1
|
Ayten H, Toker P, Turan Duman G, Olgun ÇE, Demiralay ÖD, Bınarcı B, Güpür G, Yaşar P, Akman HB, Haberkant P, Muyan M. CXXC5 is a ubiquitinated protein and is degraded by the ubiquitin-proteasome pathway. Protein Sci 2025; 34:e70140. [PMID: 40371716 PMCID: PMC12079423 DOI: 10.1002/pro.70140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025]
Abstract
CXXC5, as a member of the zinc-finger CXXC family proteins, interacts with unmodified CpG dinucleotides to modulate the expression of genes involved in cellular proliferation, differentiation, and death in physiology and pathophysiology. Various signaling pathways, including mitogenic 17β-estradiol (E2), contribute to the expression and synthesis of CXXC5. However, how signaling pathways modulate protein levels of CXXC5 in cells is largely unknown. We previously reported that some key regulators, including retinoblastoma 1 and E74-like ETS transcription factor 1, of the G1 to S phase transitions are involved in the expression of CXXC5 in estrogen-responsive MCF-7 cells, derived from a breast adenocarcinoma. We, therefore, predict that the synthesis of CXXC5 is regulated in a cell cycle-dependent manner. We report here that although E2 in synchronized MCF-7 cells augments both transcription and synthesis of CXXC5 in the G1 phase, CXXC5 protein levels are primarily mediated by ubiquitination independently of cell cycle phases. Utilizing the bioUbiquitination approach, which is based on cellular biotinylation of ubiquitin, in HEK293FT cells derived from immortalized human embryonic kidney cells, followed by sequential immunoprecipitation coupled mass spectrometry analyses, we identified ubiquitinated lysine residues of CXXC5. We show in both MCF-7 and HEK293FT cells that the ubiquitinated lysine residues contribute to the degradation of CXXC5 through the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Hazal Ayten
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| | - Pelin Toker
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| | - Gizem Turan Duman
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| | - Çağla Ece Olgun
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| | - Öykü Deniz Demiralay
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
- Present address:
Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
| | - Büşra Bınarcı
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| | - Gizem Güpür
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| | - Pelin Yaşar
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
- Present address:
Epigenetics and Stem Cell Biology Laboratory, Single Cell Dynamics GroupNational Institute of Environmental Health SciencesResearch Triangle ParkNorth CarolinaUSA
| | - Hesna Begüm Akman
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| | - Per Haberkant
- Proteomics Core FacilityEMBL HeidelbergHeidelbergGermany
| | - Mesut Muyan
- Department of Biological SciencesMiddle East Technical UniversityÇankaya‐AnkaraTürkiye
| |
Collapse
|
2
|
Toker P, Ayten H, Demiralay ÖD, Bınarcı B, Turan G, Olgun ÇE, Yaşar P, Akman HB, Muyan M. A reappraisal of cell cycle phase enrichment in synchronized estrogen receptor-positive cell models derived from breast adenocarcinomas. Sci Rep 2025; 15:5949. [PMID: 39966575 PMCID: PMC11836305 DOI: 10.1038/s41598-025-90456-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
17β-estradiol (E2), the main circulating estrogen hormone, is a critical signaling factor for the growth, differentiation, and function of breast epithelial cells. The effects of E2 on the breast tissue are primarily mediated by the estrogen receptor α (ERα). Deregulation of the E2-ERα signaling contributes to the initiation/progression of breast cancer and resistance to treatments. Cell lines from breast adenocarcinomas as in vitro model systems provide invaluable insight into cellular events, drug discovery, and drug resistance. Among ERα-synthesizing cell lines, MCF7 and T47D cells are widely used to elucidate cell cycle phase-specific molecular events that coordinate cellular proliferation mediated by E2-ERα. Due to variable results in generating phase-enriched populations with various approaches, we wanted to reassess cell cycle synchronization-coupled phase enrichment with charcoal dextran-treated fetal bovine serum, CD-FBS, as an effective hormone withdrawal approach, alone or in combination with excess thymidine, as a DNA replication inhibitor, and/or nocodazole, a microtubule poison, in MCF7 and T47D cells. We find that hormone withdrawal synchronizes both MCF7 and T47D cells at the G0/G1 phase. Supplementation of CD-FBS with E2 enriches the S phase population. E2 with nocodazole and nocodazole-coupled mitotic shake-off augments the G2/M phase population of MCF7 cells. However, the double thymidine block approach with nocodazole or nocodazole-coupled mitotic shake-off is more effective in enriching S and G2/M phase populations of T47D cells. Our results highlight the differential efficacy of synchronization approaches in MCF7 and T47D cells that could provide a framework for cell cycle-specific applications in breast cancer research.
Collapse
Affiliation(s)
- Pelin Toker
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
| | - Hazal Ayten
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
| | - Öykü Deniz Demiralay
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Büşra Bınarcı
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
| | - Gizem Turan
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
| | - Çağla Ece Olgun
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
| | - Pelin Yaşar
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
- Current Address: Epigenetics and Stem Cell Biology Laboratory, Single Cell Dynamics Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Hesna Begüm Akman
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye
| | - Mesut Muyan
- Department of Biological Sciences, Middle East Technical University, 06800, Çankaya-Ankara, Türkiye.
| |
Collapse
|
3
|
An Z, Wang J, Li C, Tang C. Signal integrator function of CXXC5 in Cancer. Cell Commun Signal 2025; 23:25. [PMID: 39806388 PMCID: PMC11730785 DOI: 10.1186/s12964-024-02005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
CXXC type zinc finger protein 5 (CXXC5) is a member of the ZF-CXXC family and plays a pivotal role in signal integration and information transfer within cell signaling network. CXXC5 acts as a regulator in various physiological processes, and abnormalities in its protein structure or function have been linked to multiple pathological processes. In this article, we correspondingly describe the composition of the ZF-CXXC family, emphatically introducing the features of the CXXC5 gene and protein, review the role of CXXC5 in cellular signaling networks, the physiological and pathological processes associated with CXXC5 dysregulation, and particularly focus on the correlation between CXXC5 and cancers. Finally, we summarize the current therapies targeting CXXC5 and their potential applications, and discuss the intriguing findings from current studies, and the opportunities and challenges in future.
Collapse
Affiliation(s)
- Zihao An
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Jiepu Wang
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chengzuo Li
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Chao Tang
- National Clinical Research Center for Child Health of Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
| |
Collapse
|
4
|
Viswanath M, Peter MCS. Thyroid hormones activate TH/E 2 receptor/regulator system and drive Na +/K +-ATPase in the ovarian wall of hypothyroid air-breathing fish (Anabas testudineus Bloch). Gen Comp Endocrinol 2025; 360:114640. [PMID: 39536982 DOI: 10.1016/j.ygcen.2024.114640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 11/09/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
In fish, as in other vertebrates, thyroid hormones (THs) act on many biological processes including growth and reproduction. Primary THs such as thyroxine (T4) and triiodothyronine (T3) are known for their direct action on osmoregulatory organs regulating ion osmotic homeostasis in many teleost fishes. However, it is unclear how these hormones interact with estradiol-17β (E2), an ovarian hormone that regulates the development of oocytes. We thus examined the short-term in vivo action of varied THs such as T4, T3 and T2, a potent TH metabolite diiodothyronine, on the expression pattern of receptors of THs and E2 in the ovarian wall of the hypothyroid climbing perch to identify the interactive pattern of TH/E2 receptor system and the molecular dynamics of Na+/K+-ATPase (NKA) subunits in the ovarian wall that provides structural and functional support to ovary. We found differential pattern of transcript abundance of NKA subunits isoforms such as nkaα1a, nkaα1b, nkaα1c atp1b1, atp1b2 and fxyd3, fxyd5, fxyd6, TH receptor isoforms (tr<, trβ, tr
Collapse
Affiliation(s)
- Meenu Viswanath
- Inter-University Centre for Evolutionary and Integrative Biology-iCEIB, School of Life Sciences, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Department of Zoology, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India
| | - M C Subhash Peter
- Inter-University Centre for Evolutionary and Integrative Biology-iCEIB, School of Life Sciences, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Department of Zoology, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Sastrajeevan Integrative Bioresearch and Education-SIBE, F17-Gandhipuram, Sreekariyam, Thiruvananthapuram 695017, Kerala, India.
| |
Collapse
|
5
|
Geng T, Sun Q, He J, Chen Y, Cheng W, Shen J, Liu B, Zhang M, Wang S, Asan K, Song M, Gao Q, Song Y, Liu R, Liu X, Ding Y, Jing A, Ye X, Ren H, Zeng K, Zhou Y, Zhang B, Ma S, Liu W, Liu S, Ji J. CXXC5 drove inflammation and ovarian cancer proliferation via transcriptional activation of ZNF143 and EGR1. Cell Signal 2024; 119:111180. [PMID: 38642782 DOI: 10.1016/j.cellsig.2024.111180] [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: 01/01/2024] [Revised: 03/28/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024]
Abstract
CXXC5, a zinc-finger protein, is known for its role in epigenetic regulation via binding to unmethylated CpG islands in gene promoters. As a transcription factor and epigenetic regulator, CXXC5 modulates various signaling processes and acts as a key coordinator. Altered expression or activity of CXXC5 has been linked to various pathological conditions, including tumorigenesis. Despite its known role in cancer, CXXC5's function and mechanism in ovarian cancer are unclear. We analyzed multiple public databases and found that CXXC5 is highly expressed in ovarian cancer, with high expression correlating with poor patient prognosis. We show that CXXC5 expression is regulated by oxygen concentration and is a direct target of HIF1A. CXXC5 is critical for maintaining the proliferative potential of ovarian cancer cells, with knockdown decreasing and overexpression increasing cell proliferation. Loss of CXXC5 led to inactivation of multiple inflammatory signaling pathways, while overexpression activated these pathways. Through in vitro and in vivo experiments, we confirmed ZNF143 and EGR1 as downstream transcription factors of CXXC5, mediating its proliferative potential in ovarian cancer. Our findings suggest that the CXXC5-ZNF143/EGR1 axis forms a network driving ovarian cell proliferation and tumorigenesis, and highlight CXXC5 as a potential therapeutic target for ovarian cancer treatment.
Collapse
Affiliation(s)
- Ting Geng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Qigang Sun
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Affiliated Hainan Hospital of Hainan Medical College, Haikou 570311, China
| | - Jingliang He
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yulu Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Wenhao Cheng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jing Shen
- Department of Obstetrics and Gynecology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Meiqi Zhang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Sen Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Kadirya Asan
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Mengwei Song
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Qi Gao
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yizhuo Song
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Ruotong Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xing Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yuanyuan Ding
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Aixin Jing
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiaoqing Ye
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Hongyu Ren
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Kaile Zeng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Ying Zhou
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Boyu Zhang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Shaojie Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| | - Wei Liu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Shunfang Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
6
|
Schlaeger L, Olejniczak I, Lehmann M, Schmidt CX, Astiz M, Oster H, Pilorz V. Estrogen-mediated coupling via gap junctions in the suprachiasmatic nucleus. Eur J Neurosci 2024; 59:1723-1742. [PMID: 38326974 DOI: 10.1111/ejn.16270] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
The circadian clock orchestrates many physiological and behavioural rhythms in mammals with 24-h periodicity, through a hierarchical organisation, with the central clock located in the suprachiasmatic nucleus (SCN) in the hypothalamus. The circuits of the SCN generate circadian rhythms with precision, relying on intrinsic coupling mechanisms, for example, neurotransmitters like arginine vasopressin (AVP), vasoactive intestinal peptide (VIP), neuronal gamma-aminobutyric acid (GABA) signalling and astrocytes connected by gap junctions composed of connexins (Cx). In female rodents, the presence of estrogen receptors (ERs) in the dorsal SCN suggests an influence of estrogen (E2) on the circuit timekeeping that could regulate circadian rhythm and coupling. To investigate this, we used SCN explants together with hypothalamic neurons and astrocytes. First, we showed that E2 stabilised the circadian amplitude in the SCN when rAVPs (receptor-associated vasopressin peptides) were inhibited. However, the phase delay induced by VIPAC2 (VIP receptors) inhibition remained unaffected by E2. We then showed that E2 exerted its effects in the SCN via ERβ (estrogen receptor beta), resulting in increased expression of Cx36 and Cx43. Notably, specific inhibition of both connexins resulted in a significant reduction in circadian amplitude within the SCN. Remarkably, E2 restored the period with inhibited Cx36 but not with Cx43 inhibition. This implies that the network between astrocytes and neurons, responsible for coupling in the SCN, can be reinforced through E2. In conclusion, these findings provide new insights into how E2 regulates circadian rhythms ex vivo in an ERβ-dependent manner, underscoring its crucial role in fortifying the SCN's rhythm.
Collapse
Affiliation(s)
- Lina Schlaeger
- Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, University of Lübeck, Lübeck, Germany
| | - Iwona Olejniczak
- Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, University of Lübeck, Lübeck, Germany
| | - Marianne Lehmann
- Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, University of Lübeck, Lübeck, Germany
| | - Cosima Xenia Schmidt
- Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, University of Lübeck, Lübeck, Germany
| | - Mariana Astiz
- Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, University of Lübeck, Lübeck, Germany
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Henrik Oster
- Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, University of Lübeck, Lübeck, Germany
| | - Violetta Pilorz
- Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, University of Lübeck, Lübeck, Germany
| |
Collapse
|
7
|
Turan G, Olgun ÇE, Ayten H, Toker P, Ashyralyyev A, Savaş B, Karaca E, Muyan M. Dynamic proximity interaction profiling suggests that YPEL2 is involved in cellular stress surveillance. Protein Sci 2024; 33:e4859. [PMID: 38145972 PMCID: PMC10804680 DOI: 10.1002/pro.4859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/27/2023]
Abstract
YPEL2 is a member of the evolutionarily conserved YPEL family involved in cellular proliferation, mobility, differentiation, senescence, and death. However, the mechanism by which YPEL2, or YPEL proteins, mediates its effects is largely unknown. Proteins perform their functions in a network of proteins whose identities, amounts, and compositions change spatiotemporally in a lineage-specific manner in response to internal and external stimuli. Here, we explored interaction partners of YPEL2 by using dynamic TurboID-coupled mass spectrometry analyses to infer a function for the protein. Our results using inducible transgene expressions in COS7 cells indicate that proximity interaction partners of YPEL2 are mainly involved in RNA and mRNA metabolic processes, ribonucleoprotein complex biogenesis, regulation of gene silencing by miRNA, and cellular responses to stress. We showed that YPEL2 interacts with the RNA-binding protein ELAVL1 and the selective autophagy receptor SQSTM1. We also found that YPEL2 localizes stress granules in response to sodium arsenite, an oxidative stress inducer, which suggests that YPEL2 participates in stress granule-related processes. Establishing a point of departure in the delineation of structural/functional features of YPEL2, our results suggest that YPEL2 may be involved in stress surveillance mechanisms.
Collapse
Affiliation(s)
- Gizem Turan
- Department of Biological SciencesMiddle East Technical UniversityAnkaraTürkiye
| | - Çağla Ece Olgun
- Department of Biological SciencesMiddle East Technical UniversityAnkaraTürkiye
| | - Hazal Ayten
- Department of Biological SciencesMiddle East Technical UniversityAnkaraTürkiye
| | - Pelin Toker
- Department of Biological SciencesMiddle East Technical UniversityAnkaraTürkiye
| | | | - Büşra Savaş
- İzmir Biomedicine and Genome CenterİzmirTürkiye
- Izmir International Biomedicine and Genome InstituteDokuz Eylül UniversityIzmirTürkiye
| | - Ezgi Karaca
- İzmir Biomedicine and Genome CenterİzmirTürkiye
- Izmir International Biomedicine and Genome InstituteDokuz Eylül UniversityIzmirTürkiye
| | - Mesut Muyan
- Department of Biological SciencesMiddle East Technical UniversityAnkaraTürkiye
- CanSyl LaboratoriesMiddle East Technical UniversityAnkaraTürkiye
| |
Collapse
|
8
|
Nuermaimaiti A, Chang L, Yan Y, Sun H, Xiao Y, Song S, Feng K, Lu Z, Ji H, Wang L. The role of sex hormones and receptors in HBV infection and development of HBV-related HCC. J Med Virol 2023; 95:e29298. [PMID: 38087447 DOI: 10.1002/jmv.29298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/02/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023]
Abstract
Gender disparity in hepatitis B virus (HBV)-related diseases has been extensively documented. Epidemiological studies consistently reported that males have a higher prevalence of HBV infection and incidence of hepatocellular carcinoma (HCC). Further investigations have revealed that sex hormone-related signal transductions play a significant role in gender disparity. Sex hormone axes showed significantly different responses to virus entry and replication. The sex hormones axes change the HBV-specific immune responses and antitumor immunity. Additionally, Sex hormone axes showed different effects on the development of HBV-related disease. But the role of sex hormones remains controversial, and researchers have not reached a consensus on the role of sex hormones and the use of hormone therapies in HCC treatment. In this review, we aim to summarize the experimental findings on sex hormones and provide a comprehensive understanding of their roles in the development of HCC and their implications for hormone-related HCC treatment.
Collapse
Affiliation(s)
- Abudulimutailipu Nuermaimaiti
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Le Chang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Ying Yan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Huizhen Sun
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingzi Xiao
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shi Song
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kaihao Feng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhuoqun Lu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Huimin Ji
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Lunan Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Erzurumlu Y, Dogan HK, Catakli D, Aydogdu E, Muhammed MT. Estrogens drive the endoplasmic reticulum-associated degradation and promote proto-oncogene c-Myc expression in prostate cancer cells by androgen receptor/estrogen receptor signaling. J Cell Commun Signal 2023; 17:793-811. [PMID: 36696010 PMCID: PMC10409964 DOI: 10.1007/s12079-022-00720-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/21/2022] [Indexed: 01/26/2023] Open
Abstract
The tumorigenic properties of prostate cancer are regulated by advanced hormonal regulation-mediated complex molecular signals. Therefore, characterizing the regulation of these signal transduction systems is crucial for understanding prostate cancer biology. Recent studies have shown that endoplasmic reticulum (ER)-localized protein quality control mechanisms, including ER-associated degradation (ERAD) and unfolded protein response (UPR) signaling contribute to prostate carcinogenesis and to the development of drug resistance. It has also been determined that these systems are tightly regulated by androgens. However, the role of estrogenic signaling in prostate cancer and its effects on protein quality control mechanisms is not fully understood. Herein, we investigated the regulatory effects of estrogens on ERAD and UPR and their impacts on prostate carcinogenesis. We found that estrogens strongly regulated the ERAD components and IRE1⍺ branch of UPR by Er⍺/β/AR axis. Besides, estrogenic signaling rigorously regulated the tumorigenicity of prostate cancer cells by promoting c-Myc expression and epithelial-mesenchymal transition (EMT). Moreover, estrogenic signal blockage significantly decreased the tumorigenic features of prostate cancer cells. Additionally, simultaneous inhibition of androgenic/estrogenic signals more efficiently inhibited tumorigenicity of prostate cancer cells, including proliferation, migration, invasion and colonial growth. Furthermore, computational-based molecular docking, molecular dynamics simulations and MMPBSA calculations supported the estrogenic stimulation of AR. Present findings suggested that ERAD components and IRE1⍺ signaling are tightly regulated by estrogen-stimulated AR and Er⍺/β. Our data suggest that treatment approaches targeting the co-inhibition of androgenic/estrogenic signals may pave the way for new treatment approaches to be developed for prostate cancer. The present model of the impact of estrogens on ERAD and UPR signaling in androgen-sensitive prostate cancer cells.
Collapse
Affiliation(s)
- Yalcin Erzurumlu
- Department of Biochemistry, Faculty of Pharmacy, Suleyman Demirel University, 32260 Isparta, Turkey
| | - Hatice Kubra Dogan
- Department of Bioengineering, Institute of Science, Suleyman Demirel University, 32260 Isparta, Turkey
| | - Deniz Catakli
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey
| | - Esra Aydogdu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Suleyman Demirel University, 32260 Isparta, Turkey
| | - Muhammed Tilahun Muhammed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Suleyman Demirel University, 32260 Isparta, Turkey
| |
Collapse
|
10
|
González-Gómez M, Reyes R, Damas-Hernández MDC, Plasencia-Cruz X, González-Marrero I, Alonso R, Bello AR. NTS, NTSR1 and ERs in the Pituitary-Gonadal Axis of Cycling and Postnatal Female Rats after BPA Treatment. Int J Mol Sci 2023; 24:ijms24087418. [PMID: 37108581 PMCID: PMC10138486 DOI: 10.3390/ijms24087418] [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: 03/09/2023] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
The neuropeptide neurotensin (NTS) is involved in regulating the reproductive axis and is expressed at each level of this axis (hypothalamus-pituitary-gonads). This dependence on estrogen levels has been widely demonstrated in the hypothalamus and pituitary. We focused on confirming the relationship of NTS with estrogens and the gonadal axis, using a particularly important environmental estrogenic molecule, bisphenol-A (BPA). Based on the experimental models or in vitro cell studies, it has been shown that BPA can negatively affect reproductive function. We studied for the first time the action of an exogenous estrogenic substance on the expression of NTS and estrogen receptors in the pituitary-gonadal axis during prolonged in vivo exposure. The exposure to BPA at 0.5 and 2 mg/kg body weight per day during gestation and lactation was monitored through indirect immunohistochemical procedures applied to the pituitary and ovary sections. Our results demonstrate that BPA induces alterations in the reproductive axis of the offspring, mainly after the first postnatal week. The rat pups exposed to BPA exhibited accelerated sexual maturation to puberty. There was no effect on the number of rats born per litter, although the fewer primordial follicles suggest a shorter fertile life.
Collapse
Affiliation(s)
- Miriam González-Gómez
- Departamento de Ciencias Médicas Básicas, Área de Anatomía Humana, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Spain
- Instituto de Tecnologías Biomédicas (ITB), 38200 La Laguna, Spain
- Instituto Universitario de Neurociencia (IUNE), Universidad de La Laguna, 38200 La Laguna, Spain
| | - Ricardo Reyes
- Instituto de Tecnologías Biomédicas (ITB), 38200 La Laguna, Spain
- Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Área de Biología Celular, Facultad de Ciencias, Sección de Biología, Universidad de La Laguna, 38200 La Laguna, Spain
- Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSP), 38296 La Laguna, Spain
| | | | - Xiomara Plasencia-Cruz
- Departamento de Ciencias Médicas Básicas, Área de Anatomía Humana, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Spain
| | - Ibrahim González-Marrero
- Departamento de Ciencias Médicas Básicas, Área de Anatomía Humana, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Spain
- Instituto Universitario de Neurociencia (IUNE), Universidad de La Laguna, 38200 La Laguna, Spain
| | - Rafael Alonso
- Instituto de Tecnologías Biomédicas (ITB), 38200 La Laguna, Spain
- Departamento de Ciencias Médicas Básicas, Área de Fisiología, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Spain
| | - Aixa R Bello
- Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Área de Biología Celular, Facultad de Ciencias, Sección de Biología, Universidad de La Laguna, 38200 La Laguna, Spain
- Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSP), 38296 La Laguna, Spain
| |
Collapse
|
11
|
Lisiak N, Dzikowska P, Wisniewska U, Kaczmarek M, Bednarczyk-Cwynar B, Zaprutko L, Rubis B. Biological Activity of Oleanolic Acid Derivatives HIMOXOL and Br-HIMOLID in Breast Cancer Cells Is Mediated by ER and EGFR. Int J Mol Sci 2023; 24:5099. [PMID: 36982173 PMCID: PMC10048893 DOI: 10.3390/ijms24065099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Breast cancer is one of the most frequently observed malignancies worldwide and represents a heterogeneous group of cancers. For this reason, it is crucial to properly diagnose every single case so a specific and efficient therapy can be adjusted. One of the most critical diagnostic parameters evaluated in cancer tissue is the status of the estrogen receptor (ER) and epidermal growth factor receptor (EGFR). Interestingly, the expression of the indicated receptors may be used in a personalized therapy approach. Importantly, the promising role of phytochemicals in the modulation of pathways controlled by ER and EGFR was also demonstrated in several types of cancer. One such biologically active compound is oleanolic acid, but due to poor water solubility and cell membrane permeability that limits its use, alternative derivative compounds were developed. These are HIMOXOL and Br-HIMOLID, which were demonstrated to be capable of inducing apoptosis and autophagy or diminishing the migratory and invasive potential of breast cancer cells in vitro. In our study, we revealed that proliferation, cell cycle, apoptosis, autophagy, and also the migratory potential of HIMOXOL and Br-HIMOLID in breast cancer cells are mediated by ER (MCF7) and EGFR (MDA-MB-231) receptors. These observations make the studied compounds interesting in the context of anticancer strategies.
Collapse
Affiliation(s)
- Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| | - Patrycja Dzikowska
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| | - Urszula Wisniewska
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Garbary 15 St., 61-866 Poznan, Poland
| | - Barbara Bednarczyk-Cwynar
- Department of Organic Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 St., 60-780 Poznan, Poland
| | - Lucjusz Zaprutko
- Department of Organic Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6 St., 60-780 Poznan, Poland
| | - Blazej Rubis
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| |
Collapse
|
12
|
Chromatin modifiers – Coordinators of estrogen action. Biomed Pharmacother 2022; 153:113548. [DOI: 10.1016/j.biopha.2022.113548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/20/2022] Open
|
13
|
Wu X, Dong W, Kong M, Ren H, Wang J, Shang L, Zhu Z, Zhu W, Shi X. Down-Regulation of CXXC5 De-Represses MYCL1 to Promote Hepatic Stellate Cell Activation. Front Cell Dev Biol 2021; 9:680344. [PMID: 34621736 PMCID: PMC8490686 DOI: 10.3389/fcell.2021.680344] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 08/24/2021] [Indexed: 12/23/2022] Open
Abstract
Liver fibrosis is mediated by myofibroblasts, a specialized cell type involved in wound healing and extracellular matrix production. Hepatic stellate cells (HSC) are the major source of myofibroblasts in the fibrotic livers. In the present study we investigated the involvement of CXXC-type zinc-finger protein 5 (CXXC5) in HSC activation and the underlying mechanism. Down-regulation of CXXC5 was observed in activated HSCs compared to quiescent HSCs both in vivo and in vitro. In accordance, over-expression of CXXC5 suppressed HSC activation. RNA-seq analysis revealed that CXXC5 influenced multiple signaling pathways to regulate HSC activation. The proto-oncogene MYCL1 was identified as a novel target for CXXC5. CXXC5 bound to the proximal MYCL1 promoter to repress MYCL1 transcription in quiescent HSCs. Loss of CXXC5 expression during HSC activation led to the removal of CpG methylation and acquisition of acetylated histone H3K9/H3K27 on the MYCL1 promoter resulting in MYCL1 trans-activation. Finally, MYCL1 knockdown attenuated HSC activation whereas MYCL1 over-expression partially relieved the blockade of HSC activation by CXXC5. In conclusion, our data unveil a novel transcriptional mechanism contributing to HSC activation and liver fibrosis.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Department of Hepatobiliary Surgery, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute of Nanjing University, Nanjing, China
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, and Center for Experimental Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Wenhui Dong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, and Center for Experimental Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, and Center for Experimental Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute of Nanjing University, Nanjing, China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute of Nanjing University, Nanjing, China
| | - Longcheng Shang
- Department of Hepatobiliary Surgery, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhengyi Zhu
- Department of Hepatobiliary Surgery, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei Zhu
- Department of Anesthesiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaolei Shi
- Department of Hepatobiliary Surgery, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute of Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Ayaz G, Turan G, Olgun ÇE, Kars G, Karakaya B, Yavuz K, Demiralay ÖD, Can T, Muyan M, Yaşar P. A prelude to the proximity interaction mapping of CXXC5. Sci Rep 2021; 11:17587. [PMID: 34475492 PMCID: PMC8413330 DOI: 10.1038/s41598-021-97060-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
CXXC5 is a member of the zinc-finger CXXC family proteins that interact with unmodified CpG dinucleotides through a conserved ZF-CXXC domain. CXXC5 is involved in the modulation of gene expressions that lead to alterations in diverse cellular events. However, the underlying mechanism of CXXC5-modulated gene expressions remains unclear. Proteins perform their functions in a network of proteins whose identities and amounts change spatiotemporally in response to various stimuli in a lineage-specific manner. Since CXXC5 lacks an intrinsic transcription regulatory function or enzymatic activity but is a DNA binder, CXXC5 by interacting with proteins could act as a scaffold to establish a chromatin state restrictive or permissive for transcription. To initially address this, we utilized the proximity-dependent biotinylation approach. Proximity interaction partners of CXXC5 include DNA and chromatin modifiers, transcription factors/co-regulators, and RNA processors. Of these, CXXC5 through its CXXC domain interacted with EMD, MAZ, and MeCP2. Furthermore, an interplay between CXXC5 and MeCP2 was critical for a subset of CXXC5 target gene expressions. It appears that CXXC5 may act as a nucleation factor in modulating gene expressions. Providing a prelude for CXXC5 actions, our results could also contribute to a better understanding of CXXC5-mediated cellular processes in physiology and pathophysiology.
Collapse
Affiliation(s)
- Gamze Ayaz
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey. .,Cancer and Stem Cell Epigenetics Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Gizem Turan
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | - Çağla Ece Olgun
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | - Gizem Kars
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | - Burcu Karakaya
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | - Kerim Yavuz
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | - Öykü Deniz Demiralay
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey
| | - Tolga Can
- Department of Computer Engineering Middle, East Technical University, 06800, Ankara, Turkey
| | - Mesut Muyan
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey. .,Cansyl Laboratories, Middle East Technical University, 06800, Ankara, Turkey.
| | - Pelin Yaşar
- Department of Biological Sciences, Middle East Technical University, 06800, Ankara, Turkey.,Epigenetics and Stem Cell Biology Laboratory, Single Cell Dynamics Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
15
|
Yaşar P, Kars G, Yavuz K, Ayaz G, Oğuztüzün Ç, Bilgen E, Suvacı Z, Çetinkol ÖP, Can T, Muyan M. A CpG island promoter drives the CXXC5 gene expression. Sci Rep 2021; 11:15655. [PMID: 34341443 PMCID: PMC8329181 DOI: 10.1038/s41598-021-95165-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
CXXC5 is a member of the zinc-finger CXXC family that binds to unmethylated CpG dinucleotides. CXXC5 modulates gene expressions resulting in diverse cellular events mediated by distinct signaling pathways. However, the mechanism responsible for CXXC5 expression remains largely unknown. We found here that of the 14 annotated CXXC5 transcripts with distinct 5' untranslated regions encoding the same protein, transcript variant 2 with the highest expression level among variants represents the main transcript in cell models. The DNA segment in and at the immediate 5'-sequences of the first exon of variant 2 contains a core promoter within which multiple transcription start sites are present. Residing in a region with high G-C nucleotide content and CpG repeats, the core promoter is unmethylated, deficient in nucleosomes, and associated with active RNA polymerase-II. These findings suggest that a CpG island promoter drives CXXC5 expression. Promoter pull-down revealed the association of various transcription factors (TFs) and transcription co-regulatory proteins, as well as proteins involved in histone/chromatin, DNA, and RNA processing with the core promoter. Of the TFs, we verified that ELF1 and MAZ contribute to CXXC5 expression. Moreover, the first exon of variant 2 may contain a G-quadruplex forming region that could modulate CXXC5 expression.
Collapse
Affiliation(s)
- Pelin Yaşar
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey.
- Epigenetics and Stem Cell Biology Laboratory, Single Cell Dynamics Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA.
| | - Gizem Kars
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey
| | - Kerim Yavuz
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey
| | - Gamze Ayaz
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey
- Cancer and Stem Cell Epigenetics Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Çerağ Oğuztüzün
- Department of Computer Engineering, Bilkent University, Ankara, 06800, Turkey
| | - Ecenaz Bilgen
- Department of Chemistry, Middle East Technical University, Ankara, 06800, Turkey
| | - Zeynep Suvacı
- Department of Chemistry, Middle East Technical University, Ankara, 06800, Turkey
| | | | - Tolga Can
- Department of Computer Engineering, Middle East Technical University, Ankara, 06800, Turkey
| | - Mesut Muyan
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey.
- Cansyl Laboratories, Middle East Technical University, Ankara, 06800, Turkey.
| |
Collapse
|
16
|
Zearalenone and the Immune Response. Toxins (Basel) 2021; 13:toxins13040248. [PMID: 33807171 PMCID: PMC8066068 DOI: 10.3390/toxins13040248] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/20/2021] [Accepted: 03/27/2021] [Indexed: 12/12/2022] Open
Abstract
Zearalenone (ZEA) is an estrogenic fusariotoxin, being classified as a phytoestrogen, or as a mycoestrogen. ZEA and its metabolites are able to bind to estrogen receptors, 17β-estradiol specific receptors, leading to reproductive disorders which include low fertility, abnormal fetal development, reduced litter size and modification at the level of reproductive hormones especially in female pigs. ZEA has also significant effects on immune response with immunostimulatory or immunosuppressive results. This review presents the effects of ZEA and its derivatives on all levels of the immune response such as innate immunity with its principal component inflammatory response as well as the acquired immunity with two components, humoral and cellular immune response. The mechanisms involved by ZEA in triggering its effects are addressed. The review cited more than 150 publications and discuss the results obtained from in vitro and in vivo experiments exploring the immunotoxicity produced by ZEA on different type of immune cells (phagocytes related to innate immunity and lymphocytes related to acquired immunity) as well as on immune organs. The review indicates that despite the increasing number of studies analyzing the mechanisms used by ZEA to modulate the immune response the available data are unsubstantial and needs further works.
Collapse
|
17
|
Astori A, Matherat G, Munoz I, Gautier EF, Surdez D, Zermati Y, Verdier F, Zaidi S, Feuillet V, Kadi A, Lauret E, Delattre O, Lefèvre C, Fontenay M, Ségal-Bendirdjian E, Dusanter-Fourt I, Bouscary D, Hermine O, Mayeux P, Pendino F. The epigenetic regulator RINF (CXXC5) maintains <i>SMAD7</i> expression in human immature erythroid cells and sustains red blood cells expansion. Haematologica 2020; 107:268-283. [PMID: 33241676 PMCID: PMC8719099 DOI: 10.3324/haematol.2020.263558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Indexed: 11/16/2022] Open
Abstract
The gene CXXC5, encoding a retinoid-inducible nuclear factor (RINF), is located within a region at 5q31.2 commonly deleted in myelodysplastic syndrome and adult acute myeloid leukemia. RINF may act as an epigenetic regulator and has been proposed as a tumor suppressor in hematopoietic malignancies. However, functional studies in normal hematopoiesis are lacking, and its mechanism of action is unknown. Here, we evaluated the consequences of RINF silencing on cytokine-induced erythroid differentiation of human primary CD34+ progenitors. We found that RINF is expressed in immature erythroid cells and that RINF-knockdown accelerated erythropoietin-driven maturation, leading to a significant reduction (~45%) in the number of red blood cells, without affecting cell viability. The phenotype induced by RINF-silencing was dependent on tumor growth factor b (TGFb) and mediated by SMAD7, a TGFb-signaling inhibitor. RINF upregulates SMAD7 expression by direct binding to its promoter and we found a close correlation between RINF and SMAD7 mRNA levels both in CD34+ cells isolated from bone marrow of healthy donors and myelodysplastic syndrome patients with del(5q). Importantly, RINF knockdown attenuated SMAD7 expression in primary cells and ectopic SMAD7 expression was sufficient to prevent the RINF knockdown-dependent erythroid phenotype. Finally, RINF silencing affects 5’-hydroxymethylation of human erythroblasts, in agreement with its recently described role as a TET2-anchoring platform in mouse. Collectively, our data bring insight into how the epigenetic factor RINF, as a transcriptional regulator of SMAD7, may fine-tune cell sensitivity to TGFb superfamily cytokines and thus play an important role in both normal and pathological erythropoiesis.
Collapse
Affiliation(s)
- Audrey Astori
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Gabriel Matherat
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Isabelle Munoz
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Emilie-Fleur Gautier
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Didier Surdez
- Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France; PSL Research University, Institut Curie Research Center, INSERM U830, Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris
| | - Yaël Zermati
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris
| | - Frédérique Verdier
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris
| | - Sakina Zaidi
- Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France; PSL Research University, Institut Curie Research Center, INSERM U830, Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris
| | - Vincent Feuillet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris
| | - Amir Kadi
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris
| | - Evelyne Lauret
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Olivier Delattre
- Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France; PSL Research University, Institut Curie Research Center, INSERM U830, Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris
| | - Carine Lefèvre
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris
| | - Michaela Fontenay
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris, France; Service d'Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Paris
| | | | - Isabelle Dusanter-Fourt
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Didier Bouscary
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Olivier Hermine
- Laboratory of Excellence GR-ex, Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris, France; Université de Paris, Institut Imagine, INSERM, CNRS, F-75015, Paris
| | - Patrick Mayeux
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris
| | - Frédéric Pendino
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France; Laboratory of Excellence GR-ex, Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer (LNCC), Paris.
| |
Collapse
|
18
|
Kovács T, Szabó-Meleg E, Ábrahám IM. Estradiol-Induced Epigenetically Mediated Mechanisms and Regulation of Gene Expression. Int J Mol Sci 2020; 21:ijms21093177. [PMID: 32365920 PMCID: PMC7246826 DOI: 10.3390/ijms21093177] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Gonadal hormone 17β-estradiol (E2) and its receptors are key regulators of gene transcription by binding to estrogen responsive elements in the genome. Besides the classical genomic action, E2 regulates gene transcription via the modification of epigenetic marks on DNA and histone proteins. Depending on the reaction partner, liganded estrogen receptor (ER) promotes DNA methylation at the promoter or enhancer regions. In addition, ERs are important regulators of passive and active DNA demethylation. Furthermore, ERs cooperating with different histone modifying enzymes and chromatin remodeling complexes alter gene transcription. In this review, we survey the basic mechanisms and interactions between estrogen receptors and DNA methylation, demethylation and histone modification processes as well as chromatin remodeling complexes. The particular relevance of these mechanisms to physiological processes in memory formation, embryonic development, spermatogenesis and aging as well as in pathophysiological changes in carcinogenesis is also discussed.
Collapse
Affiliation(s)
- Tamás Kovács
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pécs, Hungary;
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, H-7624 Pécs, Hungary;
| | - István M. Ábrahám
- Molecular Neuroendocrinology Research Group, Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Center, University of Pécs, H-7624 Pécs, Hungary;
- Correspondence:
| |
Collapse
|
19
|
Ayaz G, Razizadeh N, Yaşar P, Kars G, Kahraman DC, Saatci Ö, Şahin Ö, Çetin-Atalay R, Muyan M. CXXC5 as an unmethylated CpG dinucleotide binding protein contributes to estrogen-mediated cellular proliferation. Sci Rep 2020; 10:5971. [PMID: 32249801 PMCID: PMC7136269 DOI: 10.1038/s41598-020-62912-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Evidence suggests that the CXXC type zinc finger (ZF-CXXC) protein 5 (CXXC5) is a critical regulator/integrator of various signaling pathways that include the estrogen (E2)-estrogen receptor α (ERα). Due to its ZF-CXXC domain, CXXC5 is considered to be a member of the ZF-CXXC family, which binds to unmethylated CpG dinucleotides of DNA and through enzymatic activities for DNA methylation and/or chromatin modifications generates a chromatin state critical for gene expressions. Structural/functional features of CXXC5 remain largely unknown. CXXC5, suggested as transcription and/or epigenetic factor, participates in cellular proliferation, differentiation, and death. To explore the role of CXXC5 in E2-ERα mediated cellular events, we verified by generating a recombinant protein that CXXC5 is indeed an unmethylated CpG binder. We uncovered that CXXC5, although lacks a transcription activation/repression function, participates in E2-driven cellular proliferation by modulating the expression of distinct and mutual genes also regulated by E2. Furthermore, we found that the overexpression of CXXC5, which correlates with mRNA and protein levels of ERα, associates with poor prognosis in ER-positive breast cancer patients. Thus, CXXC5 as an unmethylated CpG binder contributes to E2-mediated gene expressions that result in the regulation of cellular proliferation and may contribute to ER-positive breast cancer progression.
Collapse
Affiliation(s)
- Gamze Ayaz
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey.,Cancer and Stem Cell Epigenetics Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Negin Razizadeh
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey
| | - Pelin Yaşar
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey
| | - Gizem Kars
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey
| | - Deniz Cansen Kahraman
- Enformatics Institute, Middle East Technical University, Ankara, 06800, Turkey.,Cansyl Laboratories, Middle East Technical University, Ankara, 06800, Turkey
| | - Özge Saatci
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Özgür Şahin
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA.,Department of Molecular Biology and Genetics, Bilkent University, Ankara, 06800, Turkey
| | - Rengül Çetin-Atalay
- Enformatics Institute, Middle East Technical University, Ankara, 06800, Turkey.,Cansyl Laboratories, Middle East Technical University, Ankara, 06800, Turkey
| | - Mesut Muyan
- Department of Biological Sciences, Middle East Technical University, Ankara, 06800, Turkey. .,Cansyl Laboratories, Middle East Technical University, Ankara, 06800, Turkey.
| |
Collapse
|
20
|
Choi S, Kim HY, Cha PH, Seo SH, Lee C, Choi Y, Shin W, Heo Y, Han G, Lee W, Choi KY. CXXC5 mediates growth plate senescence and is a target for enhancement of longitudinal bone growth. Life Sci Alliance 2019; 2:2/2/e201800254. [PMID: 30971423 PMCID: PMC6458850 DOI: 10.26508/lsa.201800254] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022] Open
Abstract
Longitudinal bone growth ceases with growth plate senescence during puberty. However, the molecular mechanisms of this phenomenon are largely unexplored. Here, we examined Wnt-responsive genes before and after growth plate senescence and found that CXXC finger protein 5 (CXXC5), a negative regulator of the Wnt/β-catenin pathway, was gradually elevated with reduction of Wnt/β-catenin signaling during senescent changes of rodent growth plate. Cxxc5 -/- mice demonstrated delayed growth plate senescence and tibial elongation. As CXXC5 functions by interacting with dishevelled (DVL), we sought to identify small molecules capable of disrupting this interaction. In vitro screening assay monitoring CXXC5-DVL interaction revealed that several indirubin analogs were effective antagonists of this interaction. A functionally improved indirubin derivative, KY19382, elongated tibial length through delayed senescence and further activation of the growth plate in adolescent mice. Collectively, our findings reveal an important role for CXXC5 as a suppressor of longitudinal bone growth involving growth plate activity.
Collapse
Affiliation(s)
- Sehee Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Hyun-Yi Kim
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Pu-Hyeon Cha
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Seol Hwa Seo
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Chulho Lee
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Yejoo Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Wookjin Shin
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Yunseok Heo
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Gyoonhee Han
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Weontae Lee
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea.,Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kang-Yell Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Korea .,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea.,CK Biotechnology Inc, Seoul, Korea
| |
Collapse
|
21
|
Coughlin SS, Kapuku G. Physical Activity, Weight Control, and Biomarkers of Prognosis and Survival among Breast Cancer Survivors. ARCHIVES OF EPIDEMIOLOGY 2018; 3:10.29011/2257-2252.100027. [PMID: 31179439 PMCID: PMC6550489 DOI: 10.29011/2257-2252.100027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Physical inactivity and obesity may increase risk of poor prognosis in breast cancer through effects on insulin or insulin-like growth factors or their binding proteins, insulin resistance, glucose metabolism, sex hormones, leptin and other adipokines, immunologic or inflammatory factors, oxidative stress, and Deoxyribonucleic acid (DNA) damage or repair capacity. The present review is based upon bibliographic searches in PubMed and relevant search terms. Articles published in English from January 1, 1980 through October 1, 2018 were identified using the following MeSH search terms and Boolean algebra commands: breast cancer survivors AND (insulin-like growth factor OR insulin resistance OR glucose metabolism OR sex hormones OR leptin OR adipokines OR immunologic OR inflammatory factors OR oxidative stress OR DNA repair capacity). After screening the abstracts or full texts of these articles and reviewing the references of previous review articles, a total of 66 studies met the eligibility criteria. Based upon published studies, it is difficult to determine the type or dose of exercise that affects inflammatory markers among breast cancer survivors. The optimal type of exercise, dose, and timing of physical activity needed to improve the inflammatory profile following a breast cancer diagnosis is unknown. Studies have used a range of physical activity types including aerobic, resistance training, yoga, and Tai Chi. A further issue is that existing studies of physical activity and biomarkers have included a range of disease stages. There is a need for a better understanding of the biological pathways through which physical activity and weight management increase survival in order to design targeted weight loss and exercise interventions for breast cancer survivors.
Collapse
Affiliation(s)
- Steven Scott Coughlin
- Department of Population Health Sciences, Augusta University, USA
- Research Service, Charlie Norwood Veterans Administration Medical Center, USA
| | - Gaston Kapuku
- Department of Population Health Sciences, Augusta University, USA
| |
Collapse
|
22
|
Xiong X, Tu S, Wang J, Luo S, Yan X. CXXC5: A novel regulator and coordinator of TGF-β, BMP and Wnt signaling. J Cell Mol Med 2018; 23:740-749. [PMID: 30479059 PMCID: PMC6349197 DOI: 10.1111/jcmm.14046] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/23/2018] [Indexed: 12/18/2022] Open
Abstract
CXXC5 is a member of the CXXC-type zinc-finger protein family. Proteins in this family play a pivotal role in epigenetic regulation by binding to unmethylated CpG islands in gene promoters through their characteristic CXXC domain. CXXC5 is a short protein (322 amino acids in length) that does not have any catalytic domain, but is able to bind to DNA and act as a transcription factor and epigenetic factor through protein-protein interactions. Intriguingly, increasing evidence indicates that expression of the CXXC5 gene is controlled by multiple signaling pathways and a variety of transcription factors, positioning CXXC5 as an important signal integrator. In addition, CXXC5 is capable of regulating various signal transduction processes, including the TGF-β, Wnt and ATM-p53 pathways, thereby acting as a novel and crucial signaling coordinator. CXXC5 plays an important role in embryonic development and adult tissue homeostasis by regulating cell proliferation, differentiation and apoptosis. In keeping with these functions, aberrant expression or altered activity of CXXC5 has been shown to be involved in several human diseases including tumourigenesis. This review summarizes the current understanding of CXXC5 as a transcription factor and signaling regulator and coordinator.
Collapse
Affiliation(s)
- Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Shuo Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Jianbin Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| |
Collapse
|
23
|
Fang L, Wang Y, Gao Y, Chen X. Overexpression of CXXC5 is a strong poor prognostic factor in ER+ breast cancer. Oncol Lett 2018; 16:395-401. [PMID: 29928427 PMCID: PMC6006432 DOI: 10.3892/ol.2018.8647] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
CXXC5 is a newly identified CXXC-type zinc finger family protein, which is encoded by the CXXC5 gene localised to the 5q31.3 chromosomal region. Previous studies revealed that CXXC5 is associated with various malignant tumours. The aim of the present study was to investigate the prognosis prediction of CXXC5 in different breast cancer subtypes via the Gene Expression Omnibus database and bc-GenExMiner. CXXC5 overexpression was observed as associated with a poor prognosis for oestrogen receptor positive (ER+) breast cancer. Basal-like breast cancer and triple-negative breast cancer also suggest a poor prognosis, however their CXXC5 expression was low and could not be used as a prognostic factor. The CXXC5 correlated genes and their enriched Gene Ontology (GO) terms were obtained. Among those enriched GO terms, GO:0070062 (extracellular exosome) had the greatest number of associated genes and the associated genes of GO:0000122 (negative regulation of transcription from RNA polymerase II promoter) and GO:0008134 (transcription factor binding) contained CXXC5. These results suggest that overexpression of CXXC5 is a strongly poor prognostic factor in ER+ breast cancer. However, the role of CXXC5 in breast cancer requires further investigation.
Collapse
Affiliation(s)
- Lei Fang
- Department of Pathology and Pathophysiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Yu Wang
- Department of Radiology and NFCR Center for Molecular Imaging, Case Western Reserve University, Cleveland, OH 44106-5065, USA
| | - Yang Gao
- Department of Oncology, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Xuejun Chen
- Department of Pathology and Pathophysiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|