epigenetics and Cancer Biology Program (PEBC), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, 08907 Barcelona, Spain, Department of Environmental Science, Second University of Naples, 81100 Caserta, Italy, Institute of Genetics and Biophysics ‘Adriano. Buzzati-Traverso,’ CNR, 80131 Naples, Italy and Fetal Growth and Development Group, Clinical and Molecular Genetics Unit, Institute of Child Health, University College London, London, WC1N 1EH UK
| |
Collapse
|
30
|
Transient neonatal diabetes mellitus gene Zac1 impairs insulin secretion in mice through Rasgrf1. Mol Cell Biol 2012; 32:2549-60. [PMID: 22547676 DOI: 10.1128/mcb.06637-11] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The biallelic expression of the imprinted gene ZAC1/PLAGL1 underlies ≈ 60% of all cases of transient neonatal diabetes mellitus (TNDM) that present with low perinatal insulin secretion. Molecular targets of ZAC1 misexpression in pancreatic β cells are unknown. Here, we identified the guanine nucleotide exchange factor Rasgrf1 as a direct Zac1/Plagl1 target gene in murine β cells. Doubling Zac1 expression reduced Rasgrf1 expression, the stimulus-induced activation of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways, and, ultimately, insulin secretion. Normalizing Rasgrf1 expression reversed this phenotype. Moreover, the transplantation of Zac1-overexpressing β cells failed to reinstate euglycemia in experimental diabetic mice. In contrast, Zac1 expression did not interfere with the signaling of the glucagon-like peptide 1 receptor (GLP-1R), and the GLP-1 analog liraglutide improved hyperglycemia in transplanted experimental diabetic mice. This study unravels a mechanism contributing to insufficient perinatal insulin secretion in TNDM and raises new prospects for therapy.
Collapse
|
31
|
Specific changes in the expression of imprinted genes in prostate cancer--implications for cancer progression and epigenetic regulation. Asian J Androl 2012; 14:436-50. [PMID: 22367183 DOI: 10.1038/aja.2011.160] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Epigenetic dysregulation comprising DNA hypermethylation and hypomethylation, enhancer of zeste homologue 2 (EZH2) overexpression and altered patterns of histone modifications is associated with the progression of prostate cancer. DNA methylation, EZH2 and histone modifications also ensure the parental-specific monoallelic expression of at least 62 imprinted genes. Although it is therefore tempting to speculate that epigenetic dysregulation may extend to imprinted genes, expression changes in cancerous prostates are only well documented for insulin-like growth factor 2 (IGF2). A literature and database survey on imprinted genes in prostate cancer suggests that the expression of most imprinted genes remains unchanged despite global disturbances in epigenetic mechanisms. Instead, selective genetic and epigenetic changes appear to lead to the inactivation of a sub-network of imprinted genes, which might function in the prostate to limit cell growth induced via the PI3K/Akt pathway, modulate androgen responses and regulate differentiation. Whereas dysregulation of IGF2 may constitute an early change in prostate carcinogenesis, inactivation of this imprinted gene network is rather associated with cancer progression.
Collapse
|
32
|
Deducing the temporal order of cofactor function in ligand-regulated gene transcription: theory and experimental verification. PLoS One 2012; 7:e30225. [PMID: 22272313 PMCID: PMC3260260 DOI: 10.1371/journal.pone.0030225] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/14/2011] [Indexed: 11/19/2022] Open
Abstract
Cofactors are intimately involved in steroid-regulated gene expression. Two critical questions are (1) the steps at which cofactors exert their biological activities and (2) the nature of that activity. Here we show that a new mathematical theory of steroid hormone action can be used to deduce the kinetic properties and reaction sequence position for the functioning of any two cofactors relative to a concentration limiting step (CLS) and to each other. The predictions of the theory, which can be applied using graphical methods similar to those of enzyme kinetics, are validated by obtaining internally consistent data for pair-wise analyses of three cofactors (TIF2, sSMRT, and NCoR) in U2OS cells. The analysis of TIF2 and sSMRT actions on GR-induction of an endogenous gene gave results identical to those with an exogenous reporter. Thus new tools to determine previously unobtainable information about the nature and position of cofactor action in any process displaying first-order Hill plot kinetics are now available.
Collapse
|
33
|
Liu ST, Chang YL, Wang WM, Chung MH, Lin WS, Chou WY, Huang SM. A non-covalent interaction between small ubiquitin-like modifier-1 and Zac1 regulates Zac1 cellular functions. Int J Biochem Cell Biol 2011; 44:547-55. [PMID: 22227369 DOI: 10.1016/j.biocel.2011.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 12/03/2011] [Accepted: 12/19/2011] [Indexed: 12/11/2022]
Abstract
Zac1, a zinc-finger protein that regulates apoptosis and cell cycle arrest 1, such as p53, can induce cell-cycle arrest and apoptosis. The transactivation and coactivation functions of Zac1 may occur at non-promyelocytic leukemia nuclear body (PML-NB) sites in the presence of other PML-NB components, including ubiquitin-conjugating 9 (Ubc9). It is unclear whether post-translational modification of Zac1 by the small ubiquitin-like modifier SUMO plays a role in the coactivation functions of Zac1 for the regulation of the p21 gene. Mutagenesis experiments revealed that the two SUMO-binding lysine residues of Zac1, K237 and K424, repress the transactivation activity of Zac1. Studies using a SUMO-1 C-terminal di-glycine motif mutant that is deficient in the ability to form covalent bonds with lysines, SUMO-1 (GA), and a dominant-negative Ubc9 construct (C93S) indicated that SUMO-1 might regulate Zac1 transactivation and coactivation via a non-covalent interaction. Unlike the wild-type Zac1, which induced apoptosis, the Zac1 (K237/424R) double mutant had the ability to induce autophagy. The functional role of p21 remains to be investigated. SUMO-1 selectively suppressed the induction of the p21 gene and protein by wild-type Zac1 but not by the Zac1 (K237/424R) double mutant. Moreover, wild-type Ubc9 but not Ubc9 (C93S) further potentiated the suppression of SUMO-1 in all Zac1-induced p21 promoter activities. Our data reveal that p21 may be an important factor for the prevention of Zac1-induced apoptosis without affecting autophagosome formation. This work indicates that Zac1 functions are regulated, at least in part, via non-covalent interactions with SUMO-1 for the induction of p21, which is important for the modulation of apoptosis.
Collapse
Affiliation(s)
- Shu-Ting Liu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
34
|
Friedlander TW, Roy R, Tomlins SA, Ngo VT, Kobayashi Y, Azameera A, Rubin MA, Pienta KJ, Chinnaiyan A, Ittmann MM, Ryan CJ, Paris PL. Common structural and epigenetic changes in the genome of castration-resistant prostate cancer. Cancer Res 2011; 72:616-25. [PMID: 22158653 DOI: 10.1158/0008-5472.can-11-2079] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Progression of primary prostate cancer to castration-resistant prostate cancer (CRPC) is associated with numerous genetic and epigenetic alterations that are thought to promote survival at metastatic sites. In this study, we investigated gene copy number and CpG methylation status in CRPC to gain insight into specific pathophysiologic pathways that are active in this advanced form of prostate cancer. Our analysis defined and validated 495 genes exhibiting significant differences in CRPC in gene copy number, including gains in androgen receptor (AR) and losses of PTEN and retinoblastoma 1 (RB1). Significant copy number differences existed between tumors with or without AR gene amplification, including a common loss of AR repressors in AR-unamplified tumors. Simultaneous gene methylation and allelic deletion occurred frequently in RB1 and HSD17B2, the latter of which is involved in testosterone metabolism. Lastly, genomic DNA from most CRPC was hypermethylated compared with benign prostate tissue. Our findings establish a comprehensive methylation signature that couples epigenomic and structural analyses, thereby offering insights into the genomic alterations in CRPC that are associated with a circumvention of hormonal therapy. Genes identified in this integrated genomic study point to new drug targets in CRPC, an incurable disease state which remains the chief therapeutic challenge.
Collapse
Affiliation(s)
- Terence W Friedlander
- Division on Genitourinary Medical Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Liu PY, Hsieh TY, Liu ST, Chang YL, Lin WS, Wang WM, Huang SM. Zac1, an Sp1-like protein, regulates human p21WAF1/Cip1 gene expression in HeLa cells. Exp Cell Res 2011; 317:2925-37. [DOI: 10.1016/j.yexcr.2011.09.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 09/26/2011] [Accepted: 09/30/2011] [Indexed: 11/26/2022]
|
36
|
|
37
|
Wang WM, Liu ST, Huang SM, Lin WS, Chen SG, Chang YL. Zac1 functional interactions mediate AP-1 transcriptional activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:2050-60. [DOI: 10.1016/j.bbamcr.2011.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 08/05/2011] [Accepted: 08/05/2011] [Indexed: 02/06/2023]
|
38
|
Cooper C, Vincett D, Yan Y, Hamedani MK, Myal Y, Leygue E. Steroid receptor RNA activator bi-faceted genetic system: Heads or Tails? Biochimie 2011; 93:1973-80. [DOI: 10.1016/j.biochi.2011.07.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 07/04/2011] [Indexed: 11/17/2022]
|
39
|
Different roles of p53 in the regulation of DNA damage caused by 1,2-heteroannelated anthraquinones and doxorubicin. Int J Biochem Cell Biol 2011; 43:1720-8. [PMID: 21856437 DOI: 10.1016/j.biocel.2011.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 08/03/2011] [Accepted: 08/08/2011] [Indexed: 12/11/2022]
Abstract
The anthracyclin antibiotic agent doxorubicin (DXR) has been widely used as a chemotherapeutic drug for more than 40 years, but its clinical use has been limited by its cardiotoxicity. The mechanism of action of DXR remains uncertain and controversial. A series of 1,2-heteroannelated anthraquinones and anthra[1,2-d]imidazole-6,11-dione compounds were synthesized and their cytotoxicity profiles were analyzed using the National Cancer Institute 60 (NCI 60) platform and human telomerase inhibition assays. In the current study, three of the 1,2-heteroannelated anthraquinones, NSC745795, NSC745885 and NSC745887, were found to differ from each other with respect to their effects on cell cycle regulation, apoptosis, autophagy, senescence and their abilities to induce DNA damage. The differences depended on the presence or absence of a heterocyclic moiety, which suggested that the differences were due, at least in part, to differential effects on specific cellular targets, such as p53. In contrast to DXR, which induced p53 expression, treatment with NSC745885 resulted in the degradation of several proteins, including p53, via proteasome-dependent and -independent pathways in HeLa cells. These results provide insights into the molecular mechanisms governing cell inhibition by 1,2-heteroannelated anthraquinone derivatives and suggest that these mechanisms could serve as the basis for new structure-based drug designs.
Collapse
|
40
|
Chung SH, Marzban H, Aldinger K, Dixit R, Millen K, Schuurmans C, Hawkes R. Zac1 plays a key role in the development of specific neuronal subsets in the mouse cerebellum. Neural Dev 2011. [PMID: 21592321 DOI: 10.1186/1749‐8104‐6‐25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND The cerebellum is composed of a diverse array of neuronal subtypes. Here we have used a candidate approach to identify Zac1, a tumor suppressor gene encoding a zinc finger transcription factor, as a new player in the transcriptional network required for the development of a specific subset of cerebellar nuclei and a population of Golgi cells in the cerebellar cortex. RESULTS We found that Zac1 has a complex expression profile in the developing cerebellum, including in two proliferating progenitor populations; the cerebellar ventricular zone and the external granular layer overlying posterior cerebellar lobules IX and X. Zac1 is also expressed in some postmitotic cerebellar neurons, including a subset of GABAergic interneurons in the medial cerebellar nuclei. Notably, GABAergic interneurons in the cerebellar nuclei are derived from the cerebellar ventricular zone, where Zac1 is also expressed, consistent with a lineage relationship between these two Zac1+ populations. Zac1 is also expressed in a small subset of cells in the posterior vermis, including some neurogranin-immunoreactive (NG+) Golgi cells, which, based on short-term birthdating, are derived from the EGL, where Zac1 is also expressed. However, Zac1+ cells and NG+ Golgi cells in the cerebellar cortex also display unique properties, as they are generated within different, albeit overlapping, time windows. Finally, consistent with the expression profile of Zac1, two conspicuous abnormalities were found in the cerebellum of Zac1 null mice: the medial cerebellar nuclei, and not the others, were significantly reduced in size; and the number of Golgi cells in cerebellar lobule IX was reduced by approximately 60% compared to wild-type littermates. CONCLUSIONS The data presented here indicate that the tumor suppressor gene Zac1 is expressed in a complex fashion in the developing cerebellum, including in two dividing progenitor populations and in specific subsets of postmitotic neurons, including Golgi cells and GABAergic neurons in the medial nuclei, which require Zac1 for their differentiation. We thus conclude that Zac1 is a critical regulator of normal cerebellar development, adding a new transcriptional regulator to the growing list of factors involved in generating neuronal diversity in the developing cerebellum.
Collapse
Affiliation(s)
- Seung-Hyuk Chung
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| | | | | | | | | | | | | |
Collapse
|
41
|
Chung SH, Marzban H, Aldinger K, Dixit R, Millen K, Schuurmans C, Hawkes R. Zac1 plays a key role in the development of specific neuronal subsets in the mouse cerebellum. Neural Dev 2011; 6:25. [PMID: 21592321 PMCID: PMC3113315 DOI: 10.1186/1749-8104-6-25] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 05/18/2011] [Indexed: 11/20/2022] Open
Abstract
Background The cerebellum is composed of a diverse array of neuronal subtypes. Here we have used a candidate approach to identify Zac1, a tumor suppressor gene encoding a zinc finger transcription factor, as a new player in the transcriptional network required for the development of a specific subset of cerebellar nuclei and a population of Golgi cells in the cerebellar cortex. Results We found that Zac1 has a complex expression profile in the developing cerebellum, including in two proliferating progenitor populations; the cerebellar ventricular zone and the external granular layer overlying posterior cerebellar lobules IX and X. Zac1 is also expressed in some postmitotic cerebellar neurons, including a subset of GABAergic interneurons in the medial cerebellar nuclei. Notably, GABAergic interneurons in the cerebellar nuclei are derived from the cerebellar ventricular zone, where Zac1 is also expressed, consistent with a lineage relationship between these two Zac1+ populations. Zac1 is also expressed in a small subset of cells in the posterior vermis, including some neurogranin-immunoreactive (NG+) Golgi cells, which, based on short-term birthdating, are derived from the EGL, where Zac1 is also expressed. However, Zac1+ cells and NG+ Golgi cells in the cerebellar cortex also display unique properties, as they are generated within different, albeit overlapping, time windows. Finally, consistent with the expression profile of Zac1, two conspicuous abnormalities were found in the cerebellum of Zac1 null mice: the medial cerebellar nuclei, and not the others, were significantly reduced in size; and the number of Golgi cells in cerebellar lobule IX was reduced by approximately 60% compared to wild-type littermates. Conclusions The data presented here indicate that the tumor suppressor gene Zac1 is expressed in a complex fashion in the developing cerebellum, including in two dividing progenitor populations and in specific subsets of postmitotic neurons, including Golgi cells and GABAergic neurons in the medial nuclei, which require Zac1 for their differentiation. We thus conclude that Zac1 is a critical regulator of normal cerebellar development, adding a new transcriptional regulator to the growing list of factors involved in generating neuronal diversity in the developing cerebellum.
Collapse
Affiliation(s)
- Seung-Hyuk Chung
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| | | | | | | | | | | | | |
Collapse
|
42
|
Zac1 is a histone acetylation-regulated NF-κB suppressor that mediates histone deacetylase inhibitor-induced apoptosis. Cell Death Differ 2011; 18:1825-35. [PMID: 21546906 DOI: 10.1038/cdd.2011.51] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors are a class of promising anticancer reagents. They are able to induce apoptosis in embryonic carcinoma (EC) cells. However, the underlying mechanism remains poorly understood. Here we show that increased expression of zinc-finger protein regulator of apoptosis and cell-cycle arrest (Zac1) is implicated in HDAC inhibitor-induced apoptosis in F9 and P19 EC cells. By chromatin immunoprecipitation analysis we identified that increased Zac1 expression is mediated by histone acetylation of the Zac1 promoter region. Knockdown of Zac1 inhibited HDAC inhibitor-induced cell apoptosis. Moreover, HDAC inhibitors repressed nuclear factor-κB (NF-κB) activity, and this effect is abrogated by Zac1 knockdown. Consistently, Zac1 overexpression suppressed cellular NF-κB activity. Further investigation showed that Zac1 inhibits NF-κB activity by interacting with the C-terminus of the p65 subunit, which suppresses the phosphorylation of p65 at Ser468 and Ser536 residues. These results indicate that Zac1 is a histone acetylation-regulated suppressor of NF-κB, which is induced and implicated in HDAC inhibitor-mediated EC cell apoptosis.
Collapse
|
43
|
Du X, Rousseau M, Ounissi-Benkalha H, Marchand L, Jetha A, Paraskevas S, Goodyer C, Polychronakos C. Differential expression pattern of ZAC in developing mouse and human pancreas. J Mol Histol 2011; 42:129-36. [PMID: 21305342 DOI: 10.1007/s10735-011-9315-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 01/20/2011] [Indexed: 01/11/2023]
Abstract
ZAC is a transcription factor and cofactor, a strong candidate for transient neonatal diabetes mellitus (TNDM). TNDM involves impaired beta-cell development and is probably due to a double dose of ZAC, which is normally expressed only from the paternal copy. ZAC and Zac1 (its mouse orthologue) are strongly expressed in the proliferating progenitor/stem cells in many systems and also in some differentiated sites in human and mouse, suggesting a dual role in cell proliferation and differentiation control. Little is known about its expression in developing pancreas, the organ affected in TNDM. In this study, we examined ZAC/Zac1 expression in developing mouse and human pancreas by real-time PCR and dual in situ hybridization and immunofluorescence. Overall pancreatic expression drastically declined during gestation and early post-natal life in the mouse, and between the second trimester and adult in the human. Zac1 was predominantly expressed in mesenchyme in the mouse embryo, while ZAC was specifically expressed in islets of the human fetus. Thus, ZAC/Zac1 may play different roles in mouse and human pancreas development. The specific expression of ZAC in the human fetal beta-cells supports it as the gene involved in TNDM and the different expression pattern of Zac1 in mice from human may explain the much milder phenotype in the mouse model of ZAC double dose.
Collapse
Affiliation(s)
- Xiaoyu Du
- Division of Pediatric Endocrinology, McGill University Health Centre Research Institute, Children's Hospital, 4060 St. Catherine west, PT-414, Montreal, QC, H3Z 2Z3, Canada
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Viral induction of Zac1b through TLR3- and IRF3-dependent pathways. Mol Immunol 2010; 48:119-27. [DOI: 10.1016/j.molimm.2010.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 08/20/2010] [Accepted: 09/14/2010] [Indexed: 01/22/2023]
|
45
|
Chooniedass-Kothari S, Hamedani MK, Auge C, Wang X, Carascossa S, Yan Y, Cooper C, Vincett D, Myal Y, Jalaguier S, Cavailles V, Leygue E. The steroid receptor RNA activator protein is recruited to promoter regions and acts as a transcriptional repressor. FEBS Lett 2010; 584:2218-24. [PMID: 20398657 DOI: 10.1016/j.febslet.2010.04.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 03/31/2010] [Accepted: 04/08/2010] [Indexed: 01/23/2023]
Abstract
Products of the steroid receptor RNA activator (SRA1) gene have the unusual property to function both at the RNA and the protein levels. SRA-RNA has long been known to increase the activity of multiple nuclear receptors. It has more recently been proposed than steroid receptor RNA activator protein (SRAP) also modulates steroid receptors activity. Herein, we show for the first time that SRAP physically interacts with multiple transcription factors and is recruited to specific promoter regions. Artificially recruiting SRAP to the promoter of a luciferase reporter gene under the control of the strong transcriptional activator VP16 leads to a decrease in transcription. Altogether we propose that SRAP could be a new transcriptional regulator, able to function as a repressor through direct association with promoters.
Collapse
|
46
|
Czubryt MP, Lamoureux L, Ramjiawan A, Abrenica B, Jangamreddy J, Swan K. Regulation of cardiomyocyte Glut4 expression by ZAC1. J Biol Chem 2010; 285:16942-50. [PMID: 20363751 DOI: 10.1074/jbc.m109.097246] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The transcription factor ZAC1 is expressed in a variety of tissues including the developing heart, but its physiological role is unclear. We examined the role of ZAC1 in regulating expression of the insulin-responsive glucose transporter GLUT4 and whether ZAC1 expression is altered in cardiomyocyte hypertrophy. We demonstrated expression of Zac1 mRNA and protein in rat cardiomyocytes by PCR and Western blotting, respectively. Using a combination of chromatin immunoprecipitation and luciferase assays, we showed that ZAC1 regulates Glut4 expression via a specific binding site in the Glut4 promoter. Overexpression of ZAC1 increased Glut4 mRNA and protein expression and resulted in increased glucose uptake in cardiomyocytes as determined by a fluorescent analog uptake assay. Induction of hypertrophy by phenylephrine or isoproterenol resulted in increased Zac1 expression. We identified a novel putative promoter in the Zac1 gene and demonstrated increased binding of MEF2 to this promoter in response to hypertrophic stimulation. MEF2 regulated transactivation of the Zac1 promoter and ZAC1 protein expression. This work identifies ZAC1 as a novel and previously unknown regulator of cardiomyocyte Glut4 expression and glucose uptake. Our results also implicate MEF2 as a regulator of ZAC1 expression in response to induction of hypertrophy.
Collapse
Affiliation(s)
- Michael P Czubryt
- Department of Physiology, University of Manitoba, Institute of Cardiovascular Sciences, St Boniface General Hospital Research Centre, Winnipeg, Manitoba R2H 2A6, Canada
| | | | | | | | | | | |
Collapse
|
47
|
Guyot R, Vincent S, Bertin J, Samarut J, Ravel-Chapuis P. The transforming acidic coiled coil (TACC1) protein modulates the transcriptional activity of the nuclear receptors TR and RAR. BMC Mol Biol 2010; 11:3. [PMID: 20078863 PMCID: PMC2822774 DOI: 10.1186/1471-2199-11-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 01/15/2010] [Indexed: 11/23/2022] Open
Abstract
Background The transcriptional activity of Nuclear hormone Receptors (NRs) is regulated by interaction with coactivator or corepressor proteins. Many of these cofactors have been shown to have a misregulated expression or to show a subcellular mislocalization in cancer cell lines or primary tumors. Therefore they can be factors involved in the process of oncogenesis. Results We describe a novel NR coregulator, TACC1, which belongs to the Transforming Acidic Coiled Coil (TACC) family. The interaction of TACC1 with Thyroid Hormone Receptors (TR) and several other NRs has been shown in a yeast two-hybrid screen and confirmed by GST pulldown, colocalization and co-immunoprecipitation experiments. TACC1 interacts preferentially with unliganded NRs. In F9 cells, endogenous TACC1 localized in the chromatin-enriched fraction of the nucleus and interacted with Retinoid Acid Receptors (RARα) in the nucleus. TACC1 depletion in the cell led to decreased RARα and TRα ligand-dependent transcriptional activity and to delocalization of TR from the nucleus to the cytoplasm. Conclusions From these experimental studies we propose that TACC1 might be a scaffold protein building up a transcriptional complex around the NRs we studied. This function of TACC1 might account for its involvement in several forms of tumour development.
Collapse
Affiliation(s)
- Romain Guyot
- Institut de Génomique Fonctionnelle de Lyon, Universitéde Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364 Lyon Cedex 07, France
| | | | | | | | | |
Collapse
|
48
|
Xu J, Wu RC, O’Malley BW. Normal and cancer-related functions of the p160 steroid receptor co-activator (SRC) family. Nat Rev Cancer 2009; 9:615-30. [PMID: 19701241 PMCID: PMC2908510 DOI: 10.1038/nrc2695] [Citation(s) in RCA: 389] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The three homologous members of the p160 SRC family (SRC1, SRC2 and SRC3) mediate the transcriptional functions of nuclear receptors and other transcription factors, and are the most studied of all the transcriptional co-activators. Recent work has indicated that the SRCgenes are subject to amplification and overexpression in various human cancers. Some of the molecular mechanisms responsible for SRC overexpression, along with the mechanisms by which SRCs promote breast and prostate cancer cell proliferation and survival, have been identified, as have the specific contributions of individual SRC family members to spontaneous breast and prostate carcinogenesis in genetically manipulated mouse models. These studies have identified new challenges for cancer research and therapy.
Collapse
Affiliation(s)
- Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Luzhou Medical College, Luzhou, Sichuan 646000, China
| | - Ray-Chang Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| |
Collapse
|
49
|
Jisa E, Graumann K, Jungbauer A. Proteins Accompanying the Estrogen Receptor α and β: A Model for Studying Protein Hetero-Complexes. BIOCATAL BIOTRANSFOR 2009. [DOI: 10.3109/10242420108992028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Williams JA, Kondo N, Okabe T, Takeshita N, Pilchak DM, Koyama E, Ochiai T, Jensen D, Chu ML, Kane MA, Napoli JL, Enomoto-Iwamoto M, Ghyselinck N, Chambon P, Pacifici M, Iwamoto M. Retinoic acid receptors are required for skeletal growth, matrix homeostasis and growth plate function in postnatal mouse. Dev Biol 2009; 328:315-27. [PMID: 19389355 DOI: 10.1016/j.ydbio.2009.01.031] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Revised: 01/20/2009] [Accepted: 01/23/2009] [Indexed: 12/21/2022]
Abstract
The retinoic acid receptors alpha, beta and gamma (RARalpha, RARbeta and RARgamma) are nuclear hormone receptors that regulate fundamental processes during embryogenesis, but their roles in skeletal development and growth remain unclear. To study skeletal-specific RAR function, we created conditional mouse mutants deficient in RAR expression in cartilage. We find that mice deficient in RARalpha and RARgamma (or RARbeta and RARgamma) exhibit severe growth retardation obvious by about 3 weeks postnatally. Their growth plates are defective and, importantly, display a major drop in aggrecan expression and content. Mice deficient in RARalpha and RARbeta, however, are virtually normal, suggesting that RARgamma is essential. In good correlation, we find that RARgamma is the most strongly expressed RAR in mouse growth plate and its expression characterizes the proliferative and pre-hypertrophic zones where aggrecan is strongly expressed also. By being avascular, those zones lack endogenous retinoids as indicated by previous RARE reporter mice and our direct biochemical measurements and thus, RARgamma is likely to exert ligand-less repressor function. Indeed, our data indicate that: aggrecan production is enhanced by RARgamma over-expression in chondrocytes under retinoid-free culture conditions; production is further boosted by co-repressor Zac1 or pharmacologic agents that enhance RAR repressor function; and RAR/Zac1 function on aggrecan expression may involve Sox proteins. In sum, our data reveal that RARs, and RARgamma in particular, exert previously unappreciated roles in growth plate function and skeletal growth and regulate aggrecan expression and content. Since aggrecan is critical for growth plate function, its deficiency in RAR-mutant mice is likely to have contributed directly to their growth retardation.
Collapse
Affiliation(s)
- Julie A Williams
- Department of Orthopaedic Surgery, Thomas Jefferson University College of Medicine, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|