1
|
Chen B, Zhao Y, Xu S, Jiang F, Nie Y, Tang A, Zhou Q. USF2 promotes autophagy and proliferation in chronic lymphocytic leukemia by inhibiting STUB1-induced NFAT5 ubiquitination. Ann Hematol 2024; 103:533-544. [PMID: 37950051 DOI: 10.1007/s00277-023-05522-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023]
Abstract
Chronic lymphocytic leukemia (CLL) mainly affects the health of older adults and is difficult to cure. Upstream stimulatory factor 2 (USF2) has been implicated in several diseases and conditions including cancers. However, the effect of USF2 on CLL has not been elucidated. To investigate the effect of USP2 on proliferation and autophagy of CLL, and to explore the underlying mechanism. The mRNA of USF2 and STIP1 homology and U-Box containing protein 1 (STUB1) was analyzed using qRT-PCR. Western blots were used to evaluate the expression level of USF2, LC3II, Beclin-1, P62, STUB1, and NFAT5. The cell proliferation was evaluated using CCK-8 and EdU assays. The cell apoptosis was evaluated using flow cytometry. Indirect fluorescent assay (IFA) was performed to analyze LC3 signal. Nuclear factor of activated T-cells 5 (NFAT5) ubiquitination was detected using immunoprecipitation (IP) assay. The CLL progression was evaluated in xenotransplantation model of nude mice. USF2 was highly expressed in CLL tissues and cell lines. USF2 knockdown suppressed the cell viability and EdU incorporation, while promoting cell apoptosis. Meanwhile, USF2 knockdown reduced the level of LC3II and Beclin-1, but increased P62, illustrating USF2 knockdown inhibiting autophagy. USF2 induced NFAT5 ubiquitination and promoted NFAT5 protein level via repressing STUB1. The downregulation of USF2 weakened CLL progression in xenotransplantation model of nude mice. CLL survival and autophagy was dependent on highly expressed USF2 which promoted the expression and ubiquitination of NFAT5 through inhibiting the transcription of STUB1, which makes USF2 a promising therapeutic candidate for CLL treatment.
Collapse
Affiliation(s)
- Beili Chen
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China.
| | - Yanyi Zhao
- Department of Oncology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Shujuan Xu
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Fang Jiang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Yuwei Nie
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Ailin Tang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| | - Qin Zhou
- Department of Hematology, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, China
| |
Collapse
|
2
|
Brooks EG, Elorriaga E, Liu Y, Duduit JR, Yuan G, Tsai CJ, Tuskan GA, Ranney TG, Yang X, Liu W. Plant Promoters and Terminators for High-Precision Bioengineering. BIODESIGN RESEARCH 2023; 5:0013. [PMID: 37849460 PMCID: PMC10328392 DOI: 10.34133/bdr.0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/12/2023] [Indexed: 10/19/2023] Open
Abstract
High-precision bioengineering and synthetic biology require fine-tuning gene expression at both transcriptional and posttranscriptional levels. Gene transcription is tightly regulated by promoters and terminators. Promoters determine the timing, tissues and cells, and levels of the expression of genes. Terminators mediate transcription termination of genes and affect mRNA levels posttranscriptionally, e.g., the 3'-end processing, stability, translation efficiency, and nuclear to cytoplasmic export of mRNAs. The promoter and terminator combination affects gene expression. In the present article, we review the function and features of plant core promoters, proximal and distal promoters, and terminators, and their effects on and benchmarking strategies for regulating gene expression.
Collapse
Affiliation(s)
- Emily G. Brooks
- Department of Horticultural Science, North Carolina State University, Raleigh, NC 27607, USA
| | - Estefania Elorriaga
- Department of Horticultural Science, North Carolina State University, Raleigh, NC 27607, USA
| | - Yang Liu
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
- The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - James R. Duduit
- Department of Horticultural Science, North Carolina State University, Raleigh, NC 27607, USA
| | - Guoliang Yuan
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
- The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Chung-Jui Tsai
- The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
- Warnell School of Forestry and Natural Resource, University of Georgia, Athens, GA 30602, USA
- Department of Plant Biology, University of Georgia, Athens, GA 30602, USA
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Gerald A. Tuskan
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
- The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Thomas G. Ranney
- Mountain Crop Improvement Lab, Department of Horticultural Science, Mountain Horticultural Crops Research and Extension Center, North Carolina State University, Mills River, NC 28759, USA
| | - Xiaohan Yang
- The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
- Warnell School of Forestry and Natural Resource, University of Georgia, Athens, GA 30602, USA
| | - Wusheng Liu
- Department of Horticultural Science, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
3
|
Zeng HB, Dong LQ, Huang YL, Xu C, Zhao XH, Wu LG. USF2 reduces BMP3 expression via transcriptional activation of miR-34a, thus promoting osteogenic differentiation of BMSCs. J Bone Miner Metab 2021; 39:997-1008. [PMID: 34350522 DOI: 10.1007/s00774-021-01254-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/18/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Osteoporosis is the most susceptible disease for people over 60. The main cause of osteoporosis is the decreased osteogenic differentiation of mesenchymal stem cells (MSCs). Here we showed that upstream stimulatory factor 2 (USF2)/microRNA-34a (miR-34a)/bone morphogenetic protein 3 (BMP3) axis regulated osteogenic differentiation of BMSCs. MATERIALS AND METHODS USF2 and miR-34a expression were examined using qPCR. Protein levels of BMP3 and osteogenic markers expression were evaluated using both western blot and qPCR. Activity of ALP was determined by ALP assay kit. Mineralization capacity of hBMSCs was assessed using ARS. Besides, CHIP assay was employed to verify whether USF2 could bind to miR-34a promoter. Finally, RIP assay and dual-luciferase reporter assay were employed to verify whether miR-34a directly bound to BMP3. RESULTS Our results suggested that miR-34a was upregulated during osteogenic differentiation of BMSCs, and miR-34a overexpression could enhance osteogenic differentiation of BMSCs. USF2 could positively regulate miR-34a expression by interacting with its promoter. USF2 overexpression enhanced osteogenic differentiation of BMSCs, while miR-34a inhibition reversed the effect. Besides, BMP3 was the target of miR-34a. MiR-34a overexpression enhanced osteogenic differentiation of BMSCs, which was abolished by BMP3 overexpression. CONCLUSION Taken together, USF2 enhanced osteogenic differentiation of BMSCs via downregulating BMP3 by interacting with miR-34a promoter.
Collapse
Affiliation(s)
- Han-Bing Zeng
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Li-Qiang Dong
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Yu-Liang Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Chao Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Xu-Hui Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China
| | - Lian-Guo Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No.318, Chaowang Road, Gongshu District, Hangzhou, 310005, Zhejiang Province, China.
| |
Collapse
|
4
|
Nobile V, Pucci C, Chiurazzi P, Neri G, Tabolacci E. DNA Methylation, Mechanisms of FMR1 Inactivation and Therapeutic Perspectives for Fragile X Syndrome. Biomolecules 2021; 11:biom11020296. [PMID: 33669384 PMCID: PMC7920310 DOI: 10.3390/biom11020296] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/13/2022] Open
Abstract
Among the inherited causes of intellectual disability and autism, Fragile X syndrome (FXS) is the most frequent form, for which there is currently no cure. In most FXS patients, the FMR1 gene is epigenetically inactivated following the expansion over 200 triplets of a CGG repeat (FM: full mutation). FMR1 encodes the Fragile X Mental Retardation Protein (FMRP), which binds several mRNAs, mainly in the brain. When the FM becomes methylated at 10-12 weeks of gestation, the FMR1 gene is transcriptionally silent. The molecular mechanisms involved in the epigenetic silencing are not fully elucidated. Among FXS families, there is a rare occurrence of males carrying a FM, which remains active because it is not methylated, thus ensuring enough FMRPs to allow for an intellectual development within normal range. Which mechanisms are responsible for sparing these individuals from being affected by FXS? In order to answer this critical question, which may have possible implications for FXS therapy, several potential epigenetic mechanisms have been described. Here, we focus on current knowledge about the role of DNA methylation and other epigenetic modifications in FMR1 gene silencing.
Collapse
Affiliation(s)
- Veronica Nobile
- Sezione di Medicina Genomica, Dipartimento Scienze della Vita e Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.N.); (C.P.); (P.C.); (G.N.)
| | - Cecilia Pucci
- Sezione di Medicina Genomica, Dipartimento Scienze della Vita e Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.N.); (C.P.); (P.C.); (G.N.)
| | - Pietro Chiurazzi
- Sezione di Medicina Genomica, Dipartimento Scienze della Vita e Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.N.); (C.P.); (P.C.); (G.N.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Genetica Medica, 00168 Rome, Italy
| | - Giovanni Neri
- Sezione di Medicina Genomica, Dipartimento Scienze della Vita e Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.N.); (C.P.); (P.C.); (G.N.)
- Greenwood Genetic Center, JC Self Research Institute, Greenwood, SC 29646, USA
| | - Elisabetta Tabolacci
- Sezione di Medicina Genomica, Dipartimento Scienze della Vita e Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.N.); (C.P.); (P.C.); (G.N.)
- Correspondence: ; Tel.: +39-06-30154606
| |
Collapse
|
5
|
Liu F, Wang X, Zheng B, Li D, Chen C, Lee IS, Zhong J, Li D, Liu Y. USF2 enhances the osteogenic differentiation of PDLCs by promoting ATF4 transcriptional activities. J Periodontal Res 2019; 55:68-76. [PMID: 31448831 DOI: 10.1111/jre.12689] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/22/2019] [Accepted: 07/11/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Our study aimed to elucidate the regulatory molecules related to the osteogenic differentiation of periodontal ligament cells (PDLCs). BACKGROUND Periodontal ligament cells are a favorable source for cell-based therapy in periodontal bone engineering and regeneration due to their potential multilineage differentiation ability. However, the molecular mechanism and signaling pathways related to the osteogenic differentiation of PDLCs are still unclear. METHODS Osteoblast-specific protein expression levels were examined by ELISA in osteogenic-induced PDLCs (induced-PDLC group). A microarray assay and a bioinformatics analysis were carried out to reveal significantly expressed genes and the related pathways in induced-PDLCs, and these findings were then confirmed by qRT-PCR and a luciferase reporter assay. Finally, overexpressing and silencing gene systems were established to identify the specific transcriptional relationship and function of the target genes on the osteogenic differentiation of PDLCs. RESULTS Osteogenically differentiated PDLCs with high levels of osteoblast-specific proteins were established. The upstream stimulatory factor 2 (USF2) and activating transcription factor 4 (ATF4) mRNA levels were upregulated the most through the MAPK signaling pathway in the induced-PDLC group. USF2 could bind to the transcriptional initiation region of ATF4 and regulate its transcriptional activities. Additionally, the overexpression of USF2 promoted osteoblast-specific gene expression and the Alizarin red staining of PDLCs, while simultaneously overexpressing USF2 and silencing ATF4 reversed the favorable osteogenic effect of the induced-PDLCs by reducing osteoblast-specific gene expression and the Alizarin red staining level. CONCLUSION Our study demonstrated that USF2 could enhance the osteogenic differentiation of PDLCs by regulating ATF4 transcriptional activities, which provides a new strategy to utilize USF2 and ATF4 as potential target molecules for periodontal bone regeneration.
Collapse
Affiliation(s)
- Fan Liu
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China.,Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Xiaohong Wang
- Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China
| | - Bowen Zheng
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Danni Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Cen Chen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - In-Seop Lee
- Institute of Natural Science, Yonsei University, Seoul, Korea
| | - Jialin Zhong
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Duo Li
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Yi Liu
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
6
|
Bhawe K, Roy D. Interplay between NRF1, E2F4 and MYC transcription factors regulating common target genes contributes to cancer development and progression. Cell Oncol (Dordr) 2018; 41:465-484. [DOI: 10.1007/s13402-018-0395-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2018] [Indexed: 12/12/2022] Open
|
7
|
Abstract
Fragile X syndrome (FXS) is one of the major causes for autism and mental retardation in humans. The etiology of FXS is linked to the expansion of the CGG trinucleotide repeats, r(CGG), suppressing the fragile X mental retardation 1 (FMR1) gene on the X chromosome, resulting in a loss of fragile X mental retardation protein (FMRP) expression, which is required for regulating normal neuronal connectivity and plasticity. Recent studies have further identified that microRNAs are involved in the mechanisms underlying FXS pathogenesis at three different developmental stages. During early embryogenesis before the blastocyst stage, an embryonic stem cell (ESC)-specific microRNA, miR-302, interferes with FMR1 mRNA translation to maintain the stem cell status and inhibit neural development. After blastocyst, the downregulation of miR-302 releases FMRP synthesis and subsequently leads to neuronal development; yet, in FXS, certain r(CGG)-derived microRNAs, such as miR-fmr1s, are expressed and accumulated and then induce DNA hypermethylation on the FMR1 gene promoter regions, resulting in transcriptional inactivation of the FMR1 gene and the loss of FMRP. In normal neuronal development, FMRP is an RNA-binding protein responsible for interacting with miR-125 and miR-132 to regulate the signaling of Group 1 metabotropic glutamate receptor (mGluR1) and N-methyl-D-aspartate receptor (NMDAR), respectively, and consequently affecting synaptic plasticity. As a result, the loss of FMRP impairs these signaling controls and eventually causes FXS-associated disorders, such as autism and mental retardation. Based on these current findings, this chapter will summarize the etiological causes of FXS and further provides significant insights into the molecular mechanisms underlying microRNA-mediated FXS pathogenesis and the related therapy development.
Collapse
|
8
|
Testosterone down regulates the expression of Fmr-1 gene in the cerebral cortex of gonadectomized old male mice. Biogerontology 2014; 15:503-15. [DOI: 10.1007/s10522-014-9521-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/17/2014] [Indexed: 10/25/2022]
|
9
|
Alterations in the Sp1 binding and Fmr-1 gene expression in the cortex of the brain during maturation and aging of mouse. Mol Biol Rep 2014; 41:6855-63. [DOI: 10.1007/s11033-014-3571-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 06/26/2014] [Indexed: 11/25/2022]
|
10
|
Rousseau F, Labelle Y, Bussières J, Lindsay C. The fragile x mental retardation syndrome 20 years after the FMR1 gene discovery: an expanding universe of knowledge. Clin Biochem Rev 2011; 32:135-162. [PMID: 21912443 PMCID: PMC3157949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The fragile X mental retardation (FXMR) syndrome is one of the most frequent causes of mental retardation. Affected individuals display a wide range of additional characteristic features including behavioural and physical phenotypes, and the extent to which individuals are affected is highly variable. For these reasons, elucidation of the pathophysiology of this disease has been an important challenge to the scientific community. 1991 marks the year of the discovery of both the FMR1 gene mutations involved in this disease, and of their dynamic nature. Although a mouse model for the disease has been available for 16 years and extensive research has been performed on the FMR1 protein (FMRP), we still understand little about how the disease develops, and no treatment has yet been shown to be effective. In this review, we summarise current knowledge on FXMR with an emphasis on the technical challenges of molecular diagnostics, on its prevalence and dynamics among populations, and on the potential of screening for FMR1 mutations.
Collapse
Affiliation(s)
- François Rousseau
- Réseau de Médecine Génétique Appliquée, Fonds de Recherche en Santé du Québec
- The APOGEE-Net/CanGèneTest Research and Knowledge Network (www.cangenetest.org)
- Unité de recherche en génétique humaine et moléculaire, Axe de recherche en évaluation des technologies et transfert des connaissances, Centre de recherche du CHUQ-Hôpital-Saint-François-d’Assise
- Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, CRCHUQ-Hôpital St-François d’Assise, 10 rue de l’Espinay, Québec, Qc, Canada G1L 3L5
| | - Yves Labelle
- The APOGEE-Net/CanGèneTest Research and Knowledge Network (www.cangenetest.org)
- Unité de recherche en génétique humaine et moléculaire, Axe de recherche en évaluation des technologies et transfert des connaissances, Centre de recherche du CHUQ-Hôpital-Saint-François-d’Assise
- Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, CRCHUQ-Hôpital St-François d’Assise, 10 rue de l’Espinay, Québec, Qc, Canada G1L 3L5
| | - Johanne Bussières
- Unité de recherche en génétique humaine et moléculaire, Axe de recherche en évaluation des technologies et transfert des connaissances, Centre de recherche du CHUQ-Hôpital-Saint-François-d’Assise
| | - Carmen Lindsay
- Unité de recherche en génétique humaine et moléculaire, Axe de recherche en évaluation des technologies et transfert des connaissances, Centre de recherche du CHUQ-Hôpital-Saint-François-d’Assise
| |
Collapse
|
11
|
Okoh V, Deoraj A, Roy D. Estrogen-induced reactive oxygen species-mediated signalings contribute to breast cancer. Biochim Biophys Acta Rev Cancer 2010; 1815:115-33. [PMID: 21036202 DOI: 10.1016/j.bbcan.2010.10.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 10/12/2010] [Accepted: 10/14/2010] [Indexed: 01/01/2023]
Abstract
Elevated lifetime estrogen exposure is a major risk factor for breast cancer. Recent advances in the understanding of breast carcinogenesis clearly indicate that induction of estrogen receptor (ER) mediated signaling is not sufficient for the development of breast cancer. The underlying mechanisms of breast susceptibility to estrogen's carcinogenic effect remain elusive. Physiologically achievable concentrations of estrogen or estrogen metabolites have been shown to generate reactive oxygen species (ROS). Recent data implicated that these ROS induced DNA synthesis, increased phosphorylation of kinases, and activated transcription factors, e.g., AP-1, NRF1, E2F, NF-kB and CREB of non-genomic pathways which are responsive to both oxidants and estrogen. Estrogen-induced ROS by increasing genomic instability and by transducing signal through influencing redox sensitive transcription factors play important role (s) in cell transformation, cell cycle, migration and invasion of the breast cancer. The present review discusses emerging data in support of the role of estrogen induced ROS-mediated signaling pathways which may contribute in the development of breast cancer. It is envisioned that estrogen induced ROS mediated signaling is a key complementary mechanism that drives the carcinogenesis process. ROS mediated signaling however occurs in the context of other estrogen induced processes such as ER-mediated signaling and estrogen reactive metabolite-associated genotoxicity. Importantly, estrogen-induced ROS can function as independent reversible modifiers of phosphatases and activate kinases to trigger the transcription factors of downstream target genes which participate in cancer progression.
Collapse
Affiliation(s)
- Victor Okoh
- Department of Environmental and Occupational Health, Florida International University, Miami, FL, USA
| | | | | |
Collapse
|
12
|
Collins SC, Bray SM, Suhl JA, Cutler DJ, Coffee B, Zwick ME, Warren ST. Identification of novel FMR1 variants by massively parallel sequencing in developmentally delayed males. Am J Med Genet A 2010; 152A:2512-20. [PMID: 20799337 PMCID: PMC2946449 DOI: 10.1002/ajmg.a.33626] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Fragile X syndrome (FXS), the most common inherited form of developmental delay, is typically caused by CGG-repeat expansion in FMR1. However, little attention has been paid to sequence variants in FMR1. Through the use of pooled-template massively parallel sequencing, we identified 130 novel FMR1 sequence variants in a population of 963 developmentally delayed males without CGG-repeat expansion mutations. Among these, we identified a novel missense change, p.R138Q, which alters a conserved residue in the nuclear localization signal of FMRP. We have also identified three promoter mutations in this population, all of which significantly reduce in vitro levels of FMR1 transcription. Additionally, we identified 10 noncoding variants of possible functional significance in the introns and 3'-untranslated region of FMR1, including two predicted splice site mutations. These findings greatly expand the catalog of known FMR1 sequence variants and suggest that FMR1 sequence variants may represent an important cause of developmental delay.
Collapse
Affiliation(s)
- Stephen C. Collins
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven M. Bray
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joshua A. Suhl
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - David J. Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bradford Coffee
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael E. Zwick
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stephen T. Warren
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Departments of Biochemistry and Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Kumari D, Usdin K. The distribution of repressive histone modifications on silenced FMR1 alleles provides clues to the mechanism of gene silencing in fragile X syndrome. Hum Mol Genet 2010; 19:4634-42. [PMID: 20843831 DOI: 10.1093/hmg/ddq394] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common heritable cause of intellectual disability and the most common known cause of autism. Most cases of FXS result from the expansion of a CGG·CCG repeat in the 5' UTR of the FMR1 gene that leads to gene silencing. It has previously been shown that silenced alleles are associated with histone H3 dimethylated at lysine 9 (H3K9Me2) and H3 trimethylated at lysine 27 (H3K27Me3), modified histones typical of developmentally repressed genes. We show here that these alleles are also associated with elevated levels of histone H3 trimethylated at lysine 9 (H3K9Me3) and histone H4 trimethylated at lysine 20 (H4K20Me3). All four of these modified histones are present on exon 1 of silenced alleles at levels comparable to that seen on pericentric heterochromatin. The two groups of histone modifications show a different distribution on fragile X alleles: H3K9Me2 and H3K27Me3 have a broad distribution, whereas H3K9Me3 and H4K20Me3 have a more focal distribution with the highest level of these marks being present in the vicinity of the repeat. This suggests that the trigger for gene silencing may be local to the repeat itself and perhaps involves a mechanism similar to that involved in the formation of pericentric heterochromatin.
Collapse
Affiliation(s)
- Daman Kumari
- Section on Gene Structure and Disease, Laboratory of Molecular and Cellular Biology, National Institute of Diabetes and Digestive and Kidney Disease/NIH, Bethesda, MD 20892-0830, USA.
| | | |
Collapse
|
14
|
Higgins PJ. The TGF-beta1/upstream stimulatory factor-regulated PAI-1 gene: potential involvement and a therapeutic target in Alzheimer's disease. J Biomed Biotechnol 2010; 2006:15792. [PMID: 17047299 PMCID: PMC1526650 DOI: 10.1155/jbb/2006/15792] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Amyloid peptide (Aβ) aggregates, derived from initial β-site proteolytic processing of the amyloid precursor
protein (APP), accumulate in the brains of Alzheimer's disease
patients. The plasmin-generating cascade appears to serve a
protective role in the central nervous system since
plasmin-mediated proteolysis of APP utilizes the α site, eventually generating nontoxic peptides, and plasmin also degrades
Aβ. The conversion of plasminogen to plasmin by tissue-type
plasminogen activator in the brain is negatively regulated by
plasminogen activator inhibitor type-1 (PAI-1) resulting in
attenuation of plasmin-dependent substrate degradation with
resultant accumulation of Aβ. PAI-1 and its major
physiological inducer TGF-β1, moreover, are increased in
models of Alzheimer's disease and have been implicated in the
etiology and progression of human neurodegenerative disorders.
This review highlights the potential role of PAI-1 and TGF-β1 in this process. Current molecular events associated with
TGF-β1-induced PAI-1 transcription are presented with
particular relevance to potential targeting of PAI-1 gene
expression as a molecular approach to the therapy of
neurodegenerative diseases associated with increased PAI-1
expression such as Alzheimer's disease.
Collapse
Affiliation(s)
- Paul J. Higgins
- Center for Cell Biology and Cancer Research, Albany Medical College,
Albany, NY 12208, USA
- *Paul J. Higgins:
| |
Collapse
|
15
|
Prasad S, Singh K. Interaction of USF1/USF2 and alpha-Pal/Nrf1 to Fmr-1 promoter increases in mouse brain during aging. Biochem Biophys Res Commun 2008; 376:347-51. [PMID: 18782566 DOI: 10.1016/j.bbrc.2008.08.155] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Accepted: 08/27/2008] [Indexed: 11/26/2022]
Abstract
Fragile X syndrome is caused due to silencing of FMR-1 gene transcription leading to loss of fragile X mental retardation protein (FMRP). To investigate whether the transcriptional mechanism is linked to aging, we have studied interaction of the transcription factors USF1/USF2 and alpha-Pal/Nrf1 to E-box and GC-box, respectively, in Fmr-1 promoter in the brain of young, adult, and old mouse using electrophoretic mobility shift assay (EMSA). Our data reveal that the interaction of these transcription factors to their respective promoter sequences increases in mouse brain as a function of age. The finding on the interaction of the above transcription factors to their cognate sequences is novel as the current investigation has been carried out in intact and aging mouse. The present finding is important in respect to age- and FMRP-dependent brain function.
Collapse
Affiliation(s)
- S Prasad
- Biochemistry & Molecular Biology Lab, CAS in Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| | | |
Collapse
|
16
|
Prasad S, Singh K. Age- and sex-dependent differential interaction of nuclear trans-acting factors with Fmr-1 promoter in mice brain. Neurochem Res 2007; 33:1028-35. [PMID: 18080753 DOI: 10.1007/s11064-007-9545-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 11/07/2007] [Indexed: 11/25/2022]
Abstract
We have investigated relation between interaction of the trans-acting factors with Fmr-1 promoter and expression of FMRP isoforms in intact mouse brain as a function of age and sex. Our EMSA data reveal that among the three complexes formed with 136 bp Fmr-1 promoter fragment, the level of complex C1 significantly increases in adult brain but decreases in old brain in comparison to that in young. The level of total FMRP significantly decreases from young to old in the brain of both the sexes, however, among the three isoforms, expression of the 80-kDa isoform significantly decreases in the brain of both the sexes where as the level of 70 kDa isoform decreases in females during aging. The present finding on relation between age- and sex-dependent interaction of trans-acting factors and expression of FMRP isoforms is novel and may be relevant for regulation of Fmr-1 gene in brain function during aging.
Collapse
Affiliation(s)
- S Prasad
- Biochemistry & Molecular Biology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, UP, India.
| | | |
Collapse
|
17
|
Singh K, Prasad S. Differential expression of Fmr-1 mRNA and FMRP in female mice brain during aging. Mol Biol Rep 2007; 35:677-84. [PMID: 17899445 DOI: 10.1007/s11033-007-9140-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2007] [Accepted: 08/29/2007] [Indexed: 10/22/2022]
Abstract
Fragile X syndrome is caused by silencing of FMR-1 gene due to unusual expansion of CGG repeats (>200 repeats) and their hypermethylation in 5'-UTR. As a consequence, the expression of the RNA binding protein FMRP is stopped. Absence of this protein leads to several morphological and neurological symptoms. The symptoms of the syndrome in males are different than that in the females. We have previously reported that the Fmr1 gene is down regulated in males as a function of age. In the present communication, we have investigated expression of Fmr-1 mRNA, FMRP and analysis of interaction of trans-acting factors with E- and GC boxes in Fmr-1 promoter in female mouse brain as a function of age. Our Northern and Western blots data reveal that the level of Fmr-1 transcript decreases in adult as compared to young mouse but significantly increases in old age and that of FMRP decreases in brain of female old mouse as compared to young and adult age. The immunohistochemical analysis supported the results obtained from Western blot studies. Our EMSA data reveal that the intensity of USF1/USF2-E Box complex gradually increases during aging having significantly highest intensity in old age mouse whereas the intensity of alpha-Pal/Nrf1- GC-Box complex gradually decreases as a function of age. The increased intensity of the complex in old age mouse is correlated to higher level of Fmr-1 transcript in old age. The elevated level of Fmr-1 transcript in old mouse brain may be attributed to USF1/USF2 dependent increased transcription of Fmr-1 gene in old age and decrease in FMRP to altered translation of the transcript or high turn over of FMRP during aging. The present finding indicates age and sex as factors affecting the expression of Fmr-1 gene in mouse brain.
Collapse
Affiliation(s)
- Kanchan Singh
- Biochemistry & Molecular Biology laboratory, CAS in Zoology, Banaras Hindu University, Varanasi, 221005, India
| | | |
Collapse
|
18
|
Taulan M, Lopez E, Guittard C, René C, Baux D, Altieri JP, DesGeorges M, Claustres M, Romey MC. First functional polymorphism in CFTR promoter that results in decreased transcriptional activity and Sp1/USF binding. Biochem Biophys Res Commun 2007; 361:775-81. [PMID: 17678620 DOI: 10.1016/j.bbrc.2007.07.091] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2007] [Accepted: 07/18/2007] [Indexed: 11/23/2022]
Abstract
Growing evidences show that functionally relevant polymorphisms in various promoters alter both transcriptional activity and affinities of existing protein-DNA interactions, and thus influence disease progression in humans. We previously reported the -94G>T CFTR promoter variant in a female CF patient in whom any known disease-causing mutation has been detected. To investigate whether the -94G>T could be a regulatory variant, we have proceeded to in silico analyses and functional studies including EMSA and reporter gene assays. Our data indicate that the promoter variant decreases basal CFTR transcriptional activity in different epithelial cells and alters binding affinities of both Sp1 and USF nuclear proteins to the CFTR promoter. The present report provides evidence for the first functional polymorphism that negatively affects the CFTR transcriptional activity and demonstrates a cooperative role of Sp1 and USF transcription factors in transactivation of the CFTR gene promoter.
Collapse
Affiliation(s)
- M Taulan
- INSERM U827, Laboratoire de Génétique de Maladies Rares, Montpellier, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Fragile X syndrome is the most common form of inherited mental retardation. The disorder is mainly caused by the expansion of the trinucleotide sequence CGG located in the 5' UTR of the FMR1 gene on the X chromosome. The abnormal expansion of this triplet leads to hypermethylation and consequent silencing of the FMR1 gene. Thus, the absence of the encoded protein (FMRP) is the basis for the phenotype. FMRP is a selective RNA-binding protein that associates with polyribosomes and acts as a negative regulator of translation. FMRP appears to play an important role in synaptic plasticity by regulating the synthesis of proteins encoded by certain mRNAs localized in the dendrite. An advancing understanding of the pathophysiology of this disorder has led to promising strategies for pharmacologic interventions.
Collapse
Affiliation(s)
- Olga Penagarikano
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | |
Collapse
|
20
|
Abstract
Fragile X syndrome, the most common heritable form of mental retardation, is caused by silencing of the FMR1 (fragile X mental retardation-1 gene). The protein product of this gene, FMRP (fragile X mental retardation protein), is thought to be involved in the translational regulation of mRNAs important for learning and memory. In mammals, there are two homologues of FMRP, namely FXR1P (fragile X-related protein 1) and FXR2P. Disruption of Fxr2 in mice produces learning and memory deficits, and Fmr1 and Fxr2 double-knockout mice have exaggerated impairments in certain neurobehavioral phenotypes relative to the single gene knockouts. This has led to the suggestion that FMR1 and FXR2 functionally overlap and that increasing the expression of FXR2P may ameliorate the symptoms of an FMRP deficiency. Interestingly, the region upstream of the FXR2 translation start site acts as a bidirectional promoter in rodents, driving transcription of an alternative transcript encoding the ABP (androgen-binding protein) [aABP (alternative ABP promoter)]. To understand the regulation of the human FXR2 gene, we cloned the evolutionarily conserved region upstream of the FXR2 translation start site and showed that it also has bidirectional promoter activity in both neuronal and muscle cells as evidenced by luciferase reporter assay studies. Alignment of the human, mouse, rat, rabbit and dog promoters reveals several highly conserved transcription factor-binding sites. Gel electrophoretic mobility-shift assays, chromatin immunoprecipitation studies and co-transfection experiments with plasmids expressing these transcription factors or dominant-negative versions of these factors showed that NF-YA (nuclear transcription factor Yalpha), AP2 (activator protein 2), Nrf1 (nuclear respiratory factor/alpha-Pal) and Sp1 (specificity protein 1) all bind to the FXR2 promoter both in vitro and in vivo and positively regulate the FXR2 promoter.
Collapse
Affiliation(s)
- Lata Mahishi
- Gene Structure and Disease Section, NIDDK (National Institute of Diabetes and Digestive and Kidney Diseases), NIH (National Institutes of Health), Bethesda, MD 20892-0830, U.S.A
| | - Karen Usdin
- Gene Structure and Disease Section, NIDDK (National Institute of Diabetes and Digestive and Kidney Diseases), NIH (National Institutes of Health), Bethesda, MD 20892-0830, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
21
|
Garber K, Smith KT, Reines D, Warren ST. Transcription, translation and fragile X syndrome. Curr Opin Genet Dev 2006; 16:270-5. [PMID: 16647847 DOI: 10.1016/j.gde.2006.04.010] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Accepted: 04/18/2006] [Indexed: 10/24/2022]
Abstract
The fragile X mental retardation protein (FMRP) plays a role in the control of local protein synthesis in the dendrites. Loss of its production in fragile X syndrome is associated with transcriptional dysregulation of the gene. Recent work demonstrates that Sp1 and NRF1 transcriptionally control this gene. Other studies reveal how the microRNA pathway and signaling are related to FMRP function through the metabotropic glutamate receptor. These studies provide new insights through which we can better understand the inactivation of the FMR1 gene and, in turn, the consequence of FMRP loss.
Collapse
Affiliation(s)
- Kathryn Garber
- Department of Human Genetics, 615 Michael Street, Room 300, Emory University, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
22
|
Smith KT, Nicholls RD, Reines D. The gene encoding the fragile X RNA-binding protein is controlled by nuclear respiratory factor 2 and the CREB family of transcription factors. Nucleic Acids Res 2006; 34:1205-15. [PMID: 16500891 PMCID: PMC1383620 DOI: 10.1093/nar/gkj521] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
FMR1 encodes an RNA-binding protein whose absence results in fragile X mental retardation. In most patients, the FMR1 gene is cytosine-methylated and transcriptionally inactive. NRF-1 and Sp1 are known to bind and stimulate the active, but not the methylated/silenced, FMR1 promoter. Prior analysis has implicated a CRE site in regulation of FMR1 in neural cells but the role of this site is controversial. We now show that a phospho-CREB/ATF family member is bound to this site in vivo. We also find that the histone acetyltransferases CBP and p300 are associated with active FMR1 but are lost at the hypoacetylated fragile X allele. Surprisingly, FMR1 is not cAMP-inducible and resides in a newly recognized subclass of CREB-regulated genes. We have also elucidated a role for NRF-2 as a regulator of FMR1 in vivo through a previously unrecognized and highly conserved recognition site in FMR1. NRF-1 and NRF-2 act additively while NRF-2 synergizes with CREB/ATF at FMR1's promoter. These data add FMR1 to the collection of genes controlled by both NRF-1 and NRF-2 and disfavor its membership in the immediate early response group of genes.
Collapse
Affiliation(s)
| | - Robert D. Nicholls
- Birth Defects Laboratories and Division of Medical Genetics, Department of Pediatrics, Children's Hospital of Pittsburgh3705 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Daniel Reines
- To whom correspondence should be addressed. Tel: +1 404 727 3361; Fax: +1 404 727 3452;
| |
Collapse
|
23
|
Lim JH, Booker AB, Luo T, Williams T, Furuta Y, Lagutin O, Oliver G, Sargent TD, Fallon JR. AP-2alpha selectively regulates fragile X mental retardation-1 gene transcription during embryonic development. Hum Mol Genet 2005; 14:2027-34. [PMID: 15930016 DOI: 10.1093/hmg/ddi207] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Fragile X syndrome (FXS) is almost always caused by silencing of the FMR1 gene. The defects observed in FXS indicate that the normal FMR1 gene has a range of functions and plays a particularly prominent role during development. However, the mechanisms regulating FMR1 expression in vivo are not known. Here, we have tested the role of the transcription factor AP-2alpha in regulating Fmr1 expression. Chromatin immunoprecipitation showed that AP-2alpha associates with the Fmr1 promoter in vivo. Furthermore, Fmr1 transcript levels are reduced >4-fold in homozygous null AP-2alpha mutant mice at embryonic day 18.5 when compared with normal littermates. Notably, AP-2alpha exhibits a strong gene dosage effect, with heterozygous mice showing approximately 2-fold reduction in Fmr1 levels. Examination of conditional AP-2alpha mutant mice indicates that this transcription factor plays a major role in regulating Fmr1 expression in embryos, but not in adults. We further investigated the role of AP-2alpha in the developmental regulation of Fmr1 expression using the Xenopus animal cap assay. Over-expression of a dominant-negative AP-2alpha in Xenopus embryos led to reduced Fmr1 levels. Moreover, exogenous wild-type AP-2alpha rescued Fmr1 expression in embryos where endogenous AP-2alpha had been suppressed. We conclude that AP-2alpha associates with the Fmr1 promoter in vivo and selectively regulates Fmr1 transcription during embryonic development.
Collapse
Affiliation(s)
- Jae H Lim
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | | | | | | | | | | | | | | | | |
Collapse
|