1
|
Jia W, Wang G, Sun S, Chen X, Xiang S, Zhang B, Huang Z. PA2G4 in health and disease: An underestimated multifunctional regulator. J Adv Res 2025:S2090-1232(25)00074-8. [PMID: 39923993 DOI: 10.1016/j.jare.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/23/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Proliferation-associated protein 2G4 (PA2G4), also known as ErbB3-binding protein 1 (EBP1), is an evolutionarily conserved, ubiquitously expressed, multifunctional factor in health and disease. In recent decades, its role as a sophisticated regulator in a broad range of biological processes has drawn widespread attention from researchers. AIM OF REVIEW We introduce the molecular structure, functional modules, and post-translational modifications of PA2G4. We further elaborate on its role and function in immune microenvironment modulation, cell growth, neural homeostasis and embryonic development. In particular, we summarize its relevance to tumorigenesis and cancer progression and describe its molecular mechanisms in regulating the hallmarks of cancers. This review aims to provide a comprehensive blueprint of PA2G4 functions and to inspire further basic and translational studies. KEY SCIENTIFIC CONCEPTS OF REVIEW Owing to its versatile domains and motifs, PA2G4 regulates a variety of molecular processes, including transcription, translation, proteostasis and epigenetic modulation, suggesting its critical roles in maintaining homeostasis. There are two isoforms of the PA2G4 protein: PA2G4-p42 and PA2G4-p48. While both isoforms regulate cellular activities, they often exert distinct or even contradictory effects. Dysfunction and aberrant expression of PA2G4 isoforms lead to the occurrence and progression of various diseases, indicating their role as predictive markers or therapeutic targets.
Collapse
Affiliation(s)
- Wenlong Jia
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaocheng Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; The Second Clinical Department, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Sun
- Department of Pharmacy, Taikang Tongji (Wuhan) Hospital, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; The Second Clinical Department, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Xiang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China; Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China; Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Li J, Wang Y, Niu X, Xing J, Dou Y, Yao Y, Liang Y, Wang Z, Wang Q, Feng H, Peng A, Guo Y, Liang H, Guo Y, Li J, Hou R, Cheng Y, Qu T, Li X, Yin G, Zhang K. Downregulation of lysine 2-hydroxyisobutyrylation of ErbB3 binding protein 1 at amino acid 210 promotes keratinocyte proliferation via induction of transcription initiation factor IA-mediated rRNA synthesis. Br J Dermatol 2024; 192:92-103. [PMID: 38752336 DOI: 10.1093/bjd/ljae194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/17/2024] [Accepted: 05/04/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Psoriasis is a prevalent chronic inflammatory dermatosis characterized by the excessive proliferation of keratinocytes (KCs). Lysine 2-hydroxyisobutyrylation (Khib) is a newly identified post-translational modification that regulates various biological processes. Abnormal Khib modification has been associated with the development of autoimmune diseases. OBJECTIVES To investigate the abnormal Khib modification profile and its pathogenic role in psoriasis. METHODS Liquid chromatography-tandem mass spectrometry was used to analyse Khib-modified proteins in the epidermis of psoriasis lesions and healthy control skin. Mutated cells and mice with downregulated Khib modification of ErbB3 binding protein 1 (EBP1) at amino acid 210 (EBP1Khib210) were generated, to investigate the functional effects of EBP1Khib210 in psoriasis. RESULTS Omics analysis revealed dysregulation of Khib modification in psoriatic lesions, exhibiting a distinct profile compared with controls. We found downregulation of EBP1Khib210 in psoriatic lesions and mice with imiquimod-induced psoriasis. Notably, expression of EBP1Khib210 was upregulated in patients with psoriasis following effective treatment. Decreased EBP1Khib210 enhanced KC viability, proliferation and survival but inhibited apoptosis in vitro. Additionally, Pa2g4K210A mice with downregulated Ebp1Khib210 exhibited more severe psoriatic lesions and enhanced KC proliferation. Moreover, we found that the EBP1K210A mutation increased the interaction between EBP1 and nuclear protein kinase B (Akt), thereby inhibiting mouse double minute 2-mediated transcription initiation factor IA (TIF-IA) ubiquitination and resulting in increased rRNA synthesis and KC proliferation. Downregulation of EBP1Khib210 was attributed to an inflammation-induced increase in histone deacetylase 2 expression. CONCLUSIONS Downregulation of EBP1Khib210 promoted KC proliferation by modulating Akt signalling and TIF-IA-mediated rRNA synthesis. These insights into Khib modification provide better understanding of the pathogenesis of psoriasis and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Junqin Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Ying Wang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Xuping Niu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Jianxiao Xing
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Yu Dou
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Yuanjun Yao
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Yanyang Liang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Zehong Wang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Qiang Wang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Haiyan Feng
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Aihong Peng
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Yi Guo
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Huifang Liang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Yaxin Guo
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Juan Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Ruixia Hou
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Yueai Cheng
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Tong Qu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Xinhua Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Guohua Yin
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, China
| |
Collapse
|
3
|
Wang Y, Xing J, Liang Y, Liang H, Liang N, Li J, Yin G, Li X, Zhang K. The structure and function of multifunctional protein ErbB3 binding protein 1 (Ebp1) and its role in diseases. Cell Biol Int 2024; 48:1069-1079. [PMID: 38884348 DOI: 10.1002/cbin.12196] [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: 12/10/2023] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024]
Abstract
ErbB3-binding protein 1(Ebp1) has two isoforms, p42 Ebp1 and p48 Ebp1, both of which can regulate cell growth and differentiation. But these isoforms often have opposite effects, including contradictory roles in regulation of cell growth in different tissues and cells. P48 Ebp1 belongs to the full-length sequence, while conformational changes in the crystal structure of p42 Ebp1 reveals a lack of an α helix at the amino terminus. Due to the differences in the structures of these two isoforms, they have different binding partners and protein modifications. Ebp1 can function as both an oncogene and a tumor suppressor factor. However, the underlying mechanisms by which these two isoforms exert opposite functions are still not fully understood. In this review, we summarize the genes and the structures of protein of these two isoforms, protein modifications, binding partners and the association of different isoforms with diseases.
Collapse
Affiliation(s)
- Ying Wang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Jianxiao Xing
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanyang Liang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Huifang Liang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Nannan Liang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Junqin Li
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Guohua Yin
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Xinhua Li
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Kaiming Zhang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
CHOUGULE BS, GAURAV K, KUMAR M, MAHADEVAPRASAD N, KRISHNA NH, PONNADA SL, DERANGULA S, NADUMANE VK. Proteomic Analysis of HepG2 Cells Reveals FAT10 and BAG2 Signaling Pathways Affected by a Protease Inhibitor from Tinospora cordifolia (Willd.) Hook. f. and Thoms Stem. Extract Among the Different Plant and Microbial Samples Analyzed. Turk J Pharm Sci 2024; 21:174-183. [PMID: 38994797 PMCID: PMC11590549 DOI: 10.4274/tjps.galenos.2023.75668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/20/2023] [Indexed: 07/13/2024]
Abstract
Objectives Dysregulation of proteolysis underlies diseases like cancer. Protease inhibitors (PIs) regulate many biological functions and hence have potential anticancer properties. With this background, the current study aimed to identify the PI from natural sources such as plants and microbes against trypsin (a protease), which was assayed against casein, using an ultraviolet spectrophotometer-based methodology. Materials and Methods PI extracted from a few plants and microbial samples were screened for their PI activity against trypsin. The PI from the most promising source in our study, Tinospora cordifolia (Willd.) Hook. f. and Thoms. stem, was partially purified using ammonium sulfate precipitation followed by dialysis. The PI activity of the partially purified inhibitor was analyzed against chymotrypsin and collagenase enzymes, and the cytotoxic effect of the PI was checked on HepG2 (liver carcinoma) cells by MTT- [3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide]- assay. Liquid Chromatograography Mass Spectrometry -based proteomic studies were performed on HepG2 cells to understand the signaling pathways affected by the PIs in the liver cancer cell line. Results Among the samples tested the PIs from T. cordifolia stem extract had the highest inhibitory activity (72.0%) against trypsin along with cytotoxicity to HepG2 cells. After partial purification by 80.0% ammonium sulfate precipitation, PI had increased inhibitory activity (83.0%) against trypsin and enhanced cytotoxicity (47.0%) to HepG2 cells. Proteomic analysis of the PI-treated HepG2 cells revealed that BAG2 and FAT10 signaling pathways were affected, which may have caused the inhibition of cancer cell proliferation. Conclusion PI from T. cordifolia stem has promising anticancer potential and hence can be used for further purification and characterization studies toward cancer drug development.
Collapse
Affiliation(s)
- Bramhi Suresh CHOUGULE
- Jain (Deemed-to-be-University), Department of Biotechnology, School of Sciences, Karnataka, India
| | - Kumar GAURAV
- Jain (Deemed-to-be-University), Department of Biotechnology, School of Sciences, Karnataka, India
| | - Mutthu KUMAR
- Jain (Deemed-to-be-University), Department of Biotechnology, School of Sciences, Karnataka, India
| | - Nayana MAHADEVAPRASAD
- Jain (Deemed-to-be-University), Department of Biotechnology, School of Sciences, Karnataka, India
| | | | - Sreya Lakshmi PONNADA
- Jain (Deemed-to-be-University), Department of Biotechnology, School of Sciences, Karnataka, India
| | - Somasekhara DERANGULA
- Jain (Deemed-to-be-University), Department of Biotechnology, School of Sciences, Karnataka, India
| | | |
Collapse
|
5
|
Naeem A, Knoer G, Avantaggiati ML, Rodriguez O, Albanese C. Provocative non-canonical roles of p53 and AKT signaling: A role for Thymosin β4 in medulloblastoma. Int Immunopharmacol 2023; 116:109785. [PMID: 36720193 DOI: 10.1016/j.intimp.2023.109785] [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: 11/23/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/30/2023]
Abstract
The PI3K/AKT and p53 pathways are key regulators of cancer cell survival and death, respectively. Contrary to their generally accepted roles, several lines of evidence, including ours in medulloblastoma, the most common childhood brain cancer, highlight non-canonical functions for both proteins and show a complex context-dependent dynamic behavior in determining cell fate. Interestingly, p53-mediated cell survival and AKT-mediated cell death can dominate in certain conditions, and these interchangeable physiological functions may potentially be manipulated for better clinical outcomes. This review article presents studies in which p53 and AKT behave contrary to their well-established functions. We discuss the factors and circumstances that may be involved in mediating these changes and the implications of these unique roles of p53 and AKT in devising therapeutic strategies. Lastly, based on our recent finding of Thymosin beta 4-mediated chemosensitivity via an AKT-p53 interaction in medulloblastoma cells, we also discuss the possible implications of Thymosin beta-4 in enhancing drug sensitivity in this deadly childhood disease.
Collapse
Affiliation(s)
- Aisha Naeem
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Health Research Governance Department, Ministry of Public Health, Qatar.
| | - Grace Knoer
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Maria Laura Avantaggiati
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Olga Rodriguez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Center for Translational Imaging, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Chris Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Radiology, Georgetown University Medical Center, Washington, DC 20057, USA; Center for Translational Imaging, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
6
|
Kim BS, Ko HR, Hwang I, Cho SW, Ahn JY. EBP1 regulates Suv39H1 stability via the ubiquitin-proteasome system in neural development. BMB Rep 2021. [PMID: 33691908 PMCID: PMC8411044 DOI: 10.5483/bmbrep.2021.54.8.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
ErbB3-binding protein 1 (EBP1) is a multifunctional protein associated with neural development. Loss of Ebp1 leads to upregulation of the gene silencing unit suppressor of variegation 3-9 homolog 1 (Suv39H1)/DNA (cytosine 5)-methyltransferase (DNMT1). EBP1 directly binds to the promoter region of DNMT1, repressing DNA methylation, and hence, promoting neural development. In the current study, we showed that EBP1 suppresses histone methyltransferase activity of Suv39H1 by promoting ubiquitin-proteasome system (UPS)-dependent degradation of Suv39H1. In addition, we showed that EBP1 directly interacts with Suv39H1, and this interaction is required for recruiting the E3 ligase MDM2 for Suv39H1 degradation. Thus, our findings suggest that EBP1 regulates UPS-dependent degradation of Suv39H1 to govern proper heterochromatin assembly during neural development.
Collapse
Affiliation(s)
- Byeong-Seong Kim
- Department of Molecular Cell Biology, Suwon 16419, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Hyo Rim Ko
- Department of Molecular Cell Biology, Suwon 16419, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Inwoo Hwang
- Department of Molecular Cell Biology, Suwon 16419, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan, College of Medicine, Seoul 05505, Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Suwon 16419, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
7
|
Seo L, Kim YI, Kim H, Hyun K, Kim J, Lee JE. Discovery of Klf2 interactors in mouse embryonic stem cells by immunoprecipitation-mass spectrometry utilizing exogenously expressed bait. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140672. [PMID: 34000451 DOI: 10.1016/j.bbapap.2021.140672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/04/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
Krüppel-like factor 2 (Klf2) is a DNA-binding transcription factor that regulates embryonic stem cell-specific gene expression. Transcription cofactors such as p300 acetyltransferase and Erk kinases interact with Klf2, providing an additional layer of transcription regulation in embryonic stem cells. To carry out a thorough survey of the Klf2 interactome in embryonic stem cells and identify novel transcription cofactors, we designed a modified immunoprecipitation-mass spectrometry (IP-MS) method. In this method, recombinant Klf2, expressed and purified from Sf9 insect cells instead of ectopically expressed in cells, was used as bait. Using this modified IP-MS method, we discovered nine Klf2-interacting proteins, including the previously reported Crebbp and p300. These proteins showed at least an 8-fold increase in signal intensity in Klf2 pull-downs compared with controls, with P-values <0.010. Among the identified Klf2-binding proteins confirmed using our IP-MS workflow was Snd1, which we found to interact directly with Klf2 and function as a transcriptional coactivator of Klf2 to drive the Oct4 gene expression. Collectively, our IP-MS protocol may offer a useful tool for identifying novel transcription cofactors in stem cells.
Collapse
Affiliation(s)
- Lin Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Yong-In Kim
- Center for Bioanalysis, Korea Research Institute of Standards and Science, Daejeon 34113, South Korea
| | - Hyoungmin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Kwangbeom Hyun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Jaehoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea.
| | - J Eugene Lee
- Division of Policy and Strategy, Korea Research Institute of Standards and Science, Daejeon 34113, South Korea.
| |
Collapse
|
8
|
Pini T, Haywood M, McCallie B, Lane SL, Schoolcraft WB, Katz-Jaffe M. Liquid chromatography-tandem mass spectrometry reveals an active response to DNA damage in human spermatozoa. F&S SCIENCE 2021; 2:153-163. [PMID: 35559750 DOI: 10.1016/j.xfss.2021.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/23/2021] [Accepted: 03/13/2021] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To investigate how endogenously elevated DNA fragmentation alters the human sperm proteome, and whether this fragmentation contributes to genomic deletions. DESIGN Research study. SETTING Commercial fertility clinic. PATIENT(S) Men with low (0%-4%, n = 7) or high (≥16%, n = 6) sperm DNA fragmentation, as assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Global sperm proteome, single-nucleotide polymorphism genotyping array. RESULT(S) A total of 78 significantly differentially abundant proteins (30 decreased, 48 increased) were observed in control vs. high DNA damage samples. DNA damage resulted in robust proteomic responses, including markers of oxidative stress and apoptosis, DNA damage repair proteins, and transcription/translation and protein turnover machinery. Several key sperm functional proteins were significantly decreased in ejaculates with high DNA damage. We were unable to substantiate a link between increased DNA fragmentation and genomic deletions in human spermatozoa. CONCLUSION(S) Developing human spermatozoa initiate an active transcriptional response to endogenous DNA damage, which manifests as alterations in the sperm proteome.
Collapse
Affiliation(s)
- Taylor Pini
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado.
| | - Mary Haywood
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado
| | - Blair McCallie
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado
| | - Sydney L Lane
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado
| | | | | |
Collapse
|
9
|
Sharma P, Nath H, Frontera A, Barcelo-Oliver M, Verma AK, Hussain S, Bhattacharyya MK. Biologically relevant unusual cooperative assemblies and fascinating infinite crown-like supramolecular nitrate–water hosts involving guest complex cations in bipyridine and phenanthroline-based Cu( ii) coordination compounds: antiproliferative evaluation and theoretical studies. NEW J CHEM 2021. [DOI: 10.1039/d1nj01004b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cytotoxicity in cancer cells with structure activity relationship has been explored in Cu(ii) compounds involving biologically relevant cooperative assemblies and fascinating crown-like nitrate–water hosts with guest complex cations.
Collapse
Affiliation(s)
- Pranay Sharma
- Department of Chemistry
- Cotton University
- Guwahati-781001
- India
| | - Hiren Nath
- Department of Chemistry
- Cotton University
- Guwahati-781001
- India
| | - Antonio Frontera
- Departament de Química
- Universitat de les Illes Balears
- 07122 Palma de Mallorca (Baleares)
- Spain
| | - Miquel Barcelo-Oliver
- Departament de Química
- Universitat de les Illes Balears
- 07122 Palma de Mallorca (Baleares)
- Spain
| | - Akalesh K. Verma
- Department of Zoology
- Cell & Biochemical Technology Laboratory
- Cotton University
- Guwahati-781001
- India
| | - Sahid Hussain
- Department of Chemistry
- Indian Institute of Technology Patna, Bihta
- Patna-801103
- India
| | | |
Collapse
|
10
|
Stevenson BW, Gorman MA, Koach J, Cheung BB, Marshall GM, Parker MW, Holien JK. A structural view of PA2G4 isoforms with opposing functions in cancer. J Biol Chem 2020; 295:16100-16112. [PMID: 32952126 DOI: 10.1074/jbc.rev120.014293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/17/2020] [Indexed: 01/04/2023] Open
Abstract
The role of proliferation-associated protein 2G4 (PA2G4), alternatively known as ErbB3-binding protein 1 (EBP1), in cancer has become apparent over the past 20 years. PA2G4 expression levels are correlated with prognosis in a range of human cancers, including neuroblastoma, cervical, brain, breast, prostate, pancreatic, hepatocellular, and other tumors. There are two PA2G4 isoforms, PA2G4-p42 and PA2G4-p48, and although both isoforms of PA2G4 regulate cellular growth and differentiation, these isoforms often have opposing roles depending on the context. Therefore, PA2G4 can function either as a contextual tumor suppressor or as an oncogene, depending on the tissue being studied. However, it is unclear how distinct structural features of the two PA2G4 isoforms translate into different functional outcomes. In this review, we examine published structures to identify important structural and functional components of PA2G4 and consider how they may explain its crucial role in the malignant phenotype. We will highlight the lysine-rich regions, protein-protein interaction sites, and post-translational modifications of the two PA2G4 isoforms and relate these to the functional cellular role of PA2G4. These data will enable a better understanding of the function and structure relationship of the two PA2G4 isoforms and highlight the care that will need to be undertaken for those who wish to conduct isoform-specific structure-based drug design campaigns.
Collapse
Affiliation(s)
| | - Michael A Gorman
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Jessica Koach
- Department of Pediatrics, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA; Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Belamy B Cheung
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia; School of Women's and Children's Health, University of New South Wales, Randwick, New South Wales, Australia
| | - Glenn M Marshall
- School of Women's and Children's Health, University of New South Wales, Randwick, New South Wales, Australia; Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - Michael W Parker
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Jessica K Holien
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Surgery, University of Melbourne, Parkville, Victoria, Australia; School of Science, College of Science, Engineering, and Health, RMIT University, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
The roles of multifunctional protein ErbB3 binding protein 1 (EBP1) isoforms from development to disease. Exp Mol Med 2020; 52:1039-1047. [PMID: 32719408 PMCID: PMC8080562 DOI: 10.1038/s12276-020-0476-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
The roles of the two isoforms of ErbB3-binding protein 1 (Ebp1) in cellular function and its regulation in disease and development is a stimulating area in current fields of biology, such as neuroscience, cancer biology, and structural biology. Over the last two decades, a growing body of studies suggests have suggested different functions for the EBP1 isoforms in various cancers, along with their specific binding partners in the ubiquitin-proteasome system. Owing to the specific cellular context or spatial/temporal expression of the EBP1 isoforms, either transcriptional repression or the activation function of EBP1 has been proposed, and epigenetic regulation by p48 EBP1 has also been observed during in the embryo development, including in brain development and neurologic disorders, such as schizophrenia, in using an Ebp1 knockout mouse model. Here, we review recent findings that have shaped our current understanding of the emerging function of EBP1 isoforms in cellular events and gene expression, from development to disease. A pair of proteins that originate from a common gene exert strikingly different effects on embryonic development as well as tumor growth and progression. RNA transcripts generated from the PA2G4 gene can undergo enzymatic processing to yield two different protein products, p42 EB1 and p48 EB1. These proteins differ by the presence or absence of 54 amino acids at one end, and Jee-Yin Ahn at the Sungkyunkwan University School of Medicine, Suwon, South Korea, and colleagues have reviewed current insights into the functional consequences of this difference. The two proteins bind to distinct sets of molecular partners. The p48 form appears to regulate a host of genes involved in brain development, but also appears to drive cancerous growth in various tumors. In contrast, p42 is scarcer during development, and appears to inhibit tumor formation.
Collapse
|
12
|
Dysregulation of Epigenetic Control Contributes to Schizophrenia-Like Behavior in Ebp1 +/- Mice. Int J Mol Sci 2020; 21:ijms21072609. [PMID: 32283721 PMCID: PMC7178112 DOI: 10.3390/ijms21072609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 11/17/2022] Open
Abstract
Dysregulation of epigenetic machinery can cause a variety of neurological disorders associated with cognitive abnormalities. In the hippocampus of postmortem Schizophrenia (SZ) patients, the most notable finding is the deregulation of GAD67 along with differential regulation of epigenetic factors associated with glutamate decarboxylase 67 (GAD67) expression. As we previously reported, ErbB3-binding protein 1 (EBP1) is a potent epigenetic regulator. EBP1 can induce repression of Dnmt1, a well-studied transcriptional repressor of GAD67. In this study, we investigated whether EBP1 contributes to the regulation of GAD67 expression in the hippocampus, controlling epigenetic machinery. In accordance with SZ-like behaviors in Ebp1(+/−) mice, heterozygous deletion of EBP1 led to a dramatic reduction of GAD67 expression, reflecting an abnormally high level of Dnmt1. Moreover, we found that EBP1 binds to the promoter region of HDAC1, which leads to histone deacetylation of GAD67, and suppresses histone deacetylase 1 (HDAC1) expression, inversely mirroring an unusually high level of HDAC1 in Ebp1(+/−) mice. However, EBP1 mutant (p.Glu 183 Ter) found in SZ patients did not elevate the expression of GAD67, failing to suppress Dnmt1 and/or HDAC1 expression. Therefore, this data supports the hypothesis that a reduced amount of EBP1 may contribute to an etiology of SZ due to a loss of transcriptional inhibition of epigenetic repressors, leading to a decreased expression of GAD67.
Collapse
|
13
|
Roles of ErbB3-binding protein 1 (EBP1) in embryonic development and gene-silencing control. Proc Natl Acad Sci U S A 2019; 116:24852-24860. [PMID: 31748268 DOI: 10.1073/pnas.1916306116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
ErbB3-binding protein 1 (EBP1) is implicated in diverse cellular functions, including apoptosis, cell proliferation, and differentiation. Here, by generating genetic inactivation of Ebp1 mice, we identified the physiological roles of EBP1 in vivo. Loss of Ebp1 in mice caused aberrant organogenesis, including brain malformation, and death between E13.5 and 15.5 owing to severe hemorrhages, with massive apoptosis and cessation of cell proliferation. Specific ablation of Ebp1 in neurons caused structural abnormalities of brain with neuron loss in [Nestin-Cre; Ebp1 flox/flox ] mice. Notably, global methylation increased with high levels of the gene-silencing unit Suv39H1/DNMT1 in Ebp1-deficient mice. EBP1 repressed the transcription of Dnmt1 by binding to its promoter region and interrupted DNMT1-mediated methylation at its target gene, Survivin promoter region. Reinstatement of EBP1 into embryo brain relived gene repression and rescued neuron death. Our findings uncover an essential role for EBP1 in embryonic development and implicate its function in transcriptional regulation.
Collapse
|
14
|
Koach J, Holien JK, Massudi H, Carter DR, Ciampa OC, Herath M, Lim T, Seneviratne JA, Milazzo G, Murray JE, McCarroll JA, Liu B, Mayoh C, Keenan B, Stevenson BW, Gorman MA, Bell JL, Doughty L, Hüttelmaier S, Oberthuer A, Fischer M, Gifford AJ, Liu T, Zhang X, Zhu S, Gustafson WC, Haber M, Norris MD, Fletcher JI, Perini G, Parker MW, Cheung BB, Marshall GM. Drugging MYCN Oncogenic Signaling through the MYCN-PA2G4 Binding Interface. Cancer Res 2019; 79:5652-5667. [PMID: 31501192 DOI: 10.1158/0008-5472.can-19-1112] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/17/2019] [Accepted: 09/03/2019] [Indexed: 11/16/2022]
Abstract
MYCN is a major driver for the childhood cancer, neuroblastoma, however, there are no inhibitors of this target. Enhanced MYCN protein stability is a key component of MYCN oncogenesis and is maintained by multiple feedforward expression loops involving MYCN transactivation target genes. Here, we reveal the oncogenic role of a novel MYCN target and binding protein, proliferation-associated 2AG4 (PA2G4). Chromatin immunoprecipitation studies demonstrated that MYCN occupies the PA2G4 gene promoter, stimulating transcription. Direct binding of PA2G4 to MYCN protein blocked proteolysis of MYCN and enhanced colony formation in a MYCN-dependent manner. Using molecular modeling, surface plasmon resonance, and mutagenesis studies, we mapped the MYCN-PA2G4 interaction site to a 14 amino acid MYCN sequence and a surface crevice of PA2G4. Competitive chemical inhibition of the MYCN-PA2G4 protein-protein interface had potent inhibitory effects on neuroblastoma tumorigenesis in vivo. Treated tumors showed reduced levels of both MYCN and PA2G4. Our findings demonstrate a critical role for PA2G4 as a cofactor in MYCN-driven neuroblastoma and highlight competitive inhibition of the PA2G4-MYCN protein binding as a novel therapeutic strategy in the disease. SIGNIFICANCE: Competitive chemical inhibition of the PA2G4-MYCN protein interface provides a basis for drug design of small molecules targeting MYC and MYCN-binding partners in malignancies driven by MYC family oncoproteins.
Collapse
Affiliation(s)
- Jessica Koach
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia.,Department of Pediatrics, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Jessica K Holien
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Hassina Massudi
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Daniel R Carter
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia.,School of Women's & Children's Health, UNSW Sydney, Randwick New South Wales, Australia.,School of Biomedical Engineering, University of Technology Sydney, Australia
| | - Olivia C Ciampa
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Mika Herath
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Taylor Lim
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Janith A Seneviratne
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Giorgio Milazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Jayne E Murray
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Joshua A McCarroll
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia.,Australian Centre for NanoMedicine, ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, UNSW, Australia
| | - Bing Liu
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Bryce Keenan
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Brendan W Stevenson
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Michael A Gorman
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Jessica L Bell
- The Section for Molecular Cell Biology, Institute of Molecular Medicine, Martin Luther University of Halle, Halle, Germany
| | - Larissa Doughty
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Stefan Hüttelmaier
- The Section for Molecular Cell Biology, Institute of Molecular Medicine, Martin Luther University of Halle, Halle, Germany
| | - Andre Oberthuer
- Department of Pediatric Oncology and Hematology, Children's Hospital, University of Cologne, Cologne, Germany.,Department of Neonatology and Pediatric Intensive Care Medicine, Children's Hospital, University of Cologne, Cologne, Germany
| | - Matthias Fischer
- Department of Pediatric Oncology and Hematology, Children's Hospital, University of Cologne, Cologne, Germany.,Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Andrew J Gifford
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia.,Department of Anatomical Pathology, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Xiaoling Zhang
- Department of Biochemistry and Molecular Biology, Cancer Center and Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Cancer Center and Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - W Clay Gustafson
- Department of Pediatrics, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Giovanni Perini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Belamy B Cheung
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia. .,School of Women's & Children's Health, UNSW Sydney, Randwick New South Wales, Australia.,School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Glenn M Marshall
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, New South Wales, Australia. .,School of Women's & Children's Health, UNSW Sydney, Randwick New South Wales, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
| |
Collapse
|
15
|
Padrón-Barthe L, Villalba-Orero M, Gómez-Salinero JM, Domínguez F, Román M, Larrasa-Alonso J, Ortiz-Sánchez P, Martínez F, López-Olañeta M, Bonzón-Kulichenko E, Vázquez J, Martí-Gómez C, Santiago DJ, Prados B, Giovinazzo G, Gómez-Gaviro MV, Priori S, Garcia-Pavia P, Lara-Pezzi E. Severe Cardiac Dysfunction and Death Caused by Arrhythmogenic Right Ventricular Cardiomyopathy Type 5 Are Improved by Inhibition of Glycogen Synthase Kinase-3β. Circulation 2019; 140:1188-1204. [PMID: 31567019 PMCID: PMC6784777 DOI: 10.1161/circulationaha.119.040366] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy/arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited cardiac disease characterized by fibrofatty replacement of the myocardium, resulting in heart failure and sudden cardiac death. The most aggressive arrhythmogenic cardiomyopathy/ARVC subtype is ARVC type 5 (ARVC5), caused by a p.S358L mutation in TMEM43 (transmembrane protein 43). The function and localization of TMEM43 are unknown, as is the mechanism by which the p.S358L mutation causes the disease. Here, we report the characterization of the first transgenic mouse model of ARVC5. METHODS We generated transgenic mice overexpressing TMEM43 in either its wild-type or p.S358L mutant (TMEM43-S358L) form in postnatal cardiomyocytes under the control of the α-myosin heavy chain promoter. RESULTS We found that mice expressing TMEM43-S358L recapitulate the human disease and die at a young age. Mutant TMEM43 causes cardiomyocyte death and severe fibrofatty replacement. We also demonstrate that TMEM43 localizes at the nuclear membrane and interacts with emerin and β-actin. TMEM43-S358L shows partial delocalization to the cytoplasm, reduced interaction with emerin and β-actin, and activation of glycogen synthase kinase-3β (GSK3β). Furthermore, we show that targeting cardiac fibrosis has no beneficial effect, whereas overexpression of the calcineurin splice variant calcineurin Aβ1 results in GSK3β inhibition and improved cardiac function and survival. Similarly, treatment of TMEM43 mutant mice with a GSK3β inhibitor improves cardiac function. Finally, human induced pluripotent stem cells bearing the p.S358L mutation also showed contractile dysfunction that was partially restored after GSK3β inhibition. CONCLUSIONS Our data provide evidence that TMEM43-S358L leads to sustained cardiomyocyte death and fibrofatty replacement. Overexpression of calcineurin Aβ1 in TMEM43 mutant mice or chemical GSK3β inhibition improves cardiac function and increases mice life span. Our results pave the way toward new therapeutic approaches for ARVC5.
Collapse
Affiliation(s)
- Laura Padrón-Barthe
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - María Villalba-Orero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Jesús M Gómez-Salinero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Fernando Domínguez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.)
| | - Marta Román
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.)
| | - Javier Larrasa-Alonso
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Paula Ortiz-Sánchez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Fernando Martínez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Marina López-Olañeta
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Elena Bonzón-Kulichenko
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Carlos Martí-Gómez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.)
| | - Demetrio J Santiago
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Belén Prados
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - Giovanna Giovinazzo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.)
| | - María Victoria Gómez-Gaviro
- Departamento de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain (M.V.G.-G.).,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain (M.V.G.-G.)
| | - Silvia Priori
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.).,Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy (S.P.)
| | - Pablo Garcia-Pavia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (L.P.-B., F.D., M.R., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,ERN GUARD-HEART (European Reference Network for Rare and Complex Diseases of the Heart) (F.D., S.P., P.G.-P.).,Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain (P.G.-P.).,Faculty of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain (P.G.-P.)
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (L.P.-B., M.V.-O., J.M.G.-S., F.D., J.L.-A., P.O.-S., F.M., M.L.-O., E.B.-K., J.V., C.M.-G., D.J.S., B.P., G.G., S.P., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (L.P.-B., F.D., E.B.-K., J.V., C.M.-G., P.G.-P., E.L.-P.).,Faculty of Medicine, National Heart & Lung Institute, Imperial College London, UK (E.L.-P.)
| |
Collapse
|
16
|
Buettner R, Nguyen LXT, Kumar B, Morales C, Liu C, Chen LS, Pemovska T, Synold TW, Palmer J, Thompson R, Li L, Hoang DH, Zhang B, Ghoda L, Kowolik C, Kontro M, Leitch C, Wennerberg K, Xu X, Chen CC, Horne D, Gandhi V, Pullarkat V, Marcucci G, Rosen ST. 8-chloro-adenosine activity in FLT3-ITD acute myeloid leukemia. J Cell Physiol 2019; 234:16295-16303. [PMID: 30770553 PMCID: PMC6697246 DOI: 10.1002/jcp.28294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 01/25/2023]
Abstract
Nucleoside analogs represent the backbone of several distinct chemotherapy regimens for acute myeloid leukemia (AML) and combination with tyrosine kinase inhibitors has improved survival of AML patients, including those harboring the poor-risk FLT3-ITD mutation. Although these compounds are effective in killing proliferating blasts, they lack activity against quiescent leukemia stem cells (LSCs), which contributes to initial treatment refractoriness or subsequent disease relapse. The reagent 8-chloro-adenosine (8-Cl-Ado) is a ribose-containing, RNA-directed nucleoside analog that is incorporated into newly transcribed RNA rather than in DNA, causing inhibition of RNA transcription. In this report, we demonstrate antileukemic activities of 8-Cl-Ado in vitro and in vivo and provide mechanistic insight into the mode of action of 8-Cl-Ado in AML. 8-Cl-Ado markedly induced apoptosis in LSC, with negligible effects on normal stem cells. 8-Cl-Ado was particularly effective against AML cell lines and primary AML blast cells harboring the FLT3-ITD mutation. FLT3-ITD is associated with high expression of miR-155. Furthermore, we demonstrate that 8-Cl-Ado inhibits miR-155 expression levels accompanied by induction of DNA-damage and suppression of cell proliferation, through regulation of miR-155/ErbB3 binding protein 1(Ebp1)/p53/PCNA signaling. Finally, we determined that combined treatment of NSG mice engrafted with FLT3-ITD + MV4-11 AML cells with 8-Cl-Ado and the FLT3 inhibitor AC220 (quizartinib) synergistically enhanced survival, compared with that of mice treated with the individual drugs, suggesting a potentially effective approach for FLT3-ITD AML patients.
Collapse
Affiliation(s)
- Ralf Buettner
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Le Xuan Truong Nguyen
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Vietnam
| | - Bijender Kumar
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Corey Morales
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Chao Liu
- Department of Cancer Genetics and Epigenetics, City of Hope National Medical Center, Duarte, CA
| | - Lisa S. Chen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tea Pemovska
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Timothy W. Synold
- Department of Cancer Biology, City of Hope National Medical Center, Duarte, CA
| | - Joycelynne Palmer
- Department of Information Sciences, City of Hope National Medical Center, Duarte, CA
| | - Ryan Thompson
- Chicago Medical School, Rosalind Franklin University, North Chicago, IL
| | - Ling Li
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Dinh Hoa Hoang
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Bin Zhang
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Lucy Ghoda
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Claudia Kowolik
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Mika Kontro
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Calum Leitch
- Center for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Xiaochun Xu
- Department of Cancer Genetics and Epigenetics, City of Hope National Medical Center, Duarte, CA
| | - Ching-Cheng Chen
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - David Horne
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vinod Pullarkat
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| | - Steven T. Rosen
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
17
|
Yagubova S, Zhanataev A, Ostrovskaya R, Anisina Е, Gudasheva Т, Durnev А, Seredenin S. Dimeric NGF Mimetic Attenuates Hyperglycaemia and DNA Damage in Mice with Streptozotocin-Induced Early-Stage Diabetes. Endocr Metab Immune Disord Drug Targets 2019; 20:453-463. [PMID: 31385776 DOI: 10.2174/1871530319666190806115623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/13/2019] [Accepted: 06/26/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND NGF deficiency is one of the reasons for reduced β-cells survival in diabetes. Our previous experiments revealed the ability of low-weight NGF mimetic, GK-2, to reduce hyperglycaemia in a model of advanced diabetes. The increase in DNA damage in advanced diabetes was repeatedly reported, while there were no data about DNA damage in the initial diabetes. AIM The study aimed to establish whether DNA damage occurs in initial diabetes and whether GK-2 is able to overcome the damage. METHODS The early-stage diabetes was modelled in Balb/c mice by streptozotocin (STZ) (130 mg/kg, i.p.). GK-2 was administered at a dose of 0.5 mg/kg, i.p., subchronically. The evaluation of DNA damage was performed using the alkaline comet assay; the percentage of DNA in the tail (%TDNA) and the percentage of the atypical DNA comets ("ghost cells") were determined. RESULTS STZ at this subthreshold dose produced a slight increase in glycemia and MDA. Meanwhile, pronounced DNA damage was observed, concerning mostly the percentage of "ghost cells" in the pancreas, the liver and kidneys. GK-2 attenuated the degree of hyperglycaemia and reduced the % of "ghost cells" and %TDNA in all the organs examined; this effect continued after discontinuation of the therapy. CONCLUSION Early-stage diabetes is accompanied by DNA damage, manifested by the increase of "ghost cells" percentage. The severity of these changes significantly exceeds the degree of hyperglycaemia and MDA accumulation. GK-2 exerts an antihyperglycaemic effect and attenuates the degree of DNA damage. Our results indicate that the comet assay is a highly informative method for search of antidiabetic medicines.
Collapse
Affiliation(s)
- Svetlana Yagubova
- Laboratory of Psychopharmacology, FSBI "Zakusov Institute of Pharmacology", Moscow, Russian Federation
| | - Aliy Zhanataev
- Laboratory of pharmacology and mutagenesis, FSBI "Zakusov Institute of Pharmacology", Moscow, Russian Federation
| | - Rita Ostrovskaya
- Laboratory of Psychopharmacology, FSBI "Zakusov Institute of Pharmacology", Moscow, Russian Federation
| | - Еlena Anisina
- Laboratory of pharmacology and mutagenesis, FSBI "Zakusov Institute of Pharmacology", Moscow, Russian Federation
| | - Тatiana Gudasheva
- Department of Medicinal Chemistry, FSBI "Zakusov Institute of Pharmacology", Moscow, Russian Federation
| | - Аndrey Durnev
- Laboratory of Drug Toxicology, FSBI "Zakusov Institute of Pharmacology", Moscow, Russian Federation
| | - Sergey Seredenin
- Department of Pharmacogenetics, FSBI "Zakusov Institute of Pharmacology", Moscow, Russian Federation
| |
Collapse
|
18
|
Wu JY, Shih YL, Lin SP, Hsieh TY, Lin YW. YC-1 Antagonizes Wnt/β-Catenin Signaling Through the EBP1 p42 Isoform in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11050661. [PMID: 31086087 PMCID: PMC6562864 DOI: 10.3390/cancers11050661] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 01/03/2023] Open
Abstract
Novel drugs targeting Wnt signaling are gradually being developed for hepatocellular carcinoma (HCC) treatment. In this study, we used a Wnt-responsive Super-TOPflash (STF) luciferase reporter assay to screen a new compound targeting Wnt signaling. 3-(5'-Hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1) was identified as a small molecule inhibitor of the Wnt/β-catenin pathway. Our coimmunoprecipitation (co-IP) data showed that YC-1 did not affect the β-catenin/TCF interaction. Then, by mass spectrometry, we identified the ErbB3 receptor-binding protein 1 (EBP1) interaction with the β-catenin/TCF complex upon YC-1 treatment. EBP1 encodes two splice isoforms, p42 and p48. We further demonstrated that YC-1 enhances p42 isoform binding to the β-catenin/TCF complex and reduces the transcriptional activity of the complex. The suppression of colony formation by YC-1 was significantly reversed after knockdown of both isoforms (p48 and p42); however, the inhibition of colony formation was maintained when only EBP1 p48 was silenced. Taken together, these results suggest that YC-1 treatment results in a reduction in Wnt-regulated transcription through EBP1 p42 and leads to the inhibition of tumor cell proliferation. These data imply that YC-1 is a drug that antagonizes Wnt/β-catenin signaling in HCC.
Collapse
Affiliation(s)
- Ju-Yun Wu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Yu-Lueng Shih
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan.
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Shih-Ping Lin
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Tsai-Yuan Hsieh
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Ya-Wen Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan.
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan.
| |
Collapse
|
19
|
Nguyen DQ, Hoang DH, Nguyen TTV, Ho HD, Huynh V, Shin JH, Ly QT, Thi Nguyen DD, Ghoda L, Marcucci G, Nguyen LXT. Ebp1 p48 promotes oncogenic activities in human colon cancer cells through regulation of TIF-90-mediated ribosomal RNA synthesis. J Cell Physiol 2019; 234:17612-17621. [PMID: 30793766 DOI: 10.1002/jcp.28385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 12/21/2022]
Abstract
The ErbB3-binding protein 1 (Ebp1) has been reported as either an oncogenic regulator or a tumor suppressor in a variety of cancers. Here, we show that Ebp1 p48, a predominant expression isoform, is highly expressed in the majority of human colon tumor cells compared with normal adjacent tissues and its expression is required for the oncogenic activities of these cells. Depletion of Ebp1 expression in primary colon cancer cells inhibits cell proliferation, colony forming, and invasion in vitro as well as tumor formation in vivo and enhances cell sensitivity to irradiation. We further demonstrated that Ebp1 interacts with TIF-90, a splice variant of transcription initiation factor IA (TIF-IA) of the RNA polymerase I complex, allowing for regulation of ribosomal RNA (rRNA) synthesis and oncogenesis in human colon cancer cells. Moreover, Ebp1 expression is essential for Akt protected TIF-90 stability by preventing TIF-90's ubiquitination by Mdm2 and hence, its proteasomal degradation. The results of the present study support a mechanism of underlying oncogenic activities by means of Ebp1 through regulation of TIF-90-mediated rRNA synthesis and suggest the potential therapeutic treatment of colon cancer by targeting Ebp1 and its signaling.
Collapse
Affiliation(s)
- Dang Quan Nguyen
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Dinh Hoa Hoang
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| | - Thanh Thao Vo Nguyen
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Huu Duc Ho
- Department of Gastrointestinal Surgery, Thong Nhat Hospital, Ho Chi Minh City, Vietnam
| | - Vu Huynh
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - June Ho Shin
- Stanford Cancer Institute, Stanford University, Stanford, California
| | - Quoc Trung Ly
- Department of Medicine, Phuong Chau International Hospital, Sóc Trăng, Vietnam
| | | | - Lucy Ghoda
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| | - Le Xuan Truong Nguyen
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| |
Collapse
|
20
|
AKT/protein kinase B associates with β-actin in the nucleus of melanoma cells. Biosci Rep 2019; 39:BSR20181312. [PMID: 30643008 PMCID: PMC6356016 DOI: 10.1042/bsr20181312] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/21/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022] Open
Abstract
The serine-threonine kinase AKT/PKB is a critical regulator of various essential cellular processes, and dysregulation of AKT has been implicated in many diseases, including cancer. Despite AKT action is known to function mainly in the cytoplasm, AKT has been reported to translocate to the nucleus. However, very little is known about the mechanism required for the nuclear import of AKT as well as its function in this cellular compartment. In the present study, we characterized the presence of endogenous nuclear AKT in human melanoma cells and addressed the possible role of AKT by exploring its potential association with key interaction nuclear partners. Confocal and Western blot analyses showed that both phosphorylated and non-phosphorylated forms of AKT are present in melanoma cells nuclei. Using mass spectrometry in combination with protein-crosslinking and co-immunoprecipitation, we identified a series of putative protein partners of nuclear AKT, including heterogeneous nuclear ribonucleoprotein (hnRNP), cytoskeleton proteins β-actin, γ-actin, β-actin-like 2 and vimentin. Confocal microscopy and biochemical analyses validated β-actin as a new nuclear AKT-interacting partner. Cofilin and active RNA Polymerase II, two proteins that have been described to interact and work in concert with nuclear actin in transcription regulation, were also found associated with nuclear AKT. Overall, the present study uncovered a yet unrecognized nuclear coupling of AKT and provides insights into the involvement of AKT in the interaction network of nuclear actin.
Collapse
|
21
|
Liu DW, Ma L, Zhang XH, Wang YY. Conditioned taste aversion memory extinction temporally induces insular cortical BDNF release and inhibits neuronal apoptosis. Neuropsychiatr Dis Treat 2019; 15:2403-2414. [PMID: 31933521 PMCID: PMC6709797 DOI: 10.2147/ndt.s215289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/05/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Memory extinction has been reported to be related to psychiatric disorders, such as post-traumatic stress disorder (PTSD). Secretion and synthesis of brain-derived neurotrophic factor (BDNF) have been shown to temporally regulate various memory processes via activation of tropomyosin-related kinase B (TrkB) receptors. However, whether memory extinction induces the synthesis and secretion of BDNF on the basis of its localization is not understood. In this study, we aim to investigate activity-dependent BDNF secretion and synthesis in the insular cortex (IC) in the setting of conditioned taste aversion (CTA) memory extinction. MATERIALS AND METHODS Rats were subjected to CTA memory extinction and BDNF antibody (or the equal volume of vehicle) was microinjected into the IC immediately after the extinction testing. Real-time polymerase chain reaction and in situ hybridization were used to detect the gene expression of BDNF, NGF and NT4. The protein levels of BDNF were determined through the enzyme-linked immunosorbent assay. In addition, the levels of phosphorylated TrkB normalized to total TrkB were evaluated using immunoprecipitation and immunoblotting. c-Fos, total extracellular signal-regulated kinase (Erk), phosphorylated Erk, and apoptosis-related protein (caspase-3), were detected by Western blotting. RESULTS We found that blocking BDNF signaling within the IC disrupts CTA extinction, suggesting that BDNF signaling in the IC is necessary for CTA extinction. Increased expression levels of c-Fos indicate the induced neuronal activity in the IC during CTA extinction. In addition, temporal changes in the gene expression and protein levels of BDNF in the IC were noted during extinction. Moreover, we found that phosphorylation of TrkB increased prior to the enhanced BDNF expression, suggesting that CTA extinction induces rapid activity-dependent BDNF secretion in the IC. Finally, we found decreased expression of caspase-3 in the IC after CTA extinction. CONCLUSION These results demonstrate that CTA memory extinction temporally induces the release and synthesis of BDNF in the IC and inhibits neuronal apoptosis.
Collapse
Affiliation(s)
- Dian-Wei Liu
- Department of Neurosurgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Ling Ma
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China
| | - Xu-Hua Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, People's Republic of China
| | - Yun-Yan Wang
- Department of Neurosurgery, QiLu Hospital of Shandong University, Jinan, Shandong 250012, People's Republic of China
| |
Collapse
|
22
|
Nguyen DQ, Hoang DH, Nguyen Vo TT, Huynh V, Ghoda L, Marcucci G, Nguyen LXT. The role of ErbB3 binding protein 1 in cancer: Friend or foe? J Cell Physiol 2018; 233:9110-9120. [PMID: 30076717 DOI: 10.1002/jcp.26951] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 06/12/2018] [Indexed: 12/20/2022]
Abstract
ErbB3, a member of the epidermal growth factor receptor family, reportedly plays an essential role in the regulation of cancer progression and therapeutic resistance. Numerous studies have indicated that ErbB3 binding protein 1 (Ebp1), a binding partner for ErbB3, plays an important regulatory role in the expression and function of ErbB3, but there is no agreement as to whether Ebp1 also has an ErbB3-independent function in cancer and how it might contribute to tumorigenesis. In this review, we will discuss the different functions of the two Ebp1 isoforms, p48 and p42, that may be responsible for the potentially dual role of Ebp1 in cancer growth.
Collapse
Affiliation(s)
- Dang Quan Nguyen
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Dinh Hoa Hoang
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| | - Thanh Thao Nguyen Vo
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Vu Huynh
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Lucy Ghoda
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| | - Le Xuan Truong Nguyen
- Department of Medical Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam.,Gehr Family Center for Leukemia Research, Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope Medical Center, Duarte, California
| |
Collapse
|
23
|
Zhang P, Yang S, Cao S, Li B. ErbB3-binding protein 1 inhibits tumor proliferation in esophageal cancer by targeting bcl-2 and p53 gene. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10205-10211. [PMID: 31966354 PMCID: PMC6965776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/18/2017] [Indexed: 06/10/2023]
Abstract
Esophageal cnacer is one of the common malignant tumors with high fatality rate and morbidity, ErbB3-binding protein 1 (EBP1) is the intracellular binding protein of ErbB3. This study aims to investigate the effect of EBP1 on esophageal cancer biological behavior and the underlying mechanism. In our study, esophageal cancer tissues and adjacent non-tumor issues were collected and the relative expression of EBP1 mRNA were examined by real-time PCR. EBP1 mimics is synthesised and transfected onto Eca109 cells. The effects of EBP1 on biological behaviors of Eca109 cells were detected by colony formation assay and flow cytometry, the expression of bcl, p53 proteins were detected by western-blot. The results showed that EBP1 was down-regulation in NSCLC. The number of colony-forming units, and the tumor size and volume in vivo in the Eca109/EBP1 group was obviously lower and the apoptosis rate was significantly higher than the Eca109/NC group and Normal group (P<0.05). The expression of p53 protein in Eca109/EBP1 group were significantly lower, and the expression of bcl-2 were dramatically increased compare with Eca109/NC group and Normal group (P<0.05). Therefore, we concluded that that E-cad may inhibit cell proliferation of esophageal cancer in vitro and in vivo by inducing apoptosis. EBP1 may play a anti-oncogene role in esophageal cancer.
Collapse
Affiliation(s)
- Peiliang Zhang
- Institute of Radiotherapy, Linyi Central HospitalShandong Province, China
| | - Shanlan Yang
- Institute of Radiotherapy, Linyi Central HospitalShandong Province, China
| | - Shuren Cao
- Institute of Radiotherapy, Linyi Central HospitalShandong Province, China
| | - Baosheng Li
- Department of Radiotherapy, Shandong Tumor HospitalShandong Province, China
| |
Collapse
|
24
|
Ko HR, Hwang I, Ahn SY, Chang YS, Park WS, Ahn JY. Neuron-specific expression of p48 Ebp1 during murine brain development and its contribution to CNS axon regeneration. BMB Rep 2017; 50:126-131. [PMID: 27916024 PMCID: PMC5422024 DOI: 10.5483/bmbrep.2017.50.3.190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Indexed: 01/06/2023] Open
Abstract
P48 Ebp1 is expressed in rapidly proliferating cells such as cancer cells and accelerates cell growth and survival. However, its expression pattern and role in central nervous system development have not been studied. Here, we demonstrated the spatiotemporal expression pattern of p48 Ebp1 during embryonic development and the postnatal period. During embryonic development, p48 Ebp1 was highly expressed in the brain. Expression gradually decreased after birth but was still more abundant than p42 expression after birth. Strikingly, we found that p48 Ebp1 was expressed in a cell type specific manner in neurons but not astrocytes. Moreover, p48 Ebp1 physically interacted with beta tubulin but not alpha tubulin. This fits with its accumulation in distal microtubule growth cone regions. Furthermore, in injured hippocampal slices, p48 Ebp1 introduction promoted axon regeneration. Thus, we speculate that p48 Ebp1 might contribute to microtubule dynamics acting as an MAP and promotes CNS axon regeneration. [BMB Reports 2017; 50(3): 126-131].
Collapse
Affiliation(s)
- Hyo Rim Ko
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Inwoo Hwang
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| |
Collapse
|
25
|
Wang Y, Zhang P, Wang Y, Zhan P, Liu C, Mao JH, Wei G. Distinct Interactions of EBP1 Isoforms with FBXW7 Elicits Different Functions in Cancer. Cancer Res 2017; 77:1983-1996. [PMID: 28209614 DOI: 10.1158/0008-5472.can-16-2246] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 01/09/2017] [Accepted: 01/25/2017] [Indexed: 01/07/2023]
Abstract
The ErbB3 receptor-binding protein EBP1 encodes two alternatively spliced isoforms P48 and P42. While there is evidence of differential roles for these isoforms in tumorigenesis, little is known about their underlying mechanisms. Here, we demonstrate that EBP1 isoforms interact with the SCF-type ubiquitin ligase FBXW7 in distinct ways to exert opposing roles in tumorigenesis. EBP1 P48 bound to the WD domain of FBXW7 as an oncogenic substrate of FBXW7. EBP1 P48 binding sequestered FBXW7α to the cytosol, modulating its role in protein degradation and attenuating its tumor suppressor function. In contrast, EBP1 P42 bound to both the F-box domain of FBXW7 as well as FBXW7 substrates. This adapter function of EBP1 P42 stabilized the interaction of FBXW7 with its substrates and promoted FBXW7-mediated degradation of oncogenic targets, enhancing its overall tumor-suppressing function. Overall, our results establish distinct physical and functional interactions between FBXW7 and EBP1 isoforms, which yield their mechanistically unique isoform-specific functions of EBP1 in cancer. Cancer Res; 77(8); 1983-96. ©2017 AACR.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Human Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Shandong, PR China
| | - Pengju Zhang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Shandong, PR China
| | - Yunshan Wang
- Department of Human Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Shandong, PR China.,Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Panpan Zhan
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Shandong, PR China
| | - Chunyan Liu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Shandong, PR China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California.
| | - Guangwei Wei
- Department of Human Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Shandong, PR China.
| |
Collapse
|
26
|
Qian Q, Liu Q, Zhou D, Pan H, Liu Z, He F, Ji S, Wang D, Bao W, Liu X, Liu Z, Zhang H, Zhang X, Zhang L, Wang M, Xu Y, Huang F, Luo B, Sun B. Brain-specific ablation of Efr3a promotes adult hippocampal neurogenesis via the brain-derived neurotrophic factor pathway. FASEB J 2017; 31:2104-2113. [PMID: 28193719 DOI: 10.1096/fj.201601207r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/17/2017] [Indexed: 11/11/2022]
Abstract
Efr3 is a newly identified plasma membrane protein and plays an important role in the phosphoinositide metabolism on the plasma membrane. However, although it is highly expressed in the brain, the functional significance of Efr3 in the brain is not clear. In the present study, we generated Efr3af/f mice and then crossed them with Nestin-Cre mice to delete Efr3a, one of the Efr3 isoforms, specifically in the brain. We found that brain-specific ablation of Efr3a promoted adult hippocampal neurogenesis by increasing survival and maturation of newborn neurons without affecting their dendritic tree morphology. Moreover, the brain-derived neurotrophic factor (BDNF)-tropomyosin-related kinase B (TrkB) signaling pathway was significantly enhanced in the hippocampus of Efr3a-deficient mice, as reflected by increased expression of BDNF, TrkB, and the downstream molecules, including phospho-MAPK and phospho-Akt. Furthermore, the number of TUNEL+ cells was decreased in the subgranular zone of dentate gyrus in Efr3a-deficient mice compared with that of control mice. Our data suggest that brain-specific deletion of Efr3a could promote adult hippocampal neurogenesis, presumably by upregulating the expression of BDNF and its receptor, TrkB, and therefore provide new insight into the roles of Efr3 in the brain.-Qian, Q., Liu, Q., Zhou, D., Pan, H., Liu, Z., He, F., Ji, S., Wang, D., Bao, W., Liu, X., Liu, Z., Zhang, H., Zhang, X., Zhang, L., Wang, M., Xu, Y., Huang, F., Luo, B., Sun B. Brain-specific ablation of Efr3a promotes adult hippocampal neurogenesis via the brain-derived neurotrophic factor pathway.
Collapse
Affiliation(s)
- Qi Qian
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and.,Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiuji Liu
- Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Shanghai, China
| | - Dongming Zhou
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and
| | - Hongyu Pan
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and
| | - Zhiwei Liu
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; and
| | - Fangping He
- Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| | - Suying Ji
- Institute of Neuroscience, Shanghai Institute of Biological Science, Chinese Academy of Sciences, Shanghai, China
| | - Dongpi Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and
| | - Wangxiao Bao
- Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Zhaoling Liu
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; and
| | - Heng Zhang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and
| | - Xiaoqin Zhang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and
| | - Ling Zhang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and
| | - Mingkai Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China.,Key Laboratory of Neurobiology of Zhejiang Province, and
| | - Ying Xu
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; and
| | - Fude Huang
- Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Shanghai, China;
| | - Benyan Luo
- Department of Neurology, First Affiliated Hospital, Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China;
| | - Binggui Sun
- Department of Neurobiology, Key Laboratory of Medical Neurobiology, Ministry of Health of China, .,Key Laboratory of Neurobiology of Zhejiang Province, and
| |
Collapse
|
27
|
Mishra P, Dixit U, Pandey AK, Upadhyay A, Pandey VN. Modulation of HCV replication and translation by ErbB3 binding protein1 isoforms. Virology 2016; 500:35-49. [PMID: 27770702 DOI: 10.1016/j.virol.2016.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/30/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023]
Abstract
We recently identified a cell-factor, ErbB3 binding protein 1 (Ebp-1), which specifically interacts with the viral RNA genome and modulates HCV replication and translation. Ebp1 has two isoforms, p48, and p42, that result from differential splicing. We found that both isoforms interact with HCV proteins NS5A and NS5B, as well as cell-factor PKR. The p48 isoform, which localizes in the cytoplasm and nuclei, promoted HCV replication, whereas the shorter p42 isoform, which resides exclusively in the cytoplasm, strongly inhibited HCV replication. Transient expression of individual isoforms in Ebp1-knockdown MH14 cells confirmed that the p48 isoform promotes HCV replication, while the p42 isoform inhibits it. We found that Ebp1-p42 significantly enhanced autophosphorylation of PKR, while Ebp1-p48 isoform strongly inhibited it. We propose that modulation of autophosphorylation of PKR by p48 isoform is an important mechanism whereby the HCV virus escapes innate antiviral immune responses by circumventing p42-mediated inhibition of its replication.
Collapse
Affiliation(s)
- Priya Mishra
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Updesh Dixit
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Ashutosh K Pandey
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Alok Upadhyay
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Virendra N Pandey
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA.
| |
Collapse
|
28
|
C-terminal domain of p42 Ebp1 is essential for down regulation of p85 subunit of PI3K, inhibiting tumor growth. Sci Rep 2016; 6:30626. [PMID: 27464702 PMCID: PMC4964336 DOI: 10.1038/srep30626] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
Potential tumor suppressor p42, ErbB3-binding protein 1 (EBP1) inhibits phosphoinositide 3-kinase (PI3K) activity reducing the p85 regulatory subunit. In this study, we demonstrated that overexpression of p42 promoted not only a reduction of wild type of p85 subunit but also oncogenic mutant forms of p85 which were identified in human cancers. Moreover, we identified the small fragment of C-terminal domain of p42 is sufficient to exhibit tumor suppressing activity of p42-WT, revealing that this small fragment (280-394) of p42 is required for the binding of both HSP70 and CHIP for a degradation of p85. Furthermore, we showed the small fragment of p42 markedly inhibited the tumor growth in mouse xenograft models of brain and breast cancer, resembling tumor suppressing activity of p42. Through identification of the smallest fragment of p42 that is responsible for its tumor suppressor activity, our findings represent a novel approach for targeted therapy of cancers that overexpress PI3K.
Collapse
|
29
|
Ko HR, Chang YS, Park WS, Ahn JY. Opposing roles of the two isoforms of ErbB3 binding protein 1 in human cancer cells. Int J Cancer 2016; 139:1202-8. [DOI: 10.1002/ijc.30165] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/25/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Hyo Rim Ko
- Department of Molecular Cell Biology; Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine; Suwon Korea
| | - Yun Sil Chang
- Department of Pediatrics; Samsung Medical Center, Sungkyunkwan University School of Medicine; Seoul Korea
| | - Won Soon Park
- Department of Pediatrics; Samsung Medical Center, Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology; Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine; Suwon Korea
| |
Collapse
|
30
|
Akt attenuates apoptotic death through phosphorylation of H2A under hydrogen peroxide-induced oxidative stress in PC12 cells and hippocampal neurons. Sci Rep 2016; 6:21857. [PMID: 26899247 PMCID: PMC4761890 DOI: 10.1038/srep21857] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 02/02/2016] [Indexed: 01/24/2023] Open
Abstract
Although the essential role of protein kinase B (PKB)/Akt in cell survival signaling has been clearly established, the mechanism by which Akt mediates the cellular response to hydrogen peroxide (H2O2)-induced oxidative stress remains unclear. We demonstrated that Akt attenuated neuronal apoptosis through direct association with histone 2A (H2A) and phosphorylation of H2A at threonine 17. At early time points during H2O2 exposure of PC12 cells and primary hippocampal neurons, when the cells can tolerate the level of DNA damage, Akt was activated and phosphorylated H2A, leading to inhibition of apoptotic death. At later time points, Akt delivered the NAD+-dependent protein deacetylase Sirtuin 2 (Sirt 2) to the vicinity of phosphorylated H2A in response to irreversible DNA damage, thereby inducing H2A deacetylation and subsequently leading to apoptotic death. Ectopically expressed T17A-substituted H2A minimally interacted with Akt and failed to prevent apoptosis under oxidative stress. Thus Akt-mediated H2A phosphorylation has an anti-apoptotic function in conditions of H2O2-induced oxidative stress in neurons and PC12 cells.
Collapse
|
31
|
Nguyen LXT, Zhu L, Lee Y, Ta L, Mitchell BS. Expression and Role of the ErbB3-Binding Protein 1 in Acute Myelogenous Leukemic Cells. Clin Cancer Res 2016; 22:3320-7. [PMID: 26813358 DOI: 10.1158/1078-0432.ccr-15-2282] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/21/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE The ErbB3-binding protein 1 (Ebp1) has been implicated in diverse cancers as having either oncogenic or tumor suppressor activities. The present study was undertaken to determine the effects of Ebp1 expression in AML cells and to determine the mechanisms by which Ebp1 promotes cell proliferation in these cells. EXPERIMENTAL DESIGN The expression of Ebp1 was studied in mononuclear cells obtained from the peripheral blood of 54 patients with AML by Western blot analysis. The effects of Ebp1 expression on proliferating cell nuclear antigen (PCNA) expression and cell proliferation was measured using Western blot analysis, immunoprecipitation, in vitro ubiquitination, and colony-forming assays. The role of Ebp1 in promoting rRNA synthesis and cell proliferation was evaluated by measuring the level of pre-rRNA and the recruitment of Pol I to rDNA. RESULTS Ebp1 is highly expressed in acute myelogenous leukemia (AML) cells and regulates the level of ribosomal RNA (rRNA) synthesis by binding to RNA Polymerase I (Pol I) and enhancing the formation of the Pol I initiation complex. Ebp1 also increases the stability of PCNA protein by preventing its interaction with Mdm2, for which it is a substrate. CONCLUSIONS These results demonstrate an important role of Ebp1 in promoting cell proliferation in AML cells through the regulation of both rRNA synthesis and PCNA expression. Clin Cancer Res; 22(13); 3320-7. ©2016 AACR.
Collapse
Affiliation(s)
- Le Xuan Truong Nguyen
- Departments of Medicine and Chemical and Systems Biology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Li Zhu
- Departments of Medicine and Chemical and Systems Biology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Yunqin Lee
- Departments of Medicine and Chemical and Systems Biology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Lynn Ta
- Departments of Medicine and Chemical and Systems Biology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Beverly S Mitchell
- Departments of Medicine and Chemical and Systems Biology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
32
|
Cheng H, Chen X, Zhu J, Huang H. Overexpression of a Hevea brasiliensis ErbB-3 Binding protein 1 Gene Increases Drought Tolerance and Organ Size in Arabidopsis. FRONTIERS IN PLANT SCIENCE 2016; 7:1703. [PMID: 27895658 PMCID: PMC5107689 DOI: 10.3389/fpls.2016.01703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/28/2016] [Indexed: 05/04/2023]
Abstract
Rubber trees are economically important tropical tree species and the major source of natural rubber, which is an essential industrial material. This tropical perennial tree is susceptible to cold stress and other abiotic stresses, especially in the marginal northern tropics. Recent years, the genome sequencing and RNA-seq projects produced huge amount of sequence data, which greatly facilitated the functional genomics study. However, the characterization of individual functional gene is in urgent demands, especially for those involved in stress resistance. Here we identified and characterized the rubber tree gene ErbB-3 binding protein 1, which undergoes changes in expression in response to cold, drought stress and ABA treatment. HbEBP1 overexpression (OE) in Arabidopsis increased organ size, facilitated root growth and increased adult leaf number by delaying the vegetative-to-reproductive transition. In addition, HbEBP1 OE enhanced the resistance of the Arabidopsis plants to freezing and drought stress, demonstrating that this gene participates in the regulation of abiotic stress resistance. RD29a, RD22 and CYCD3;1 expression was also greatly enhanced by HbEBP1 OE, which explains its regulatory roles in organ size and stress resistance. The regulation of drought stress resistance is a novel function identified in plant EBP1 genes, which expands our understanding of the roles of EBP1 gene in response to the environment. Our results provide information that may lead to the use of HbEBP1 in genetically engineered crops to increase both biomass and abiotic stress resistance.
Collapse
Affiliation(s)
- Han Cheng
- *Correspondence: Han Cheng, Huasun Huang,
| | | | | | | |
Collapse
|
33
|
Arora A, Singh S, Bhatt AN, Pandey S, Sandhir R, Dwarakanath BS. Interplay Between Metabolism and Oncogenic Process: Role of microRNAs. TRANSLATIONAL ONCOGENOMICS 2015; 7:11-27. [PMID: 26740741 PMCID: PMC4696840 DOI: 10.4137/tog.s29652] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/11/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022]
Abstract
Cancer is a complex disease that arises from the alterations in the composition and regulation of several genes leading to the disturbances in signaling pathways, resulting in the dysregulation of cell proliferation and death as well as the ability of transformed cells to invade the host tissue and metastasize. It is increasingly becoming clear that metabolic reprograming plays a critical role in tumorigenesis and metastasis. Therefore, targeting this phenotype is considered as a promising approach for the development of therapeutics and adjuvants. The process of metabolic reprograming is linked to the activation of oncogenes and/or suppression of tumor suppressor genes, which are further regulated by microRNAs (miRNAs) that play important roles in the interplay between oncogenic process and metabolic reprograming. Looking at the advances made in the recent past, it appears that the translation of knowledge from research in the areas of metabolism, miRNA, and therapeutic response will lead to paradigm shift in the management of this disease.
Collapse
Affiliation(s)
- Aastha Arora
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.; Department of Biochemistry, Panjab University, Chandigarh, India
| | - Saurabh Singh
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Anant Narayan Bhatt
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Sanjay Pandey
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.; Dr B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Bilikere S Dwarakanath
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.; Sri Ramachandra University, Chennai, India
| |
Collapse
|
34
|
Ko HR, Nguyen TL, Kim CK, Park Y, Lee KH, Ahn JY. P42 Ebp1 functions as a tumor suppressor in non-small cell lung cancer. BMB Rep 2015; 48:159-65. [PMID: 24998263 PMCID: PMC4453029 DOI: 10.5483/bmbrep.2015.48.3.130] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Indexed: 12/03/2022] Open
Abstract
Although the short isoform of ErbB3-binding protein 1 (Ebp1), p42 has been considered to be a potent tumor suppressor in a number of human cancers, whether p42 suppresses tumorigenesis of lung cancer cells has never been clarified. In the current study we investigated the tumor suppressor role of p42 in non-small cell lung cancer cells. Our data suggest that the expression level of p42 is inversely correlated with the cancerous properties of NSCLC cells and that ectopic expression of p42 is sufficient to inhibit cell proliferation, anchorage-independent growth, and invasion as well as tumor growth in vivo. Interestingly, p42 suppresses Akt activation and overexpression of a constitutively active form of Akt restores the tumorigenic activity of A549 cells that is ablated by exogenous p42 expression. Thus, we propose that p42 Ebp1 functions as a potent tumor suppressor of NSCLC through interruption of Akt signaling. [BMB Reports 2015; 48(3): 159-165]
Collapse
Affiliation(s)
- Hyo Rim Ko
- Department of Molecular Cell Biology; Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | - Truong Lx Nguyen
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | - Chung Kwon Kim
- Department of Molecular Cell Biology; Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | - Youngbin Park
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kyung-Hoon Lee
- Anatomy; Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology; Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| |
Collapse
|
35
|
ErbB3 binding protein 1 (EBP1) participates in the regulation of intestinal inflammation via mediating Akt signaling pathway. Mol Immunol 2015; 67:540-51. [DOI: 10.1016/j.molimm.2015.07.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 07/24/2015] [Accepted: 07/26/2015] [Indexed: 01/22/2023]
|
36
|
Zhang F, Liu Y, Wang Z, Sun X, Yuan J, Wang T, Tian R, Ji W, Yu M, Zhao Y, Niu R. A novel Anxa2-interacting protein Ebp1 inhibits cancer proliferation and invasion by suppressing Anxa2 protein level. Mol Cell Endocrinol 2015; 411:75-85. [PMID: 25917452 DOI: 10.1016/j.mce.2015.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 03/31/2015] [Accepted: 04/16/2015] [Indexed: 11/16/2022]
Abstract
Anxa2 is dysregulated in many types of carcinomas and implicated in several pivotal biological functions, such as angiogenesis, cell proliferation, invasion, and metastasis. We previously demonstrated that upregulation of Anxa2 enhances the proliferation and invasion of breast cancer cells. However, the detailed mechanism remains unclear. In this study, co-immunoprecipitation and LC-MS/MS-based interactome approach were employed to screen potential Anxa2 binding proteins. A total of 312 proteins were identified as candidate Anxa2 interacting partners. Using Gene Ontology, pathway annotation, and protein-protein interaction analyses, we constructed a connected network for Anxa2 interacting proteins, and Ebp1 may function as a "hub" in the Anxa2 interaction network. Moreover, Ebp1 knockdown resulted in enhanced cell proliferation and invasion, as well as increased expression of Anxa2. Furthermore, the abundance of cyclin D1 and the phosphorylation of Erk1/2 were increased in Ebp1 inhibited cells. This finding is consistent with a previous study, in which upregulation of Anxa2 results in an increased cyclin D1 expression and Erk1/2 activation. Our results suggest a novel function of Ebp1 as a binding protein and negative regulator of Anxa2. The functional association between Anxa2 and EBP1 may also participate in regulating cancer cell proliferation and invasion, thereby contributing to cancer progression.
Collapse
Affiliation(s)
- Fei Zhang
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| | - Yuan Liu
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Zhiyong Wang
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Xiumei Sun
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Jie Yuan
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Tong Wang
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Ran Tian
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Wei Ji
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Man Yu
- Ontario Cancer Institute/Princess Margaret Hospital, University of Toronto, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - Yuanyuan Zhao
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Ruifang Niu
- Public Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| |
Collapse
|
37
|
High expression of ErbB3 binding protein 1 (EBP1) predicts poor prognosis of pancreatic ductal adenocarcinoma (PDAC). Tumour Biol 2015; 36:9189-99. [PMID: 26088450 DOI: 10.1007/s13277-015-3625-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/27/2015] [Indexed: 02/04/2023] Open
Abstract
Recent studies have identified that ErbB3 binding protein 1 (EBP1) is broadly expressed in various cancer tissues and critically involved in plenty of biological processes in this regard. However, the functional role of EBP1 in pancreatic ductal adenocarcinoma (PDAC) has never been elucidated. In this study, we found that EBP1 could serve as a prognostic biomarker of PDAC. Western blot analysis revealed that EBP1 was remarkably upregulated in PDAC tissues and cell lines. Using immunohistochemical analysis, we showed that the expression of EBP1 was correlated with tumor size (P = 0.004), histological differentiation (P = 0.041), and tumor node metastasis (TNM) stage (P = 0.000). Notably, Kaplan-Meier curve showed that high expression of EBP1 predicted significantly worsened prognosis of PDAC patients (P = 0.001). In addition, knockdown of EBP1 expression suppressed PDAC cell proliferation and retarded cell cycle progression. Furthermore, depletion of EBP1 induced the apoptosis of Panc-1 cells. Of great interest, we found that EBP1 interacted with anti-apoptotic protein, Bcl-xL, and promoted its accumulation. In summary, our results suggest that EBP1 is a novel prognostic indicator and potential therapeutic target of PDAC, shedding new insights into the important role of EBP1 in cancer development.
Collapse
|
38
|
Nguyen LXT, Lee Y, Urbani L, Utz PJ, Hamburger AW, Sunwoo JB, Mitchell BS. Regulation of ribosomal RNA synthesis in T cells: requirement for GTP and Ebp1. Blood 2015; 125:2519-29. [PMID: 25691158 PMCID: PMC4400289 DOI: 10.1182/blood-2014-12-616433] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/04/2015] [Indexed: 11/20/2022] Open
Abstract
Mycophenolic acid (MPA) is the active metabolite of mycophenolate mofetil, an effective immunosuppressive drug. Both MPA and mycophenolate mofetil are highly specific inhibitors of guanine nucleotide synthesis and of T-cell activation. However, the mechanism by which guanine nucleotide depletion suppresses T-cell activation is unknown. Depletion of GTP inhibits ribosomal RNA synthesis in T cells by inhibiting transcription initiation factor I (TIF-IA), a GTP-binding protein that recruits RNA polymerase I to the ribosomal DNA promoter. TIF-IA-GTP binds the ErbB3-binding protein 1, and together they enhance the transcription of proliferating cell nuclear antigen (PCNA). GTP binding by TIF-IA and ErbB3-binding protein 1 phosphorylation by protein kinase C δ are both required for optimal PCNA expression. The protein kinase C inhibitor sotrastaurin markedly potentiates the inhibition of ribosomal RNA synthesis, PCNA expression, and T-cell activation induced by MPA, suggesting that the combination of the two agents are more highly effective than either alone in inducing immunosuppression.
Collapse
Affiliation(s)
| | - Yunqin Lee
- Department of Otolaryngology (Head and Neck Surgery), Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Lenore Urbani
- Departments of Medicine and Chemical and Systems Biology, and
| | - Paul J Utz
- Division of Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford CA; and
| | - Anne W Hamburger
- Department of Pathology and Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - John B Sunwoo
- Department of Otolaryngology (Head and Neck Surgery), Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | |
Collapse
|
39
|
Kumaraswamy P, Sethuraman S, Krishnan UM. Tjernberg peptide: a double edged sword in Alzheimer’s disease. RSC Adv 2015. [DOI: 10.1039/c5ra10746f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
KLVFF peptide acts as an anti-oxidant through regulation of SOD levels by altering the levels of AP-1 and NF-κB below a concentration of 100 μM while at higher concentrations it turns toxic through activation of MAP kinases and p53.
Collapse
Affiliation(s)
- Priyadharshini Kumaraswamy
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB)
- School of Chemical & Biotechnology
- SASTRA University
- Thanjavur-613 401
- India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB)
- School of Chemical & Biotechnology
- SASTRA University
- Thanjavur-613 401
- India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB)
- School of Chemical & Biotechnology
- SASTRA University
- Thanjavur-613 401
- India
| |
Collapse
|
40
|
Ahn JY. Neuroprotection signaling of nuclear akt in neuronal cells. Exp Neurobiol 2014; 23:200-6. [PMID: 25258566 PMCID: PMC4174610 DOI: 10.5607/en.2014.23.3.200] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/11/2014] [Accepted: 08/12/2014] [Indexed: 12/29/2022] Open
Abstract
Akt is one of the central kinases that perform a pivotal function in mediating survival signaling in a wide range of neuronal cell types in response to growth factor stimulation. The recent findings of a number of targets for Akt suggest that it prohibits neuronal death by both impinging on the cytoplasmic cell death machinery and by regulating nuclear proteins. The presence of active Akt in the nuclei of mammalian cells is no longer debatable, and this has been corroborated by the finding of multiple targets in the nucleus of PC12 cells. However, it is also clear that the nuclear Akt signaling exists independent of the cytosolic Akt signaling, thereby showing a distinctive feature of nuclear Akt signaling as opposed to its cytosolic counterpart. The principal objective of this review is to summarize our current state of knowledge regarding nuclear Akt signaling in neuronal survival, and to introduce current theories regarding the roles of nuclear Akt in neuron.
Collapse
Affiliation(s)
- Jee-Yin Ahn
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| |
Collapse
|
41
|
Ko HR, Kim CK, Ahn JY. Phosphorylation of the N-terminal domain of p48 Ebp1 by CDK2 is required for tumorigenic function of p48. Mol Carcinog 2014; 54:1283-91. [DOI: 10.1002/mc.22203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 06/07/2014] [Accepted: 06/11/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Hyo Rim Ko
- Department of Molecular Cell Biology; Center for Molecular Medicine; Samsung Biomedical Research Institute; Sungkyunkwan University School of Medicine; Suwon Korea
| | - Chung Kwon Kim
- Department of Molecular Cell Biology; Center for Molecular Medicine; Samsung Biomedical Research Institute; Sungkyunkwan University School of Medicine; Suwon Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology; Center for Molecular Medicine; Samsung Biomedical Research Institute; Sungkyunkwan University School of Medicine; Suwon Korea
| |
Collapse
|
42
|
Deoxyribonuclease inhibitors. Eur J Med Chem 2014; 88:101-11. [PMID: 25042005 DOI: 10.1016/j.ejmech.2014.07.040] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 02/06/2023]
Abstract
Deoxyribonucleases (DNases) are a class of enzymes able to catalyze DNA hydrolysis. DNases play important roles in cell function, while DNase inhibitors control or modify their activities. This review focuses on DNase inhibitors. Some DNase inhibitors have been isolated from various natural sources, such as humans, animals (beef, calf, rabbit and rat), plants (Nicotiana tabacum), and microorganisms (some Streptomyces and Adenovirus species, Micromonospora echinospora and Escherichia coli), while others have been obtained by chemical synthesis. They differ in chemical structure (various proteins, nucleotides, anthracycline and aminoglycoside antibiotics, synthetic organic and inorganic compounds) and mechanism of action (forming complexes with DNases or DNA). Some of the inhibitors are specific toward only one type of DNase, while others are active towards two or more. Physico-chemical properties of DNase inhibitors are calculated using the Molinspiration tool and most of them meet all criteria for good solubility and permeability. DNase inhibitors may be used as pharmaceuticals for preventing, monitoring and treating various diseases.
Collapse
|
43
|
Ko HR, Kim CK, Lee SB, Song J, Lee KH, Kim KK, Park KW, Cho SW, Ahn JY. P42 Ebp1 regulates the proteasomal degradation of the p85 regulatory subunit of PI3K by recruiting a chaperone-E3 ligase complex HSP70/CHIP. Cell Death Dis 2014; 5:e1131. [PMID: 24651434 PMCID: PMC3973206 DOI: 10.1038/cddis.2014.79] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 01/28/2023]
Abstract
The short isoform of ErbB3-binding protein 1 (Ebp1), p42, is considered to be a potent tumor suppressor in a number of human cancers, although the mechanism by which it exerts this tumor-suppressive activity is unclear. Here, we report that p42 interacts with the cSH2 domain of the p85 subunit of phosphathidyl inositol 3-kinase (PI3K), leading to inhibition of its lipid kinase activity. Importantly, we found that p42 induces protein degradation of the p85 subunit and further identified HSP70/CHIP complex as a novel E3 ligase for p85 that is responsible for p85 ubiquitination and degradation. In this process, p42 couples p85 to the HSP70/CHIP-mediated ubiquitin–proteasomal system (UPS), thereby promoting a reduction of p85 levels both in vitro and in vivo. Thus, the tumor-suppressing effects of p42 in cancer cells are driven by negative regulation of the p85 subunit of PI3K.
Collapse
Affiliation(s)
- H R Ko
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - C K Kim
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - S B Lee
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - J Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - K-H Lee
- Department of Anatomy, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - K K Kim
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - K W Park
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon, Korea
| | - S-W Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan, College of Medicine, Seoul, Korea
| | - J-Y Ahn
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
44
|
The nitric oxide-cGKII system relays death and survival signals during embryonic retinal development via AKT-induced CREB1 activation. Cell Death Differ 2014; 21:915-28. [PMID: 24531539 DOI: 10.1038/cdd.2014.11] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 12/21/2013] [Accepted: 01/08/2014] [Indexed: 01/28/2023] Open
Abstract
During early neurogenesis, retinal neuronal cells display a conserved differentiation program in vertebrates. Previous studies established that nitric oxide (NO) and cGMP accumulation regulate essential events in retinal physiology. Here we used pharmacological and genetic loss-of-function to investigate the effects of NO and its downstream signaling pathway in the survival of developing avian retinal neurons in vitro and in vivo. Six-day-old (E6) chick retinal cells displayed increased calcium influx and produced higher amounts of NO when compared with E8 cells. L-arginine (substrate for NO biosynthesis) and S-nitroso-N-acetyl-D,L-penicillamine (SNAP; a nitrosothiol NO donor) promoted extensive cell death in E6 retinas, whereas in E8 both substances decreased apoptosis. The effect of NO at both periods was mediated by soluble guanylyl cyclase (sGC) and cGMP-dependent kinase (cGK) activation. In addition, shRNA-mediated cGKII knockdown prevented NO-induced cell death (E6) and cell survival (E8). This, NO-induced cell death or cell survival was not correlated with an early inhibition of retinal cell proliferation. E6 cells also responded differentially from E8 neurons regarding cyclic AMP-responsive element-binding protein (CREB) activation in the retina in vivo. NO strongly decreased nuclear phospho-CREB staining in E6 but it robustly enhanced CREB phosphorylation in the nuclei of E8 neurons, an effect that was completely abrogated by cGKII shRNAs at both embryonic stages. The ability of NO in regulating CREB differentially during retinal development relied on the capacity of cGKII in decreasing (E6) or increasing (E8) nuclear AKT (V-Akt murine thymoma viral oncogene) activation. Accordingly, inhibiting AKT prevented both cGKII shRNA-mediated CREB upregulation in E6 and SNAP-induced CREB activation in E8. Furthermore, shRNA-mediated in vivo cGKII or in vitro CREB1 knockdown confirmed that NO/cGKII dualistically regulated the downstream CREB1 pathway and caspase activation in the chick retina to modulate neuronal viability. These data demonstrate that NO-mediated cGKII signaling may function to control the viability of neuronal cells during early retinal development via AKT/CREB1 activity.
Collapse
|
45
|
Norris-Mullins B, VanderKolk K, Vacchina P, Joyce MV, Morales MA. LmaPA2G4, a homolog of human Ebp1, is an essential gene and inhibits cell proliferation in L. major. PLoS Negl Trop Dis 2014; 8:e2646. [PMID: 24421916 PMCID: PMC3888471 DOI: 10.1371/journal.pntd.0002646] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/03/2013] [Indexed: 11/18/2022] Open
Abstract
We have identified LmaPA2G4, a homolog of the human proliferation-associated 2G4 protein (also termed Ebp1), in a phosphoproteomic screening. Multiple sequence alignment and cluster analysis revealed that LmaPA2G4 is a non-peptidase member of the M24 family of metallopeptidases. This pseudoenzyme is structurally related to methionine aminopeptidases. A null mutant system based on negative selection allowed us to demonstrate that LmaPA2G4 is an essential gene in Leishmania major. Over-expression of LmaPA2G4 did not alter cell morphology or the ability to differentiate into metacyclic and amastigote stages. Interestingly, the over-expression affected cell proliferation and virulence in mouse footpad analysis. LmaPA2G4 binds a synthetic double-stranded RNA polyriboinosinic polyribocytidylic acid [poly(I∶C)] as shown in an electrophoretic mobility shift assay (EMSA). Quantitative proteomics revealed that the over-expression of LmaPA2G4 led to accumulation of factors involved in translation initiation and elongation. Significantly, we found a strong reduction of de novo protein biosynthesis in transgenic parasites using a non-radioactive metabolic labeling assay. In conclusion, LmaPA2G4 is an essential gene and is potentially implicated in fundamental biological mechanisms, such as translation, making it an attractive target for therapeutic intervention.
Collapse
Affiliation(s)
- Brianna Norris-Mullins
- Eck Institute for Global Health. Department of Biological Sciences. University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Kaitlin VanderKolk
- Eck Institute for Global Health. Department of Biological Sciences. University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Paola Vacchina
- Eck Institute for Global Health. Department of Biological Sciences. University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Michelle V. Joyce
- Mass Spectrometry and Proteomics Facility, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Miguel A. Morales
- Eck Institute for Global Health. Department of Biological Sciences. University of Notre Dame, Notre Dame, Indiana, United States of America
- * E-mail:
| |
Collapse
|
46
|
Yip PY, Cooper WA, Kohonen-Corish MRJ, Lin BPC, McCaughan BC, Boyer MJ, Kench JG, Horvath LG. Phosphorylated Akt expression is a prognostic marker in early-stage non-small cell lung cancer. J Clin Pathol 2013; 67:333-40. [PMID: 24265323 DOI: 10.1136/jclinpath-2013-201870] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
AIMS To determine the prognostic significance of pAkt expression in order to identify high-risk stage IB patients with non-small cell lung cancer (NSCLC) in an exploratory study. METHODS We identified 471 consecutive patients with stage IB primary NSCLC according to the American Joint Commission on Cancer 6th edition tumour-node-metastasis (TNM) staging system, who underwent surgical resection between 1990 and 2008. Patients who received neoadjuvant or adjuvant treatments were excluded. Pathology reports were reviewed, and pathological characteristics were extracted. Expression of phosphorylated Akt (pAkt) in both cytoplasmic and nuclear locations was assessed by immunohistochemistry, and clinicopathological factors were analysed against 10-year overall survival using Kaplan-Meier and Cox proportional hazards model. RESULTS 455 (96.6%) cancers were adequate for pAkt immunohistochemical analysis. The prevalence of pAkt expression in the cytoplasm and nucleus of the cancers was 60.7% and 43.7%, respectively. Patients whose cancers expressed higher levels of cytoplasmic pAkt had a trend towards longer overall survival than those with lower levels (p=0.06). Conversely, patients whose cancers expressed higher levels of nuclear pAkt had a poorer prognosis than those with lower levels of expression (p=0.02). Combined low cytoplasmic/high nuclear expression of pAkt was an independent predictor of overall survival (HR=2.86 (95% CI 1.35 to 6.04); p=0.006) when modelled with age (HR=1.05 (95% CI 1.03 to 1.07); p<0.001), extent of operation (HR=2.11 (95% CI 1.48 to 3.01); p<0.001), visceral pleural invasion (HR=1.63 (95% CI 1.24 to 2.15); p<0.001), gender, tumour size, histopathological type and grade (p>0.05). CONCLUSIONS Level of expression of pAkt in the cytoplasm and nucleus is an independent prognostic factor that may help to select patients with high-risk disease.
Collapse
Affiliation(s)
- P Y Yip
- Department of Medical Oncology, Sydney Cancer Centre, Royal Prince Alfred Hospital, , Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Expression of ErbB3-binding protein-1 (EBP1) during primordial follicle formation: role of estradiol-17ß. PLoS One 2013; 8:e67068. [PMID: 23840586 PMCID: PMC3688617 DOI: 10.1371/journal.pone.0067068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/14/2013] [Indexed: 01/18/2023] Open
Abstract
The formation of primordial follicles involves the interaction between the oocytes and surrounding somatic cells, which differentiate into granulosa cells. Estradiol-17ß (E) promotes primordial follicle formation in vivo and in vitro; however, the underlying mechanisms are poorly understood. The expression of an ERBB3-binding protein 1 (EBP1) is downregulated in 8-day old hamster ovaries concurrent with the increase in serum estradiol levels and the formation of primordial follicles. The objectives of the present study were to determine the spatio-temporal expression and putative E regulation of EBP1 in ovarian cells during perinatal development with respect to primordial follicle formation. Hamster EBP1 nucleic acid and amino acid sequences were more than 93% and 98% similar, respectively, to those of mouse and human, and contained nucleolar localization signal, RNA-binding domain and several phosphorylation sites. EBP1 protein was present in somatic cells and oocytes from E15, and declined in oocytes by P1 and in somatic cells by P5. Thereafter, EBP1 expression increased through P7 with a transient decline on P8 primarily in interstitial cells. EBP1 mRNA levels mirrored protein expression pattern. E treatment on P1 and P4 upregulated EBP1 expression by P8 whereas E treatment on P4 downregulated it by 72 h suggesting a compensatory upregulation due to E pretreatment. Treatment with an FSH-antiserum, which suppressed primordial follicle formation, prevented the decline in EBP1 levels, and the effect was reversed by E treatment. Therefore, the results provide the first evidence that EBP1 may play an important role in mediating the effect of E in the differentiation of somatic cells into granulosa cells during primordial follicle formation.
Collapse
|
48
|
Uranga RM, Katz S, Salvador GA. Enhanced phosphatidylinositol 3-kinase (PI3K)/Akt signaling has pleiotropic targets in hippocampal neurons exposed to iron-induced oxidative stress. J Biol Chem 2013; 288:19773-84. [PMID: 23687303 DOI: 10.1074/jbc.m113.457622] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The PI3K/Akt pathway is a key component in synaptic plasticity and neuronal survival. The aim of this work was to investigate the participation of the PI3K/Akt pathway and its outcome on different molecular targets such as glycogen synthase kinase 3β (GSK3β) and Forkhead box-O (FoxO) transcription factors during mild oxidative stress triggered by iron overload. The exposure of mouse hippocampal neurons (HT22) to different concentrations of Fe(2+) (25-200 μm) for 24 h led us to define a mild oxidative injury status (50 μm Fe(2+)) in which cell morphology showed changes typical of neuronal damage with increased lipid peroxidation and cellular oxidant levels but no alteration of cellular viability. There was a simultaneous increase in both Akt and GSK3β phosphorylation. Levels of phospho-FoxO3a (inactive form) increased in the cytosolic fraction of cells treated with iron in a PI3K-dependent manner. Moreover, PI3K and Akt translocated to the nucleus in response to oxidative stress. Iron-overloaded cells harboring a constitutively active form of Akt showed decreased oxidants levels. Indeed, GSH synthesis under oxidative stress conditions was regulated by activated Akt. Our results show that activation of the PI3K/Akt pathway during iron-induced neurotoxicity regulates multiple targets such as GSK3β, FoxO transcriptional activity, and glutathione metabolism, thus modulating the neuronal response to oxidative stress.
Collapse
Affiliation(s)
- Romina María Uranga
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur, 8000 Bahía Blanca, Argentina
| | | | | |
Collapse
|
49
|
Effective identification of Akt interacting proteins by two-step chemical crosslinking, co-immunoprecipitation and mass spectrometry. PLoS One 2013; 8:e61430. [PMID: 23613850 PMCID: PMC3629208 DOI: 10.1371/journal.pone.0061430] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 03/12/2013] [Indexed: 11/19/2022] Open
Abstract
Akt is a critical protein for cell survival and known to interact with various proteins. However, Akt binding partners that modulate or regulate Akt activation have not been fully elucidated. Identification of Akt-interacting proteins has been customarily achieved by co-immunoprecipitation combined with western blot and/or MS analysis. An intrinsic problem of the method is loss of interacting proteins during procedures to remove non-specific proteins. Moreover, antibody contamination often interferes with the detection of less abundant proteins. Here, we developed a novel two-step chemical crosslinking strategy to overcome these problems which resulted in a dramatic improvement in identifying Akt interacting partners. Akt antibody was first immobilized on protein A/G beads using disuccinimidyl suberate and allowed to bind to cellular Akt along with its interacting proteins. Subsequently, dithiobis[succinimidylpropionate], a cleavable crosslinker, was introduced to produce stable complexes between Akt and binding partners prior to the SDS-PAGE and nanoLC-MS/MS analysis. This approach enabled identification of ten Akt partners from cell lysates containing as low as 1.5 mg proteins, including two new potential Akt interacting partners. None of these but one protein was detectable without crosslinking procedures. The present method provides a sensitive and effective tool to probe Akt-interacting proteins. This strategy should also prove useful for other protein interactions, particularly those involving less abundant or weakly associating partners.
Collapse
|
50
|
Iglesias-Guimarais V, Gil-Guiñon E, Sánchez-Osuna M, Casanelles E, García-Belinchón M, Comella JX, Yuste VJ. Chromatin collapse during caspase-dependent apoptotic cell death requires DNA fragmentation factor, 40-kDa subunit-/caspase-activated deoxyribonuclease-mediated 3'-OH single-strand DNA breaks. J Biol Chem 2013; 288:9200-15. [PMID: 23430749 DOI: 10.1074/jbc.m112.411371] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Apoptotic nuclear morphology and oligonucleosomal double-strand DNA fragments (also known as DNA ladder) are considered the hallmarks of apoptotic cell death. From a classic point of view, these two processes occur concomitantly. Once activated, DNA fragmentation factor, 40-kDa subunit (DFF40)/caspase-activated DNase (CAD) endonuclease hydrolyzes the DNA into oligonucleosomal-size pieces, facilitating the chromatin package. However, the dogma that the apoptotic nuclear morphology depends on DNA fragmentation has been questioned. Here, we use different cellular models, including MEF CAD(-/-) cells, to unravel the mechanism by which DFF40/CAD influences chromatin condensation and nuclear collapse during apoptosis. Upon apoptotic insult, SK-N-AS cells display caspase-dependent apoptotic nuclear alterations in the absence of internucleosomal DNA degradation. The overexpression of a wild-type form of DFF40/CAD endonuclease, but not of different catalytic-null mutants, restores the cellular ability to degrade the chromatin into oligonucleosomal-length fragments. We show that apoptotic nuclear collapse requires a 3'-OH endonucleolytic activity even though the internucleosomal DNA degradation is impaired. Moreover, alkaline unwinding electrophoresis and In Situ End-Labeling (ISEL)/In Situ Nick Translation (ISNT) assays reveal that the apoptotic DNA damage observed in the DNA ladder-deficient SK-N-AS cells is characterized by the presence of single-strand nicks/breaks. Apoptotic single-strand breaks can be impaired by DFF40/CAD knockdown, abrogating nuclear collapse and disassembly. In conclusion, the highest order of chromatin compaction observed in the later steps of caspase-dependent apoptosis relies on DFF40/CAD-mediated DNA damage by generating 3'-OH ends in single-strand rather than double-strand DNA nicks/breaks.
Collapse
Affiliation(s)
- Victoria Iglesias-Guimarais
- Cell Death, Senescence, and Survival Group, Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|