1
|
Lin XJ, Wang ML, Kong WW, Mo BX. Molecular Studies on Plant Telomeres: Expanding Horizons in Plant Biology. ACS Synth Biol 2025. [PMID: 40340407 DOI: 10.1021/acssynbio.4c00846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The integrity of plant genomes is intricately safeguarded by telomeres, the protective caps located at the ends of the chromosome. This review provides a comprehensive analysis of the molecular mechanisms governing the structure, maintenance, and dynamics of plant telomeres, highlighting their genetic and epigenetic regulation and their pivotal roles in plant development, longevity, stress adaptation, and disease resistance. Recent advancements, such as next-generation sequencing and single-molecule imaging, have revolutionized our understanding of telomere biology, unveiling new insights into telomerase activity and telomere-associated genetic variants. Additionally, the review also discusses the challenges and future directions of telomere research, including the potential applications of telomere biology in plant breeding and genetic engineering.
Collapse
Affiliation(s)
- Xiao J Lin
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Ming L Wang
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Wen W Kong
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Bei X Mo
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
2
|
He L, Zhang X, Zhang S, Wang Y, Hu W, Li J, Liu Y, Liao Y, Peng X, Li J, Zhao H, Wang L, Lv Y, Hu C, Yang S. H. Pylori-Facilitated TERT/Wnt/β-Catenin Triggers Spasmolytic Polypeptide-Expressing Metaplasia and Oxyntic Atrophy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401227. [PMID: 39587848 PMCID: PMC11744579 DOI: 10.1002/advs.202401227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 10/23/2024] [Indexed: 11/27/2024]
Abstract
Persistent H. pylori infection triggers the repair program of the mucosa, such as spasmolytic polypeptide-expressing metaplasia (SPEM). However, the mechanism underlying the initiation of SPEM in gastric tissues by H. pylori remains unclear. Here, an increase in telomerase reverse transcriptase (TERT) protein expression is observed in chief cells upon infection with cagA-positive H. pylori. Tert knockout significantly ameliorated H. pylori-induced SPEM and single-cell RNA sequencing demonstrated that the Wnt/β-Catenin pathway is suppressed in gastric cells with Tert knockout. Mechanism study revealed that CagA elevated TERT abundance by disrupting the interaction between TERT and its novel E3 ligase, SYVN1. Interestingly, Nitazoxanide effectively relieved SPEM via inhibition of the Wnt/β-Catenin signaling in vivo. This results clarified the mechanism underlying which CagA activated the TERT/Wnt/β-Catenin pathway, thus promoting the dedifferentiation of chief cells and the occurrence of SPEM in gastric mucosa. This highlights a molecular basis for targeting CagA-activated Wnt signaling in chief cells for the treatment of gastric precancerous lesions.
Collapse
Affiliation(s)
- Lijiao He
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Xiao Zhang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Cancer Center of Daping HospitalArmy Medical UniversityChongqing400000China
| | - Shengwei Zhang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Department of GastroenterologyThe 987th Hospital of the Joint Logistics Support Force of the People's Liberation Army of China, BaojiShaanxi721000China
| | - Yi Wang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Biological Science Research CenterSouthwest UniversityChongqing400715China
| | - Weichao Hu
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Jie Li
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Yunyi Liu
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Yu Liao
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Xue Peng
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Jianjun Li
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Haiyan Zhao
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Liting Wang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
- Central LaboratoryArmy Medical UniversityChongqing400038China
| | - Yang‐Fan Lv
- Department of PathologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Chang‐Jiang Hu
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| | - Shi‐Ming Yang
- Department of GastroenterologyThe Second Affiliated Hospital of Army Medical UniversityChongqing400037China
| |
Collapse
|
3
|
Zuo WF, Pang Q, Zhu X, Yang QQ, Zhao Q, He G, Han B, Huang W. Heat shock proteins as hallmarks of cancer: insights from molecular mechanisms to therapeutic strategies. J Hematol Oncol 2024; 17:81. [PMID: 39232809 PMCID: PMC11375894 DOI: 10.1186/s13045-024-01601-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Heat shock proteins are essential molecular chaperones that play crucial roles in stabilizing protein structures, facilitating the repair or degradation of damaged proteins, and maintaining proteostasis and cellular functions. Extensive research has demonstrated that heat shock proteins are highly expressed in cancers and closely associated with tumorigenesis and progression. The "Hallmarks of Cancer" are the core features of cancer biology that collectively define a series of functional characteristics acquired by cells as they transition from a normal state to a state of tumor growth, including sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, enabled replicative immortality, the induction of angiogenesis, and the activation of invasion and metastasis. The pivotal roles of heat shock proteins in modulating the hallmarks of cancer through the activation or inhibition of various signaling pathways has been well documented. Therefore, this review provides an overview of the roles of heat shock proteins in vital biological processes from the perspective of the hallmarks of cancer and summarizes the small-molecule inhibitors that target heat shock proteins to regulate various cancer hallmarks. Moreover, we further discuss combination therapy strategies involving heat shock proteins and promising dual-target inhibitors to highlight the potential of targeting heat shock proteins for cancer treatment. In summary, this review highlights how targeting heat shock proteins could regulate the hallmarks of cancer, which will provide valuable information to better elucidate and understand the roles of heat shock proteins in oncology and the mechanisms of cancer occurrence and development and aid in the development of more efficacious and less toxic novel anticancer agents.
Collapse
Affiliation(s)
- Wei-Fang Zuo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiwen Pang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xinyu Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian-Qian Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian Zhao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gu He
- Department of Dermatology and Venereology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
4
|
Lee SE, Lee SB, Roh JI, Kim KP, Lee JH, Lee HW. SIRT1 regulates the localization and stability of telomerase protein by direct interaction. Biochem Biophys Res Commun 2024; 720:150098. [PMID: 38749190 DOI: 10.1016/j.bbrc.2024.150098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/05/2024]
Abstract
Telomerase reverse transcriptase (TERT) not only upholds telomeric equilibrium but also plays a pivotal role in multiple non-canonical cellular mechanisms, particularly in the context of aging, cancer, and genomic stability. Though depletion of SIRT1 in mouse embryonic fibroblasts has demonstrated telomere shortening, the impact of SIRT1 on enabling TERT to regulate telomeric homeostasis remains enigmatic. Here, we reveal that SIRT1 directly interacts with TERT, and promotes the nuclear localization and stability of TERT. Reverse transcriptase (RT) domain of TERT and N-terminus of SIRT1 mainly participated in their direct interaction. TERT, concomitantly expressed with intact SIRT1, exhibits nuclear localization, whereas TERT co-expressed with N-terminal-deleted SIRT1 remains in the cytosol. Furthermore, overexpression of SIRT1 enhances the nuclear localization and protein stability of TERT, akin to overexpression of deacetylase-inactive SIRT1, whereas N-terminal-deleted SIRT1 has no effect on TERT. These findings suggest a novel regulatory role of SIRT1 for TERT through direct interaction. This interaction provides new insights into the fields of aging, cancer, and genome stability governed by TERT and SIRT1.
Collapse
Affiliation(s)
- Seung Eon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Su Bin Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae-Il Roh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea; ReCerise Therapeutics Inc., 400-12, Yangcheon-ro, Gangseo-gu, Seoul, 07573, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin-si, 17104, Gyeonggi-do, Republic of Korea
| | - Jae Hoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea; GEMCRO, Inc., Seoul, 03722, Republic of Korea.
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea; GEMCRO, Inc., Seoul, 03722, Republic of Korea.
| |
Collapse
|
5
|
Liu M, Zhang Y, Jian Y, Gu L, Zhang D, Zhou H, Wang Y, Xu ZX. The regulations of telomerase reverse transcriptase (TERT) in cancer. Cell Death Dis 2024; 15:90. [PMID: 38278800 PMCID: PMC10817947 DOI: 10.1038/s41419-024-06454-7] [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: 09/22/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/28/2024]
Abstract
Abnormal activation of telomerase occurs in most cancer types, which facilitates escaping from cell senescence. As the key component of telomerase, telomerase reverse transcriptase (TERT) is regulated by various regulation pathways. TERT gene changing in its promoter and phosphorylation respectively leads to TERT ectopic expression at the transcription and protein levels. The co-interacting factors play an important role in the regulation of TERT in different cancer types. In this review, we focus on the regulators of TERT and these downstream functions in cancer regulation. Determining the specific regulatory mechanism will help to facilitate the development of a cancer treatment strategy that targets telomerase and cancer cell senescence. As the most important catalytic subunit component of telomerase, TERT is rapidly regulated by transcriptional factors and PTM-related activation. These changes directly influence TERT-related telomere maintenance by regulating telomerase activity in telomerase-positive cancer cells, telomerase assembly with telomere-binding proteins, and recruiting telomerase to the telomere. Besides, there are also non-canonical functions that are influenced by TERT, including the basic biological functions of cancer cells, such as proliferation, apoptosis, cell cycle regulation, initiating cell formation, EMT, and cell invasion. Other downstream effects are the results of the influence of transcriptional factors by TERT. Currently, some small molecular inhibitors of TERT and TERT vaccine are under research as a clinical therapeutic target. Purposeful work is in progress.
Collapse
Affiliation(s)
- Mingdi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Yuning Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Yongping Jian
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Liting Gu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Dan Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China.
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China.
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
6
|
Shepelev N, Dontsova O, Rubtsova M. Post-Transcriptional and Post-Translational Modifications in Telomerase Biogenesis and Recruitment to Telomeres. Int J Mol Sci 2023; 24:5027. [PMID: 36902458 PMCID: PMC10003056 DOI: 10.3390/ijms24055027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Telomere length is associated with the proliferative potential of cells. Telomerase is an enzyme that elongates telomeres throughout the entire lifespan of an organism in stem cells, germ cells, and cells of constantly renewed tissues. It is activated during cellular division, including regeneration and immune responses. The biogenesis of telomerase components and their assembly and functional localization to the telomere is a complex system regulated at multiple levels, where each step must be tuned to the cellular requirements. Any defect in the function or localization of the components of the telomerase biogenesis and functional system will affect the maintenance of telomere length, which is critical to the processes of regeneration, immune response, embryonic development, and cancer progression. An understanding of the regulatory mechanisms of telomerase biogenesis and activity is necessary for the development of approaches toward manipulating telomerase to influence these processes. The present review focuses on the molecular mechanisms involved in the major steps of telomerase regulation and the role of post-transcriptional and post-translational modifications in telomerase biogenesis and function in yeast and vertebrates.
Collapse
Affiliation(s)
- Nikita Shepelev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Olga Dontsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
- Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Maria Rubtsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| |
Collapse
|
7
|
Marinaccio J, Micheli E, Udroiu I, Di Nottia M, Carrozzo R, Baranzini N, Grimaldi A, Leone S, Moreno S, Muzzi M, Sgura A. TERT Extra-Telomeric Roles: Antioxidant Activity and Mitochondrial Protection. Int J Mol Sci 2023; 24:ijms24054450. [PMID: 36901881 PMCID: PMC10002448 DOI: 10.3390/ijms24054450] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/18/2023] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
Telomerase reverse transcriptase (TERT) is the catalytic subunit of telomerase holoenzyme, which adds telomeric DNA repeats on chromosome ends to counteract telomere shortening. In addition, there is evidence of TERT non-canonical functions, among which is an antioxidant role. In order to better investigate this role, we tested the response to X-rays and H2O2 treatment in hTERT-overexpressing human fibroblasts (HF-TERT). We observed in HF-TERT a reduced induction of reactive oxygen species and an increased expression of the proteins involved in the antioxidant defense. Therefore, we also tested a possible role of TERT inside mitochondria. We confirmed TERT mitochondrial localization, which increases after oxidative stress (OS) induced by H2O2 treatment. We next evaluated some mitochondrial markers. The basal mitochondria quantity appeared reduced in HF-TERT compared to normal fibroblasts and an additional reduction was observed after OS; nevertheless, the mitochondrial membrane potential and morphology were better conserved in HF-TERT. Our results suggest a protective function of TERT against OS, also preserving mitochondrial functionality.
Collapse
Affiliation(s)
| | - Emanuela Micheli
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
- Correspondence:
| | - Ion Udroiu
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
| | - Michela Di Nottia
- Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Rosalba Carrozzo
- Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Nicolò Baranzini
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Stefano Leone
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Sandra Moreno
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Maurizio Muzzi
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Antonella Sgura
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
| |
Collapse
|
8
|
Prodromou C, Aran-Guiu X, Oberoi J, Perna L, Chapple JP, van der Spuy J. HSP70-HSP90 Chaperone Networking in Protein-Misfolding Disease. Subcell Biochem 2023; 101:389-425. [PMID: 36520314 DOI: 10.1007/978-3-031-14740-1_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Molecular chaperones and their associated co-chaperones are essential in health and disease as they are key facilitators of protein-folding, quality control and function. In particular, the heat-shock protein (HSP) 70 and HSP90 molecular chaperone networks have been associated with neurodegenerative diseases caused by aberrant protein-folding. The pathogenesis of these disorders usually includes the formation of deposits of misfolded, aggregated protein. HSP70 and HSP90, plus their co-chaperones, have been recognised as potent modulators of misfolded protein toxicity, inclusion formation and cell survival in cellular and animal models of neurodegenerative disease. Moreover, these chaperone machines function not only in folding but also in proteasome-mediated degradation of neurodegenerative disease proteins. This chapter gives an overview of the HSP70 and HSP90 chaperones, and their respective regulatory co-chaperones, and explores how the HSP70 and HSP90 chaperone systems form a larger functional network and its relevance to counteracting neurodegenerative disease associated with misfolded proteins and disruption of proteostasis.
Collapse
Affiliation(s)
| | - Xavi Aran-Guiu
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Jasmeen Oberoi
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Laura Perna
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - J Paul Chapple
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | |
Collapse
|
9
|
FOXM1 increases hTERT protein stability and indicates poor prognosis in gastric cancer. Neoplasia 2022; 36:100863. [PMID: 36528911 PMCID: PMC9792884 DOI: 10.1016/j.neo.2022.100863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Gastric cancer is one of most lethal diseases across the world. However, the underlying mechanism of gastric cancer carcinogenesis and development is still not fully known. Forkhead box M1 (FOXM1) belongs to the FOX family and has crucial roles in transactivation of multiple oncogenes in several cancer types, including gastric cancer. Recent studies have also shown the non-transcriptional function of FOXM1 via protein-protein interactions. Human telomerase reverse transcriptase (hTERT) is the core subunit of telomerase that facilitates cancer initiation and progression by maintaining cell immortalization, promoting cell proliferation and inhibiting cell apoptosis. However, the relationship between FOXM1 and hTERT in gastric cancer is still unclear. In our study, we found that FOXM1 and hTERT were convergent to the cell cycle-related pathways and they were positively related with advanced gastric cancer stages and poor outcomes. Simultaneous high levels of FOXM1 and hTERT predicted the worst prognosis. FOXM1 could increase hTERT protein rather than mRNA levels in a non-transcriptional manner. Mechanistically, FOXM1 interrupted the interaction between the E3 ligase MKRN1 and hTERT and decreased hTERT protein degradation. Further studies revealed that FOXM1 interacted with hTERT through its DNA-binding domain (DBD) region. Finally, we found that hTERT played important roles in FOXM1-mediated activation of the Wnt/β-catenin pathway to promote gastric cancer cell proliferation. Taken together, we found a novel non-classical function of FOXM1 to increase hTERT protein stability. Targeting the FOXM1-hTERT pathway may be a potential therapeutic strategy in treating gastric cancer.
Collapse
|
10
|
Udroiu I, Marinaccio J, Sgura A. Many Functions of Telomerase Components: Certainties, Doubts, and Inconsistencies. Int J Mol Sci 2022; 23:ijms232315189. [PMID: 36499514 PMCID: PMC9736166 DOI: 10.3390/ijms232315189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
A growing number of studies have evidenced non-telomeric functions of "telomerase". Almost all of them, however, investigated the non-canonical effects of the catalytic subunit TERT, and not the telomerase ribonucleoprotein holoenzyme. These functions mainly comprise signal transduction, gene regulation and the increase of anti-oxidative systems. Although less studied, TERC (the RNA component of telomerase) has also been shown to be involved in gene regulation, as well as other functions. All this has led to the publication of many reviews on the subject, which, however, are often disseminating personal interpretations of experimental studies of other researchers as original proofs. Indeed, while some functions such as gene regulation seem ascertained, especially because mechanistic findings have been provided, other ones remain dubious and/or are contradicted by other direct or indirect evidence (e.g., telomerase activity at double-strand break site, RNA polymerase activity of TERT, translation of TERC, mitochondrion-processed TERC). In a critical study of the primary evidence so far obtained, we show those functions for which there is consensus, those showing contradictory results and those needing confirmation. The resulting picture, together with some usually neglected aspects, seems to indicate a link between TERT and TERC functions and cellular stemness and gives possible directions for future research.
Collapse
|
11
|
Kumar S, Basu M, Ghosh MK. Chaperone-assisted E3 ligase CHIP: A double agent in cancer. Genes Dis 2022; 9:1521-1555. [PMID: 36157498 PMCID: PMC9485218 DOI: 10.1016/j.gendis.2021.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022] Open
Abstract
The carboxy-terminus of Hsp70-interacting protein (CHIP) is a ubiquitin ligase and co-chaperone belonging to Ubox family that plays a crucial role in the maintenance of cellular homeostasis by switching the equilibrium of the folding-refolding mechanism towards the proteasomal or lysosomal degradation pathway. It links molecular chaperones viz. HSC70, HSP70 and HSP90 with ubiquitin proteasome system (UPS), acting as a quality control system. CHIP contains charged domain in between N-terminal tetratricopeptide repeat (TPR) and C-terminal Ubox domain. TPR domain interacts with the aberrant client proteins via chaperones while Ubox domain facilitates the ubiquitin transfer to the client proteins for ubiquitination. Thus, CHIP is a classic molecule that executes ubiquitination for degradation of client proteins. Further, CHIP has been found to be indulged in cellular differentiation, proliferation, metastasis and tumorigenesis. Additionally, CHIP can play its dual role as a tumor suppressor as well as an oncogene in numerous malignancies, thus acting as a double agent. Here, in this review, we have reported almost all substrates of CHIP established till date and classified them according to the hallmarks of cancer. In addition, we discussed about its architectural alignment, tissue specific expression, sub-cellular localization, folding-refolding mechanisms of client proteins, E4 ligase activity, normal physiological roles, as well as involvement in various diseases and tumor biology. Further, we aim to discuss its importance in HSP90 inhibitors mediated cancer therapy. Thus, this report concludes that CHIP may be a promising and worthy drug target towards pharmaceutical industry for drug development.
Collapse
Affiliation(s)
- Sunny Kumar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Paraganas, West Bengal 743372, India
| | - Mrinal K. Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
12
|
Yilmaz S, Bedir E, Ballar Kirmizibayrak P. The role of cycloastragenol at the intersection of NRF2/ARE, telomerase, and proteasome activity. Free Radic Biol Med 2022; 188:105-116. [PMID: 35718303 DOI: 10.1016/j.freeradbiomed.2022.06.230] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 10/18/2022]
Abstract
Aging is well-characterized by the gradual decline of cellular functionality. As redox balance, proteostasis, and telomerase systems have been found to be associated with aging and age-related diseases, targeting these systems with small compounds has been considered a promising therapeutic approach. Cycloastragenol (CA), a small molecule telomerase activator obtained from Astragalus species, has been reported to positively affect several age-related pathophysiologies, but the mechanisms underlying CA activity have yet to be reported. Here, we presented that CA increased NRF2 nuclear localization and activity leading to upregulation of cytoprotective enzymes and attenuation of oxidative stress-induced ROS levels. Furthermore, CA-mediated induction of telomerase activity was found to be regulated by NRF2. CA not only increased the expression of hTERT but also its nuclear localization via upregulating the Hsp90-chaperon complex. In addition to modulating nuclear hTERT levels at unstressed conditions, CA alleviated oxidative stress-induced mitochondrial hTERT levels while increasing nuclear hTERT levels. Concomitantly, H2O2-induced mitochondrial ROS level was found to be significantly decreased by CA administration. Our data also revealed that CA strongly enhanced proteasome activity and assembly. More importantly, the proteasome activator effect of CA is dependent on the induction of telomerase activity, which is mediated by NRF2 system. In conclusion, our results not only revealed the cross-talk among NRF2, telomerase, and proteasome systems but also that CA functions at the intersection of these three major aging-related cellular pathways.
Collapse
Affiliation(s)
- Sinem Yilmaz
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Bornova, Izmir, Turkey; Department of Bioengineering, Faculty of Engineering, University of Alanya Aladdin Keykubat, Antalya, Turkey
| | - Erdal Bedir
- Department of Bioengineering, Izmir Institute of Technology, 35430, Urla, Izmir, Turkey.
| | | |
Collapse
|
13
|
Wang K, Liu J, Li YL, Li JP, Zhang R. Ubiquitination/de-ubiquitination: A promising therapeutic target for PTEN reactivation in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188723. [DOI: 10.1016/j.bbcan.2022.188723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023]
|
14
|
Effect of oxidative stress on telomere maintenance in aortic smooth muscle cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166397. [PMID: 35346819 DOI: 10.1016/j.bbadis.2022.166397] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) and telomere dysfunction are both associated with aging and the development of age-related diseases. Although there is evidence for a direct relationship between ROS and telomere dysfunction as well as an independent association of oxidative stress and telomere attrition with age-related disorders, there has not been sufficient exploration of how the interaction between oxidative stress and telomere function may contribute to the pathophysiology of cardiovascular diseases (CVD). To better understand the complex relationships between oxidative stress, telomerase biology and pathophysiology, we examined the telomere biology of aortic smooth muscle cells (ASMCs) isolated from mutant mouse models of oxidative stress. We discovered that telomere lengths were significantly shorter in ASMCs isolated from superoxide dismutase 2 heterozygous (Sod2+/-) mice, which exhibit increased arterial stiffness with aging, and the observed telomere attrition occurred over time. Furthermore, the telomere erosion occurred even though telomerase activity increased. In contrast, telomeres remained stable in wild-type and superoxide dismutase 1 heterozygous (Sod1+/-) mice, which do not exhibit CVD phenotypes. The data indicate that mitochondrial oxidative stress, in particular elevated superoxide levels and decreased hydrogen peroxide levels, induces telomere erosion in the ASMCs of the Sod2+/- mice. This reduction in telomere length occurs despite an increase in telomerase activity and correlates with the onset of disease phenotype. Our results suggest that the oxidative stress caused by imbalance in mitochondrial ROS, from deficient SOD2 activity as a model for mitochondrial dysfunction results in telomere dysfunction, which may contribute to pathogenesis of CVD.
Collapse
|
15
|
Davis JA, Chakrabarti K. Telomerase ribonucleoprotein and genome integrity-An emerging connection in protozoan parasites. WILEY INTERDISCIPLINARY REVIEWS. RNA 2021; 13:e1710. [PMID: 34973045 DOI: 10.1002/wrna.1710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
Telomerase has an established role in telomere maintenance in eukaryotes. However, recent studies have begun to implicate telomerase in cellular roles beyond telomere maintenance. Specifically, evidence is emerging of cross-talks between telomerase mediated telomere homeostasis and DNA repair pathways. Telomere shortening due to the end replication problem is a constant threat to genome integrity in eukaryotic cells. This poses a particular problem in unicellular parasitic protists because their major virulence genes are located at the subtelomeric loci. Although telomerase is the major regulator of telomere lengthening in eukaryotes, it is less studied in the ancient eukaryotes, including clinically important human pathogens. Recent research is highlighting interplay between telomerase and the DNA damage response in human parasites. The importance of this interplay in pathogen virulence is only beginning to be illuminated, including the potential to highlight novel developmental regulation of telomerase in parasites who transition between multiple developmental stages throughout their life cycle. In this review, we will discuss the telomerase ribonucleoprotein enzyme and DNA repair pathways with emerging views in human parasites to give a broader perspective of the possible connection of telomere, telomerase, and DNA repair pathways across eukaryotic lineages and highlight their potential role in pathogen virulence. This article is categorized under: RNA Structure and Dynamics > Influence of RNA Structure in Biological Systems RNA Evolution and Genomics > RNA and Ribonucleoprotein Evolution RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
| | - Kausik Chakrabarti
- University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| |
Collapse
|
16
|
Lin KH, Ali A, Kuo CH, Yang PC, Kumar VB, Padma VV, Lo JF, Huang CY, Kuo WW. Carboxyl terminus of HSP70-interacting protein attenuates advanced glycation end products-induced cardiac injuries by promoting NFκB proteasomal degradation. J Cell Physiol 2021; 237:1888-1901. [PMID: 34958118 DOI: 10.1002/jcp.30660] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 11/06/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022]
Abstract
Advanced glycation end products (AGEs), which are highly reactive molecules resulting from persistent high-glucose levels, can lead to the generation of oxidative stress and cardiac complications. The carboxyl terminus of HSP70 interacting protein (CHIP) has been demonstrated to have a protective role in several diseases, including cardiac complications; however, the role in preventing AGE-induced cardiac damages remains poorly understood. Here, we found that elevated AGE levels impaired cardiac CHIP expression in streptozotocin-induced diabetes and high-fat diet-administered animals, representing AGE exposure models. We used the TUNEL assay, hematoxylin and eosin, Masson's trichrome staining, and western blotting to prove that cardiac injuries were induced in diabetic animals and AGE-treated cardiac cells. Interestingly, our results collectively indicated that CHIP overexpression significantly rescued the AGE-induced cardiac injuries and promoted cell survival. Moreover, CHIP knockdown-mediated stabilization of nuclear factor κB (NFκB) was attenuated by overexpressing CHIP in the cells. Furthermore, co-immunoprecipitation and immunoblot assay revealed that CHIP promotes the ubiquitination and proteasomal degradation of AGE-induced NFκB. Importantly, fluorescence microscopy, a luciferase reporter assay, electrophoretic mobility shift assay, and subcellular fractionation further demonstrated that CHIP overexpression inhibits AGE-induced NFκB nuclear translocation, reduced its binding ability with the promoter sequences of the receptor of AGE, consequently inhibiting the translocation of the receptor AGE to the cell membrane for its proper function. Overall, our current study findings suggest that CHIP can target NFκB for ubiquitin-mediated proteasomal degradation, and thereby potentially rescue AGE-induced cardiac damages.
Collapse
Affiliation(s)
- Kuan-Ho Lin
- College of Medicine, China Medical University, Taichung, Taiwan.,Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ayaz Ali
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Pei-Chen Yang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | | | | | - Jeng-Fan Lo
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Genome Research Centre, National Yang-Ming University, Taipei, Taiwan.,Cancer Progression Centre of Excellence, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yang Huang
- Department of Biotechnology, Asia University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Centre of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| |
Collapse
|
17
|
Nersisyan L, Simonyan A, Binder H, Arakelyan A. Telomere Maintenance Pathway Activity Analysis Enables Tissue- and Gene-Level Inferences. Front Genet 2021; 12:662464. [PMID: 33897770 PMCID: PMC8058386 DOI: 10.3389/fgene.2021.662464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/16/2021] [Indexed: 12/31/2022] Open
Abstract
Telomere maintenance is one of the mechanisms ensuring indefinite divisions of cancer and stem cells. Good understanding of telomere maintenance mechanisms (TMM) is important for studying cancers and designing therapies. However, molecular factors triggering selective activation of either the telomerase dependent (TEL) or the alternative lengthening of telomeres (ALT) pathway are poorly understood. In addition, more accurate and easy-to-use methodologies are required for TMM phenotyping. In this study, we have performed literature based reconstruction of signaling pathways for the ALT and TEL TMMs. Gene expression data were used for computational assessment of TMM pathway activities and compared with experimental assays for TEL and ALT. Explicit consideration of pathway topology makes bioinformatics analysis more informative compared to computational methods based on simple summary measures of gene expression. Application to healthy human tissues showed high ALT and TEL pathway activities in testis, and identified genes and pathways that may trigger TMM activation. Our approach offers a novel option for systematic investigation of TMM activation patterns across cancers and healthy tissues for dissecting pathway-based molecular markers with diagnostic impact.
Collapse
Affiliation(s)
- Lilit Nersisyan
- Bioinformatics Group, Institute of Molecular Biology, National Academy of Sciences, Yerevan, Armenia.,Pathverse, Yerevan, Armenia
| | - Arman Simonyan
- Bioinformatics Group, Institute of Molecular Biology, National Academy of Sciences, Yerevan, Armenia
| | - Hans Binder
- Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Arsen Arakelyan
- Bioinformatics Group, Institute of Molecular Biology, National Academy of Sciences, Yerevan, Armenia.,Pathverse, Yerevan, Armenia
| |
Collapse
|
18
|
Seo HR, Jeong D, Lee S, Lee HS, Lee SA, Kang SW, Kwon J. CHIP and BAP1 Act in Concert to Regulate INO80 Ubiquitination and Stability for DNA Replication. Mol Cells 2021; 44:101-115. [PMID: 33658435 PMCID: PMC7941006 DOI: 10.14348/molcells.2021.2258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
The INO80 chromatin remodeling complex has roles in many essential cellular processes, including DNA replication. However, the mechanisms that regulate INO80 in these processes remain largely unknown. We previously reported that the stability of Ino80, the catalytic ATPase subunit of INO80, is regulated by the ubiquitin proteasome system and that BRCA1-associated protein-1 (BAP1), a nuclear deubiquitinase with tumor suppressor activity, stabilizes Ino80 via deubiquitination and promotes replication fork progression. However, the E3 ubiquitin ligase that targets Ino80 for proteasomal degradation was unknown. Here, we identified the C-terminus of Hsp70-interacting protein (CHIP), the E3 ubiquitin ligase that functions in cooperation with Hsp70, as an Ino80-interacting protein. CHIP polyubiquitinates Ino80 in a manner dependent on Hsp70. Contrary to our expectation that CHIP degrades Ino80, CHIP instead stabilizes Ino80 by extending its halflife. The data suggest that CHIP stabilizes Ino80 by inhibiting degradative ubiquitination. We also show that CHIP works together with BAP1 to enhance the stabilization of Ino80, leading to its chromatin binding. Interestingly, both depletion and overexpression of CHIP compromise replication fork progression with little effect on fork stalling, as similarly observed for BAP1 and Ino80, indicating that an optimal cellular level of Ino80 is important for replication fork speed but not for replication stress suppression. This work therefore idenitifes CHIP as an E3 ubiquitin ligase that stabilizes Ino80 via nondegradative ubiquitination and suggests that CHIP and BAP1 act in concert to regulate Ino80 ubiquitination to fine-tune its stability for efficient DNA replication.
Collapse
Affiliation(s)
- Hye-Ran Seo
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Daun Jeong
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Sunmi Lee
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| | - Han-Sae Lee
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Shin-Ai Lee
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
- Present address: Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Sang Won Kang
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| | - Jongbum Kwon
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
19
|
Engin AB, Engin A. The Connection Between Cell Fate and Telomere. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:71-100. [PMID: 33539012 DOI: 10.1007/978-3-030-49844-3_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Abolition of telomerase activity results in telomere shortening, a process that eventually destabilizes the ends of chromosomes, leading to genomic instability and cell growth arrest or death. Telomere shortening leads to the attainment of the "Hayflick limit", and the transition of cells to state of senescence. If senescence is bypassed, cells undergo crisis through loss of checkpoints. This process causes massive cell death concomitant with further telomere shortening and spontaneous telomere fusions. In functional telomere of mammalian cells, DNA contains double-stranded tandem repeats of TTAGGG. The Shelterin complex, which is composed of six different proteins, is required for the regulation of telomere length and stability in cells. Telomere protection by telomeric repeat binding protein 2 (TRF2) is dependent on DNA damage response (DDR) inhibition via formation of T-loop structures. Many protein kinases contribute to the DDR activated cell cycle checkpoint pathways, and prevent DNA replication until damaged DNA is repaired. Thereby, the connection between cell fate and telomere length-associated telomerase activity is regulated by multiple protein kinase activities. Contrarily, inactivation of DNA damage checkpoint protein kinases in senescent cells can restore cell-cycle progression into S phase. Therefore, telomere-initiated senescence is a DNA damage checkpoint response that is activated with a direct contribution from dysfunctional telomeres. In this review, in addition to the above mentioned, the choice of main repair pathways, which comprise non-homologous end joining and homologous recombination in telomere uncapping telomere dysfunctions, are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
20
|
Yuan X, Dai M, Xu D. Telomere-related Markers for Cancer. Curr Top Med Chem 2020; 20:410-432. [PMID: 31903880 PMCID: PMC7475940 DOI: 10.2174/1568026620666200106145340] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/03/2019] [Accepted: 12/14/2019] [Indexed: 02/06/2023]
Abstract
Telomeres are structurally nucleoprotein complexes at termini of linear chromosomes and essential to chromosome stability/integrity. In normal human cells, telomere length erodes progressively with each round of cell divisions, which serves as an important barrier to uncontrolled proliferation and malignant transformation. In sharp contrast, telomere maintenance is a key feature of human malignant cells and required for their infinite proliferation and maintenance of other cancer hallmarks as well. Thus, a telomere-based anti-cancer strategy has long been suggested. However, clinically efficient and specific drugs targeting cancer telomere-maintenance have still been in their infancy thus far. To achieve this goal, it is highly necessary to elucidate how exactly cancer cells maintain functional telomeres. In the last two decades, numerous studies have provided profound mechanistic insights, and the identified mechanisms include the aberrant activation of telomerase or the alternative lengthening of telomere pathway responsible for telomere elongation, dysregulation and mutation of telomere-associated factors, and other telomere homeostasis-related signaling nodes. In the present review, these various strategies employed by malignant cells to regulate their telomere length, structure and function have been summarized, and potential implications of these findings in the rational development of telomere-based cancer therapy and other clinical applications for precision oncology have been discussed.
Collapse
Affiliation(s)
- Xiaotian Yuan
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
| | - Mingkai Dai
- Central Research Laboratory, Shandong University Second Hospital, Jinan, 250033, China.,Karolinska Institute Collaborative Laboratory for Cancer and Stem Cell Research, Shandong University Second Hospital, Jinan, 250033, China
| | - Dawei Xu
- Karolinska Institute Collaborative Laboratory for Cancer and Stem Cell Research, Shandong University Second Hospital, Jinan, 250033, China.,Department of Medicine, Division of Hematology, Center for Molecular Medicine (CMM) and Bioclinicum, Karolinska Institute and Karolinska University Hospital Solna, Solna 171 64, Sweden
| |
Collapse
|
21
|
Telomerase Biogenesis and Activities from the Perspective of Its Direct Interacting Partners. Cancers (Basel) 2020; 12:cancers12061679. [PMID: 32599885 PMCID: PMC7352425 DOI: 10.3390/cancers12061679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022] Open
Abstract
Telomerase reverse transcriptase (TERT)—the catalytic subunit of telomerase—is reactivated in up to 90% of all human cancers. TERT is observed in heterogenous populations of protein complexes, which are dynamically regulated in a cell type- and cell cycle-specific manner. Over the past two decades, in vitro protein–protein interaction detection methods have discovered a number of endogenous TERT binding partners in human cells that are responsible for the biogenesis and functionalization of the telomerase holoenzyme, including the processes of TERT trafficking between subcellular compartments, assembly into telomerase, and catalytic action at telomeres. Additionally, TERT have been found to interact with protein species with no known telomeric functions, suggesting that these complexes may contribute to non-canonical activities of TERT. Here, we survey TERT direct binding partners and discuss their contributions to TERT biogenesis and functions. The goal is to review the comprehensive spectrum of TERT pro-malignant activities, both telomeric and non-telomeric, which may explain the prevalence of its upregulation in cancer.
Collapse
|
22
|
Zheng JT, Zhang N, Yu YH, Gong PT, Li XH, Wu N, Wang C, Wang XC, Li X, Li JH, Zhang XC. Identification of a TRBD zinc finger-interacting protein in Giardia duodenalis and its regulation of telomerase. Parasit Vectors 2019; 12:568. [PMID: 31783771 PMCID: PMC6884763 DOI: 10.1186/s13071-019-3821-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/21/2019] [Indexed: 11/10/2022] Open
Abstract
Background Giardia duodenalis causes giardiasis, with diarrhea as the primary symptom. The trophozoite proliferation of this zoonotic parasite is mainly affected by telomerase, although the mechanism of telomerase regulation has not been thoroughly analyzed. Methods This study was performed to identify the telomerase RNA-binding domain (TRBD)-interacting protein in G. duodenalis and its regulation of telomerase. Interaction between TRBD and interacting proteins was verified via pulldown assays and co-immunoprecipitation (co-IP) techniques, and the subcellular localization of the protein interactions was determined in vivo via split SNAP-tag labeling. The hammerhead ribozyme was designed to deplete the mRNA of TRBD-interacting proteins. Results Using TRBD as bait, we identified zinc-finger domain (ZFD)-containing proteins and verified it via pulldown and co-IP experiments. Protein-protein interaction occurred in the nuclei of 293T cells and both nuclei of G. duodenalis. The hammerhead ribozyme depleted ZFD mRNA levels, which reduced the reproduction rate of G. duodenalis, telomerase activity and telomere length. Conclusions Our findings suggest that ZFD may regulate telomere function in G. duodenalis nuclei.
Collapse
Affiliation(s)
- Jing-Tong Zheng
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China.,Department of Pathogenobiology, College of Basic Medicine, Jilin University, Changchun, 130021, Jilin, China
| | - Nan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China.,State and Local Joint Engineering Laboratory for Animal Models of Human Diseases, Academy of Translational Medicine, First Hospital, Jilin University, Changchun, 130021, China
| | - Yan-Hui Yu
- Clinical Laboratory of Second Hospital, Jilin University, Changchun, 130021, China
| | - Peng-Tao Gong
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Xian-He Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Na Wu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Can Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Xiao-Cen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Xin Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China
| | - Jian-Hua Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China.
| | - Xi-Chen Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, 130062, China.
| |
Collapse
|
23
|
Tanwar K, Pati U. Inhibition of apoptosis via CHIP-mediated proteasomal degradation of TAp73α. J Cell Biochem 2019; 120:11091-11103. [PMID: 30714204 DOI: 10.1002/jcb.28386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 11/22/2018] [Accepted: 11/29/2018] [Indexed: 01/24/2023]
Abstract
TAp73, a homologous of tumor suppressor p53, regulates apoptosis in a p53-independent manner and its suppressive as well as stimulatory role in promoting angiogenesis has been reported. It exists in multiple isoforms which varies structurally in their N-terminus and C-terminus region and crucial interplay among them guides the decision of cell survival and death. As molecular chaperones control both stability and degradation of TAp73, selective regulation of p73 isoforms has implication upon developing new therapeutic for hypoxic tumor. We have discovered that under DNA damage carboxy terminus Hsp70 interacting protein (CHIP's) antiapoptotic function is displayed via its E3 ligase activity that inhibits exclusively TAp73α-mediated apoptosis in cancer cell. The decrease in TAp73α level by CHIP as it is supported by increased ubiquitination pattern is reverted back by sh-CHIP. Further, the transactivation of p53-downstream apoptotic genes BAX, PUMA and PIG3 by TAp73α is also shown to be subsequently inhibited by CHIP. The tetratricopeptide TPR-domain of CHIP in its amino-terminus interacts with the carboxy-terminus of TAp73α and ΔNp73α and as a result, U-BOX domain of CHIP in the carboxy-terminus is able to ubiquitinate TAp73α for proteasomal degradation. Due to lack of C-terminus in TAp73β, CHIP fails to interact with and degrade it. In conclusion, we have thus uncovered for the first time a novel mechanism of chaperone-assisted regulation of p73 stability as well as its apoptotic functions by CHIP that might be utilized to develop new anticancer strategies.
Collapse
Affiliation(s)
- Kamia Tanwar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Uttam Pati
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
24
|
Jie MM, Chang X, Zeng S, Liu C, Liao GB, Wu YR, Liu CH, Hu CJ, Yang SM, Li XZ. Diverse regulatory manners of human telomerase reverse transcriptase. Cell Commun Signal 2019; 17:63. [PMID: 31186051 PMCID: PMC6560729 DOI: 10.1186/s12964-019-0372-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/17/2019] [Indexed: 12/22/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) is the core subunit of human telomerase and plays important roles in human cancers. Aberrant expression of hTERT is closely associated with tumorigenesis, cancer cell stemness maintaining, cell proliferation, apoptosis inhibition, senescence evasion and metastasis. The molecular basis of hTERT regulation is highly complicated and consists of various layers. A deep and full-scale comprehension of the regulatory mechanisms of hTERT is pivotal in understanding the pathogenesis and searching for therapeutic approaches. In this review, we summarize the recent advances regarding the diverse regulatory mechanisms of hTERT, including the transcriptional (promoter mutation, promoter region methylation and histone acetylation), post-transcriptional (mRNA alternative splicing and non-coding RNAs) and post-translational levels (phosphorylation and ubiquitination), which may provide novel perspectives for further translational diagnosis or therapeutic strategies targeting hTERT.
Collapse
Affiliation(s)
- Meng-Meng Jie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Xing Chang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Shuo Zeng
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Cheng Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Guo-Bin Liao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ya-Ran Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Chun-Hua Liu
- Teaching evaluation center of Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
| | - Xin-Zhe Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
| |
Collapse
|
25
|
Quintana-Gallardo L, Martín-Benito J, Marcilla M, Espadas G, Sabidó E, Valpuesta JM. The cochaperone CHIP marks Hsp70- and Hsp90-bound substrates for degradation through a very flexible mechanism. Sci Rep 2019; 9:5102. [PMID: 30911017 PMCID: PMC6433865 DOI: 10.1038/s41598-019-41060-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/22/2019] [Indexed: 11/26/2022] Open
Abstract
Some molecular chaperones are involved not only in assisting the folding of proteins but also, given appropriate conditions, in their degradation. This is the case for Hsp70 and Hsp90 which, in concert with the cochaperone CHIP, direct their bound substrate to degradation through ubiquitination. We generated complexes between the chaperones (Hsp70 or Hsp90), the cochaperone CHIP and, as substrate, a p53 variant containing the GST protein (p53-TMGST). Both ternary complexes (Hsp70:p53-TMGST:CHIP and Hsp90:p53-TMGST:CHIP) ubiquitinated the substrate at a higher efficiency than in the absence of the chaperones. The 3D structures of the two complexes, obtained using a combination of cryoelectron microscopy and crosslinking mass spectrometry, showed the substrate located between the chaperone and the cochaperone, suggesting a ubiquitination mechanism in which the chaperone-bound substrate is presented to CHIP. These complexes are inherently flexible, which is important for the ubiquitination process.
Collapse
Affiliation(s)
| | | | - Miguel Marcilla
- Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049, Madrid, Spain
| | - Guadalupe Espadas
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | - Eduard Sabidó
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Proteomics Unit, Universitat Pompeu Fabra, Barcelona, Spain
| | | |
Collapse
|
26
|
Seo J, Han SY, Seong D, Han HJ, Song J. Multifaceted C-terminus of HSP70-interacting protein regulates tumorigenesis via protein quality control. Arch Pharm Res 2019; 42:63-75. [PMID: 30600426 DOI: 10.1007/s12272-018-1101-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 12/11/2018] [Indexed: 10/27/2022]
Abstract
C-terminus of heat shock protein 70 (HSP70)-interacting protein (CHIP) is an E3 ligase involved in a variety of protein homeostasis events implicated in diverse signaling pathways. Its involvement in varied and even opposite signaling circuits might be due to its hallmark signature of associating with molecular chaperones, including HSP90 and HSP70. Together, these proteins may be pivotal in implementing protein quality control. A curious and puzzling aspect of the function of CHIP is its capability to induce protein degradation via the proteasome- or lysosome-dependent pathways. In addition, these pathways are combined with ubiquitin-dependent or -independent pathways. This review focuses on the role of CHIP in the development or suppression of tumorigenesis. CHIP can act as a tumor suppressor by downregulating various oncogenes. CHIP also displays an oncogenic feature involving the inhibition of diverse tumor suppressors, including proteins related to intrinsic and extrinsic apoptotic pathways. The ability of CHIP to exhibit dual roles in determining the fate of cells has not been studied analytically. However, its association with various proteins involved in protein quality control might play a major role. In this review, the mechanistic roles of CHIP in tumor formation based on the regulation of diverse proteins are discussed.
Collapse
Affiliation(s)
- Jinho Seo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Su Yeon Han
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Daehyeon Seong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Hyun-Ji Han
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
27
|
Chen B, Zhang Y, Yang Y, Chen S, Xu A, Wu L, Xu S. Involvement of telomerase activity inhibition and telomere dysfunction in silver nanoparticles anticancer effects. Nanomedicine (Lond) 2018; 13:2067-2082. [PMID: 30203702 DOI: 10.2217/nnm-2018-0036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIM To investigate the possible mechanisms of telomerase and telomere underlying the anticancer effects of silver nanoparticles (AgNPs). MATERIALS & METHODS 25nm polyvinylpyrrolidone-coated AgNPs were used. The telomerase activity and telomere function were evaluated. The anticancer effects of AgNPs were gauged with cell viability assay under different statement of telomerase and telomere. RESULTS & CONCLUSION AgNPs could inhibit telomerase activity and lead to telomere shortening and dysfunction. Overexpression of telomerase attenuated the anticancer activity of AgNPs, whereas downregulation of telomerase activity or dysfunction of the telomere enhanced the cytotoxicity of AgNPs in HeLa cells. Our findings provided strong evidence that the anticancer effects of AgNPs were mediated via interference with the telomerase/telomere.
Collapse
Affiliation(s)
- Biao Chen
- School of Environmental Science & Optoelectronic Technology, University of Science & Technology of China, Hefei, Anhui, 230026, PR China.,Key Laboratory of High Magnetic Field & Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Yajun Zhang
- Key Laboratory of High Magnetic Field & Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China.,Institute of Physical & Information Technology, Anhui University, Hefei, Anhui, 230601, PR China
| | - Yaning Yang
- School of Environmental Science & Optoelectronic Technology, University of Science & Technology of China, Hefei, Anhui, 230026, PR China.,Key Laboratory of High Magnetic Field & Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China
| | - Shaopeng Chen
- Key Laboratory of High Magnetic Field & Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China.,Institute of Physical & Information Technology, Anhui University, Hefei, Anhui, 230601, PR China
| | - An Xu
- School of Environmental Science & Optoelectronic Technology, University of Science & Technology of China, Hefei, Anhui, 230026, PR China.,Key Laboratory of High Magnetic Field & Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China.,Institute of Physical & Information Technology, Anhui University, Hefei, Anhui, 230601, PR China
| | - Lijun Wu
- School of Environmental Science & Optoelectronic Technology, University of Science & Technology of China, Hefei, Anhui, 230026, PR China.,Key Laboratory of High Magnetic Field & Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China.,Key Laboratory of Environmental Toxicology & Pollution Control Technology of Anhui Province, Hefei, Anhui, 230031, PR China.,Institute of Physical & Information Technology, Anhui University, Hefei, Anhui, 230601, PR China
| | - Shengmin Xu
- Key Laboratory of High Magnetic Field & Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, PR China.,Key Laboratory of Environmental Toxicology & Pollution Control Technology of Anhui Province, Hefei, Anhui, 230031, PR China
| |
Collapse
|
28
|
Maan M, Pati U. CHIP promotes autophagy-mediated degradation of aggregating mutant p53 in hypoxic conditions. FEBS J 2018; 285:3197-3214. [PMID: 29953728 DOI: 10.1111/febs.14602] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/05/2018] [Accepted: 06/26/2018] [Indexed: 12/18/2022]
Abstract
Tumor suppressor protein p53 aggregates in the hypoxic core of solid tumors. C terminus of Hsc70-interacting protein (CHIP) displays chaperone as well as E3 ligase activities in both stabilizing and degrading wild-type and mutant p53. In this study, we have discovered that CHIP selectively degrades aggregating mutant p53 under both normal and hypoxic conditions. Silencing of CHIP alleviates degradation of aggregating mutant p53 in both normoxia and hypoxia, but has no significant effect on the level of nonaggregating mutant p53. Although both U-box and TPR domains of CHIP are responsible for p53 degradation, the U-box domain selectively binds to aggregating mutant p53, whereas the TPR domain interacts with nonaggregating mutant p53. The degradation of mutant p53 by CHIP is shown to be via autophagy through K63-linked polyubiquitination. Both in normoxia and under physiological hypoxia, the level of aggregating mutant p53 in the presence of CHIP was reduced threefold, whereas under serum starvation, it was reduced fivefold. Interestingly, both wild-type and mutant p53 interact with and stabilize CHIP at the post-translational level, suggesting a chaperone synergy between p53 and CHIP. This finding may have strong therapeutic significance via selective degradation of oncogenic mutant p53 in regressing hypoxic tumors.
Collapse
Affiliation(s)
- Meenu Maan
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Uttam Pati
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
29
|
Peng J, Wang H, Lu J, Hui W, Wang Y, Shang X. Identifying term relations cross different gene ontology categories. BMC Bioinformatics 2017; 18:573. [PMID: 29297309 PMCID: PMC5751813 DOI: 10.1186/s12859-017-1959-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background The Gene Ontology (GO) is a community-based bioinformatics resource that employs ontologies to represent biological knowledge and describes information about gene and gene product function. GO includes three independent categories: molecular function, biological process and cellular component. For better biological reasoning, identifying the biological relationships between terms in different categories are important. However, the existing measurements to calculate similarity between terms in different categories are either developed by using the GO data only or only take part of combined gene co-function network information. Results We propose an iterative ranking-based method called CroGO2 to measure the cross-categories GO term similarities by incorporating level information of GO terms with both direct and indirect interactions in the gene co-function network. Conclusions The evaluation test shows that CroGO2 performs better than the existing methods. A genome-specific term association network for yeast is also generated by connecting terms with the high confidence score. The linkages in the term association network could be supported by the literature. Given a gene set, the related terms identified by using the association network have overlap with the related terms identified by GO enrichment analysis.
Collapse
Affiliation(s)
- Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Honggang Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Junya Lu
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Weiwei Hui
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China.
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China.
| |
Collapse
|
30
|
Majerská J, Schrumpfová PP, Dokládal L, Schořová Š, Stejskal K, Obořil M, Honys D, Kozáková L, Polanská PS, Sýkorová E. Tandem affinity purification of AtTERT reveals putative interaction partners of plant telomerase in vivo. PROTOPLASMA 2017; 254:1547-1562. [PMID: 27853871 DOI: 10.1007/s00709-016-1042-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/04/2016] [Indexed: 05/15/2023]
Abstract
The life cycle of telomerase involves dynamic and complex interactions between proteins within multiple macromolecular networks. Elucidation of these associations is a key to understanding the regulation of telomerase under diverse physiological and pathological conditions from telomerase biogenesis, through telomere recruitment and elongation, to its non-canonical activities outside of telomeres. We used tandem affinity purification coupled to mass spectrometry to build an interactome of the telomerase catalytic subunit AtTERT, using Arabidopsis thaliana suspension cultures. We then examined interactions occurring at the AtTERT N-terminus, which is thought to fold into a discrete domain connected to the rest of the molecule via a flexible linker. Bioinformatic analyses revealed that interaction partners of AtTERT have a range of molecular functions, a subset of which is specific to the network around its N-terminus. A significant number of proteins co-purifying with the N-terminal constructs have been implicated in cell cycle and developmental processes, as would be expected of bona fide regulatory interactions and we have confirmed experimentally the direct nature of selected interactions. To examine AtTERT protein-protein interactions from another perspective, we also analysed AtTERT interdomain contacts to test potential dimerization of AtTERT. In total, our results provide an insight into the composition and architecture of the plant telomerase complex and this will aid in delineating molecular mechanisms of telomerase functions.
Collapse
Affiliation(s)
- Jana Majerská
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Královopolská 135, CZ-61265, Brno, Czech Republic
- Central European Institute of Technology and Faculty of Science, Masaryk University, Kotlářská 2, CZ-61137, Brno, Czech Republic
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Petra Procházková Schrumpfová
- Central European Institute of Technology and Faculty of Science, Masaryk University, Kotlářská 2, CZ-61137, Brno, Czech Republic
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Ladislav Dokládal
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Královopolská 135, CZ-61265, Brno, Czech Republic
| | - Šárka Schořová
- Central European Institute of Technology and Faculty of Science, Masaryk University, Kotlářská 2, CZ-61137, Brno, Czech Republic
| | - Karel Stejskal
- Central European Institute of Technology and Faculty of Science, Masaryk University, Kotlářská 2, CZ-61137, Brno, Czech Republic
| | - Michal Obořil
- Central European Institute of Technology and Faculty of Science, Masaryk University, Kotlářská 2, CZ-61137, Brno, Czech Republic
| | - David Honys
- Institute of Experimental Biology, Academy of Sciences of the Czech Republic, v.v.i., Rozvojová 263, CZ-165 02, Prague, Czech Republic
| | - Lucie Kozáková
- Central European Institute of Technology and Faculty of Science, Masaryk University, Kotlářská 2, CZ-61137, Brno, Czech Republic
| | - Pavla Sováková Polanská
- Central European Institute of Technology and Faculty of Science, Masaryk University, Kotlářská 2, CZ-61137, Brno, Czech Republic
| | - Eva Sýkorová
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Královopolská 135, CZ-61265, Brno, Czech Republic.
| |
Collapse
|
31
|
Yalçin Z, Selenz C, Jacobs JJL. Ubiquitination and SUMOylation in Telomere Maintenance and Dysfunction. Front Genet 2017; 8:67. [PMID: 28588610 PMCID: PMC5440461 DOI: 10.3389/fgene.2017.00067] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 05/10/2017] [Indexed: 12/14/2022] Open
Abstract
Telomeres are essential nucleoprotein structures at linear chromosomes that maintain genome integrity by protecting chromosome ends from being recognized and processed as damaged DNA. In addition, they limit the cell’s proliferative capacity, as progressive loss of telomeric DNA during successive rounds of cell division eventually causes a state of telomere dysfunction that prevents further cell division. When telomeres become critically short, the cell elicits a DNA damage response resulting in senescence, apoptosis or genomic instability, thereby impacting on aging and tumorigenesis. Over the past years substantial progress has been made in understanding the role of post-translational modifications in telomere-related processes, including telomere maintenance, replication and dysfunction. This review will focus on recent findings that establish an essential role for ubiquitination and SUMOylation at telomeres.
Collapse
Affiliation(s)
- Zeliha Yalçin
- Department of Molecular Oncology, Netherlands Cancer InstituteAmsterdam, Netherlands
| | - Carolin Selenz
- Department of Molecular Oncology, Netherlands Cancer InstituteAmsterdam, Netherlands
| | - Jacqueline J L Jacobs
- Department of Molecular Oncology, Netherlands Cancer InstituteAmsterdam, Netherlands
| |
Collapse
|
32
|
Heat Shock Protein 90 Ensures Efficient Mumps Virus Replication by Assisting with Viral Polymerase Complex Formation. J Virol 2017; 91:JVI.02220-16. [PMID: 28053100 DOI: 10.1128/jvi.02220-16] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/21/2016] [Indexed: 12/30/2022] Open
Abstract
Paramyxoviral RNAs are synthesized by a viral RNA-dependent RNA polymerase (RdRp) consisting of the large (L) protein and its cofactor phosphoprotein (P protein). The L protein is a multifunctional protein that catalyzes RNA synthesis, mRNA capping, and mRNA polyadenylation. Growing evidence shows that the stability of several paramyxovirus L proteins is regulated by heat shock protein 90 (Hsp90). In this study, we demonstrated that Hsp90 activity was important for mumps virus (MuV) replication. The Hsp90 activity was required for L-protein stability and activity because an Hsp90-specific inhibitor, 17-allylamino-17-demethoxygeldanamycin (17-AAG), destabilized the MuV L protein and suppressed viral RNA synthesis. However, once the L protein formed a mature polymerase complex with the P protein, Hsp90 activity was no longer required for the stability and activity of the L protein. When the Hsp90 activity was inhibited, the MuV L protein was degraded through the CHIP (C terminus of Hsp70-interacting protein)-mediated proteasomal pathway. High concentrations of 17-AAG showed strong cytotoxicity to certain cell types, but combined use of an Hsp70 inhibitor, VER155008, potentiated degradation of the L protein, allowing a sufficient reduction of 17-AAG concentration to block MuV replication with minimum cytotoxicity. Regulation of the L protein by Hsp90 and Hsp70 chaperones was also demonstrated for another paramyxovirus, the measles virus. Collectively, our data show that the Hsp90/Hsp70 chaperone machinery assists in the maturation of the paramyxovirus L protein and thereby in the formation of a mature RdRp complex and efficient viral replication.IMPORTANCE Heat shock protein 90 (Hsp90) is nearly universally required for viral protein homeostasis. Here, we report that Hsp90 activity is required for efficient propagation of mumps virus (MuV). Hsp90 functions in the maintenance of the catalytic subunit of viral polymerase, the large (L) protein, prior to formation of a mature polymerase complex with the polymerase cofactor of L, phosphoprotein. Hsp70 collaborates with Hsp90 to regulate biogenesis of the MuV L protein. The functions of these chaperones on the viral polymerase may be common among paramyxoviruses because the L protein of measles virus is also similarly regulated. Our data provide important insights into the molecular mechanisms of paramyxovirus polymerase maturation as well as a basis for the development of novel antiviral drugs.
Collapse
|
33
|
Im E, Yoon JB, Lee HW, Chung KC. Human Telomerase Reverse Transcriptase (hTERT) Positively Regulates 26S Proteasome Activity. J Cell Physiol 2017; 232:2083-2093. [PMID: 27648923 DOI: 10.1002/jcp.25607] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/19/2016] [Indexed: 02/02/2023]
Abstract
Human telomerase reverse transcriptase (hTERT) is the catalytic subunit of telomerase, an RNA-dependent DNA polymerase that elongates telomeric DNA. hTERT displays several extra-telomeric functions that are independent of its telomere-regulatory function, including tumor progression, and neuronal cell death regulation. In this study, we evaluated these additional hTERT non-telomeric functions. We determined that hTERT interacts with several 19S and 20S proteasome subunits. The 19S regulatory particle and 20S core particle are part of 26S proteasome complex, which plays a central role in ubiquitin-dependent proteolysis. In addition, hTERT positively regulated 26S proteasome activity independent of its enzymatic activity. Moreover, hTERT enhanced subunit interactions, which may underlie hTERT's ability of hTERT to stimulate the 26S proteasome. Furthermore, hTERT displayed cytoprotective effect against ER stress via the activation of 26S proteasome in acute myeloid leukemia cells. Our data suggest that hTERT acts as a novel chaperone to promote 26S proteasome assembly and maintenance. J. Cell. Physiol. 232: 2083-2093, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eunju Im
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Jong Bok Yoon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
34
|
Hsp90-binding immunophilin FKBP52 modulates telomerase activity by promoting the cytoplasmic retrotransport of hTERT. Biochem J 2016; 473:3517-3532. [DOI: 10.1042/bcj20160344] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/08/2016] [Indexed: 11/17/2022]
Abstract
Telomerase is a unique ribonucleoprotein enzyme that is required for continued cell proliferation. To generate catalytically active telomerase, human telomerase reverse transcriptase (hTERT) must translocate to the nucleus and assemble with the RNA component of telomerase. The molecular chaperones heat shock protein 90 (Hsp90) and p23 maintain hTERT in a conformation that enables nuclear translocation. However, the regulatory role of chaperones in nuclear transport of hTERT remains unclear. In this work, we demonstrate that immunophilin FK506-binding protein (FKBP)52 linked the hTERT–Hsp90 complex to the dynein–dynactin motor, thereby promoting the transport of hTERT to the nucleus along microtubules. FKBP52 interacted with the hTERT–Hsp90 complex through binding of the tetratricopeptide repeat domain to Hsp90 and binding of the dynamitin (Dyt) component of the dynein-associated dynactin complex to the peptidyl prolyl isomerase domain. The depletion of FKBP52 inhibited nuclear transport of hTERT, resulting in cytoplasmic accumulation. Cytoplasmic hTERT was rapidly degraded through ubiquitin (Ub)-dependent proteolysis, thereby abrogating telomerase activity. In addition, overexpression of dynamitin, which is known to dissociate the dynein–dynactin motor from its cargoes, reduced telomerase activity. Collectively, these results provide a molecular mechanism by which FKBP52 modulates telomerase activity by promoting dynein–dynactin-dependent nuclear import of hTERT.
Collapse
|
35
|
Joshi V, Amanullah A, Upadhyay A, Mishra R, Kumar A, Mishra A. A Decade of Boon or Burden: What Has the CHIP Ever Done for Cellular Protein Quality Control Mechanism Implicated in Neurodegeneration and Aging? Front Mol Neurosci 2016; 9:93. [PMID: 27757073 PMCID: PMC5047891 DOI: 10.3389/fnmol.2016.00093] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/20/2016] [Indexed: 01/13/2023] Open
Abstract
Cells regularly synthesize new proteins to replace old and abnormal proteins for normal cellular functions. Two significant protein quality control pathways inside the cellular milieu are ubiquitin proteasome system (UPS) and autophagy. Autophagy is known for bulk clearance of cytoplasmic aggregated proteins, whereas the specificity of protein degradation by UPS comes from E3 ubiquitin ligases. Few E3 ubiquitin ligases, like C-terminus of Hsc70-interacting protein (CHIP) not only take part in protein quality control pathways, but also plays a key regulatory role in other cellular processes like signaling, development, DNA damage repair, immunity and aging. CHIP targets misfolded proteins for their degradation through proteasome, as well as autophagy; simultaneously, with the help of chaperones, it also regulates folding attempts for misfolded proteins. The broad range of CHIP substrates and their associations with multiple pathologies make it a key molecule to work upon and focus for future therapeutic interventions. E3 ubiquitin ligase CHIP interacts and degrades many protein inclusions formed in neurodegenerative diseases. The presence of CHIP at various nodes of cellular protein-protein interaction network presents this molecule as a potential candidate for further research. In this review, we have explored a wide range of functionality of CHIP inside cells by a detailed presentation of its co-chaperone, E3 and E4 enzyme like functions, with central focus on its protein quality control roles in neurodegenerative diseases. We have also raised many unexplored but expected fundamental questions regarding CHIP functions, which generate hopes for its future applications in research, as well as drug discovery.
Collapse
Affiliation(s)
- Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Ribhav Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| | - Amit Kumar
- Centre for Biosciences and Biomedical Engineering, Indian Institute of Technology Indore Madhya Pradesh, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur Rajasthan, India
| |
Collapse
|
36
|
MacNeil DE, Bensoussan HJ, Autexier C. Telomerase Regulation from Beginning to the End. Genes (Basel) 2016; 7:genes7090064. [PMID: 27649246 PMCID: PMC5042394 DOI: 10.3390/genes7090064] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022] Open
Abstract
The vast body of literature regarding human telomere maintenance is a true testament to the importance of understanding telomere regulation in both normal and diseased states. In this review, our goal was simple: tell the telomerase story from the biogenesis of its parts to its maturity as a complex and function at its site of action, emphasizing new developments and how they contribute to the foundational knowledge of telomerase and telomere biology.
Collapse
Affiliation(s)
- Deanna Elise MacNeil
- Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte Ste-Catherine Road, Montréal, QC H3T 1E2, Canada.
- Room M-29, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montréal, QC H3A 0C7, Canada.
| | - Hélène Jeanne Bensoussan
- Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte Ste-Catherine Road, Montréal, QC H3T 1E2, Canada.
- Room M-29, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montréal, QC H3A 0C7, Canada.
| | - Chantal Autexier
- Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte Ste-Catherine Road, Montréal, QC H3T 1E2, Canada.
- Room M-29, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montréal, QC H3A 0C7, Canada.
- Department of Experimental Medicine, McGill University, 1110 Pins Avenue West, Room 101, Montréal, QC H3A 1A3, Canada.
| |
Collapse
|
37
|
Lagadari M, Zgajnar NR, Gallo LI, Galigniana MD. Hsp90-binding immunophilin FKBP51 forms complexes with hTERT enhancing telomerase activity. Mol Oncol 2016; 10:1086-98. [PMID: 27233944 PMCID: PMC5423183 DOI: 10.1016/j.molonc.2016.05.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/25/2016] [Accepted: 05/09/2016] [Indexed: 11/25/2022] Open
Abstract
FK506-binding proteins are members of the immunophilin family of proteins. Those immunophilins associated to the 90-kDa-heat-shock protein, Hsp90, have been proposed as potential modulators of signalling cascade factors chaperoned by Hsp90. FKBP51 and FKBP52 are the best characterized Hsp90-bound immunophilins first described associated to steroid-receptors. The reverse transcriptase subunit of telomerase, hTERT, is also an Hsp90 client-protein and is highly expressed in cancer cells, where it is required to compensate the loss of telomeric DNA after each successive cell division. Because FKBP51 is also a highly expressed protein in cancer tissues, we analyzed its potential association with hTERT·Hsp90 complexes and its possible biological role. In this study it is demonstrated that both immunophilins, FKBP51 and FKBP52, co-immunoprecipitate with hTERT. The Hsp90 inhibitor radicicol disrupts the heterocomplex and favors the partial cytoplasmic relocalization of hTERT in similar manner as the overexpression of the TPR-domain peptide of the immunophilin. While confocal microscopy images show that FKBP51 is primarily localized in mitochondria and hTERT is totally nuclear, upon the onset of oxidative stress, FKBP51 (but not FKBP52) becomes mostly nuclear colocalizing with hTERT, and longer exposure times to peroxide favors hTERT export to mitochondria. Importantly, telomerase activity of hTERT is significantly enhanced by FKBP51. These observations support the emerging role assigned to FKBP51 as antiapoptotic factor in cancer development and progression, and describe for the first time the potential role of this immunophilin favoring the clonal expansion by enhancing telomerase activity.
Collapse
Affiliation(s)
- Mariana Lagadari
- Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, C1428ADN, Argentina
| | - Nadia R Zgajnar
- Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, C1428ADN, Argentina
| | - Luciana I Gallo
- Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET) & Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Mario D Galigniana
- Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, C1428ADN, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina.
| |
Collapse
|
38
|
Tingting Q, Jiao W, Qingfeng W, Yancheng L, Shijun YU, Zhaoqi W, Dongmei S, ShiLong W. CHIP involves in non-small cell lung cancer prognosis through VEGF pathway. Biomed Pharmacother 2016; 83:271-276. [PMID: 27392029 DOI: 10.1016/j.biopha.2016.06.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 05/21/2016] [Accepted: 06/09/2016] [Indexed: 11/30/2022] Open
Abstract
AIM CHIP (c-terminal Hsp70-interacting protein) is an E3 ligase playing vital roles in various cancers. The VEGF pathway has become an important therapeutic target in non-small cell lung cancer (NSCLC). However, little is known about the role of CHIP and the relationship between CHIP and VEGF-VEGFR2 (VEGF receptor 2) pathway in NSCLC. In this study we aimed to investigate the clinical function of CHIP in NSCLC and explore the relevant regulatory mechanism. METHODS QRT-PCR was performed to detect CHIP expression in NSCLC tissues. The association of CHIP expression and clinical parameters was analyzed using the Chi-square test. Kaplan- Meier and Cox analyses were performed to identify the role of CHIP in the prognosis of NSCLC patients. ELISA test was used to detect the VEGF secretion of NSCLC cells and western blot were used to detected the protein expression of VEGFR2 in NSCLC cells. RESULTS and the results revealed that CHIP expression was decreased in NSCLC tissues and significantly correlated with clinical stages, lymph node metastasis and distant metastasis (P<0.05). Moreover, Kaplan-Meier and Cox regression analyses showed that patients with negative expression of CHIP had a shorter survival time and CHIP could be an independent prognostic biomarker. In addition, ELISA tests showed that CHIP negatively regulated the secretion level of VEGF. Furthermore, western blot assay indicated that the VEGFR2 protein level was reduced after CHIP over-expression. CONCLUSIONS Taken together, our findings demonstrate for the first time that CHIP may serve as a promising prognostic biomarker for NSCLC patients and it may be involved in NSCLC angiogenesis through regulating VEGF secretion and expression of VEGFR2.
Collapse
Affiliation(s)
- Qian Tingting
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China.
| | - Wang Jiao
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - Wang Qingfeng
- College of Grain Science and Technology, Shenyang Normal University, Shenyang, China
| | - Liu Yancheng
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, PR China
| | - Y U Shijun
- Chuzhou University, School of Biology and Food Engineering, Chuzhou, Anhui 239000, PR China
| | - Wang Zhaoqi
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - Sun Dongmei
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - Wang ShiLong
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| |
Collapse
|
39
|
Hong J, Lee JH, Chung IK. Telomerase activates transcription of cyclin D1 gene through an interaction with NOL1. J Cell Sci 2016; 129:1566-79. [PMID: 26906424 DOI: 10.1242/jcs.181040] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/15/2016] [Indexed: 12/28/2022] Open
Abstract
Telomerase is a ribonucleoprotein enzyme that is required for the maintenance of telomere repeats. Although overexpression of telomerase in normal human somatic cells is sufficient to overcome replicative senescence, the ability of telomerase to promote tumorigenesis requires additional activities that are independent of its role in telomere extension. Here, we identify proliferation-associated nucleolar antigen 120 (NOL1, also known as NOP2) as a telomerase RNA component (TERC)-binding protein that is found in association with catalytically active telomerase. Although NOL1 is highly expressed in the majority of human tumor cells, the molecular mechanism by which NOL1 contributes to tumorigenesis remained unclear. We show that NOL1 binds to the T-cell factor (TCF)-binding element of the cyclin D1 promoter and activates its transcription. Interestingly, telomerase is also recruited to the cyclin D1 promoter in a TERC-dependent manner through the interaction with NOL1, further enhancing transcription of the cyclin D1 gene. Depletion of NOL1 suppresses cyclin D1 promoter activity, thereby leading to induction of growth arrest and altered cell cycle distributions. Collectively, our findings suggest that NOL1 represents a new route by which telomerase activates transcription of cyclin D1 gene, thus maintaining cell proliferation capacity.
Collapse
Affiliation(s)
- Juyeong Hong
- Department of Integrated Omics for Biomedical Science, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Ji Hoon Lee
- Department of Integrated Omics for Biomedical Science, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - In Kwon Chung
- Department of Integrated Omics for Biomedical Science, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
40
|
Qing H, Aono J, Findeisen HM, Jones KL, Heywood EB, Bruemmer D. Differential Regulation of Telomerase Reverse Transcriptase Promoter Activation and Protein Degradation by Histone Deacetylase Inhibition. J Cell Physiol 2015; 231:1276-82. [PMID: 26505494 DOI: 10.1002/jcp.25226] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 12/17/2022]
Abstract
Telomerase reverse transcriptase (TERT) maintains telomeres and is rate limiting for replicative life span. While most somatic tissues silence TERT transcription resulting in telomere shortening, cells derived from cancer or cardiovascular diseases express TERT and activate telomerase. In the present study, we demonstrate that histone deacetylase (HDAC) inhibition induces TERT transcription and promoter activation. At the protein level in contrast, HDAC inhibition decreases TERT protein abundance through enhanced degradation, which decreases telomerase activity and induces senescence. Finally, we demonstrate that HDAC inhibition decreases TERT expression during vascular remodeling in vivo. These data illustrate a differential regulation of TERT transcription and protein stability by HDAC inhibition and suggest that TERT may constitute an important target for the anti-proliferative efficacy of HDAC inhibitors.
Collapse
Affiliation(s)
- Hua Qing
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky.,Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky.,Department of Endocrinology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Aono
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky
| | - Hannes M Findeisen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky
| | - Karrie L Jones
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky
| | - Elizabeth B Heywood
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky
| | - Dennis Bruemmer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky.,Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
41
|
Lee JH, Jeong SA, Khadka P, Hong J, Chung IK. Involvement of SRSF11 in cell cycle-specific recruitment of telomerase to telomeres at nuclear speckles. Nucleic Acids Res 2015; 43:8435-51. [PMID: 26286192 PMCID: PMC4787792 DOI: 10.1093/nar/gkv844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 08/09/2015] [Indexed: 01/13/2023] Open
Abstract
Telomerase, a unique ribonucleoprotein complex that contains the telomerase reverse transcriptase (TERT), the telomerase RNA component (TERC) and the TERC-binding protein dyskerin, is required for continued cell proliferation in stem cells and cancer cells. Here we identify SRSF11 as a novel TERC-binding protein that localizes to nuclear speckles, subnuclear structures that are enriched in pre-messenger RNA splicing factors. SRSF11 associates with active telomerase enzyme through an interaction with TERC and directs it to nuclear speckles specifically during S phase of the cell cycle. On the other hand, a subset of telomeres is shown to be constitutively present at nuclear speckles irrespective of cell cycle phase, suggesting that nuclear speckles could be the nuclear sites for telomerase recruitment to telomeres. SRSF11 also associates with telomeres through an interaction with TRF2, which facilitates translocation of telomerase to telomeres. Depletion of SRSF11 prevents telomerase from associating with nuclear speckles and disrupts telomerase recruitment to telomeres, thereby abrogating telomere elongation by telomerase. These findings suggest that SRSF11 acts as a nuclear speckle-targeting factor that is essential for telomerase association with telomeres through the interactions with TERC and TRF2, and provides a potential target for modulating telomerase activity in cancer.
Collapse
Affiliation(s)
- Ji Hoon Lee
- Departments of Systems Biology and Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| | - Sun Ah Jeong
- Departments of Systems Biology and Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| | - Prabhat Khadka
- Departments of Systems Biology and Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| | - Juyeong Hong
- Departments of Systems Biology and Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| | - In Kwon Chung
- Departments of Systems Biology and Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
42
|
Singh AK, Pati U. CHIP stabilizes amyloid precursor protein via proteasomal degradation and p53-mediated trans-repression of β-secretase. Aging Cell 2015; 14:595-604. [PMID: 25773675 PMCID: PMC4531073 DOI: 10.1111/acel.12335] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2015] [Indexed: 12/22/2022] Open
Abstract
In patient with Alzheimer’s disease (AD), deposition of amyloid-beta Aβ, a proteolytic cleavage of amyloid precursor protein (APP) by β-secretase/BACE1, forms senile plaque in the brain. BACE1 activation is caused due to oxidative stresses and dysfunction of ubiquitin–proteasome system (UPS), which is linked to p53 inactivation. As partial suppression of BACE1 attenuates Aβ generation and AD-related pathology, it might be an ideal target for AD treatment. We have shown that both in neurons and in HEK-APP cells, BACE1 is a new substrate of E3-ligase CHIP and an inverse relation exists between CHIP and BACE1 level. CHIP inhibits ectopic BACE1 level by promoting its ubiquitination and proteasomal degradation, thus reducing APP processing; it stabilizes APP in neurons, thus reducing Aβ. CHIPUbox domain physically interacts with BACE1; however, both U-box and TPR domain are essential for ubiquitination and degradation of BACE1. Further, BACE1 is a downstream target of p53 and overexpression of p53 decreases BACE1 level. In HEK-APP cells, CHIP is shown to negatively regulate BACE1 promoter through stabilization of p53’s DNA-binding conformation and its binding upon 5′ UTR element (+127 to +150). We have thus discovered that CHIP regulates p53-mediated trans-repression of BACE1 at both transcriptional and post-translational level. We propose that a CHIP–BACE1–p53 feedback loop might control APP stabilization, which could further be utilized for new therapeutic intervention in AD.
Collapse
Affiliation(s)
- Amir Kumar Singh
- School of Biotechnology Jawaharlal Nehru University New Delhi 110067 India
| | - Uttam Pati
- School of Biotechnology Jawaharlal Nehru University New Delhi 110067 India
| |
Collapse
|
43
|
Huang Y, Sun L, Liu N, Wei Q, Jiang L, Tong X, Ye X. Polo-like Kinase 1 (Plk1) Up-regulates Telomerase Activity by Affecting Human Telomerase Reverse Transcriptase (hTERT) Stability. J Biol Chem 2015; 290:18865-73. [PMID: 26070557 PMCID: PMC4513140 DOI: 10.1074/jbc.m114.635375] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 06/11/2015] [Indexed: 12/13/2022] Open
Abstract
Maintenance of telomere is regulated by active telomerase complex, including telomerase holoenzyme and its associated proteins. The activity of telomerase is precisely controlled in cells, and its dysregulation is one of the hallmarks of cancer. The telomerase catalytic subunit human telomerase reverse transcriptase (hTERT) plays a central role for telomerase activity. In this study, we indentified that Polo-like kinase 1 (Plk1) is a novel telomerase-associated protein. Plk1 can interact with hTERT independently of its kinase activity. More importantly, we found that Plk1 is associated with active telomerase complex. In addition, we demonstrated that knockdown of Plk1 caused the reduction of telomerase activity, whereas overexpression of Plk1 increased telomerase activity. Further analysis showed that overexpression of Plk1 led to a significant increase of hTERT protein by prolonging its half-life but did not affect the level of hTERT mRNA. Furthermore, we found that Plk1 enhanced the chromatin loading of hTERT and inhibited its ubiquitination. This implied that Plk1 affected hTERT stability by inhibiting its ubiquitin-mediated degradation. Collectively, these observations suggested that Plk1 is a positive modulator of telomerase by enhancing the stability of hTERT.
Collapse
Affiliation(s)
- Yan Huang
- From the CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS) and the University of Chinese Academy of Sciences, Beijing 100101, China
| | - Liping Sun
- From the CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS) and the University of Chinese Academy of Sciences, Beijing 100101, China
| | - Ningning Liu
- From the CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS) and the University of Chinese Academy of Sciences, Beijing 100101, China
| | - Qian Wei
- From the CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS) and the University of Chinese Academy of Sciences, Beijing 100101, China
| | - Liangzhen Jiang
- From the CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS) and the University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaomei Tong
- From the CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS) and
| | - Xin Ye
- From the CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS) and
| |
Collapse
|
44
|
Jeong SA, Kim K, Lee JH, Cha JS, Khadka P, Cho HS, Chung IK. Akt-mediated phosphorylation increases the binding affinity of hTERT for importin α to promote nuclear translocation. J Cell Sci 2015; 128:2287-301. [DOI: 10.1242/jcs.166132] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 04/10/2015] [Indexed: 01/09/2023] Open
Abstract
ABSTRACT
Telomeres are essential for chromosome integrity and protection, and their maintenance requires the ribonucleoprotein enzyme telomerase. Previously, we have shown that human telomerase reverse transcriptase (hTERT) contains a bipartite nuclear localization signal (NLS; residues 222–240) that is responsible for nuclear import, and that Akt-mediated phosphorylation of residue S227 is important for efficient nuclear import of hTERT. Here, we show that hTERT binds to importin-α proteins through the bipartite NLS and that this heterodimer then forms a complex with importin-β proteins to interact with the nuclear pore complex. Depletion of individual importin-α proteins results in a failure of hTERT nuclear import, and the resulting cytoplasmic hTERT is degraded by ubiquitin-dependent proteolysis. Crystallographic analysis reveals that the bipartite NLS interacts with both the major and minor sites of importin-α proteins. We also show that Akt-mediated phosphorylation of S227 increases the binding affinity for importin-α proteins and promotes nuclear import of hTERT, thereby resulting in increased telomerase activity. These data provide details of a binding mechanism that enables hTERT to interact with the nuclear import receptors and of the control of the dynamic nuclear transport of hTERT through phosphorylation.
Collapse
Affiliation(s)
- Sun Ah Jeong
- Department of Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| | - Kuglae Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Ji Hoon Lee
- Department of Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| | - Jeong Seok Cha
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Prabhat Khadka
- Department of Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
| | - Hyun-Soo Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - In Kwon Chung
- Department of Integrated Omics for Biomedical Science, Yonsei University, Seoul 120-749, Korea
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
45
|
Cao Z, Xu J, Huang H, Shen P, You L, Zhou L, Zheng L, Zhang T, Zhao Y. MiR-1178 promotes the proliferation, G1/S transition, migration and invasion of pancreatic cancer cells by targeting CHIP. PLoS One 2015; 10:e0116934. [PMID: 25635996 PMCID: PMC4312055 DOI: 10.1371/journal.pone.0116934] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 12/16/2014] [Indexed: 12/21/2022] Open
Abstract
CHIP, a co-chaperone protein that interacts with Hsc/Hsp70, has been shown to be under-expressed in pancreatic cancer cells and has demonstrated a potential tumor suppressor property. Nevertheless, the underlying mechanisms of CHIP regulation in pancreatic cancer cells remain unknown. In this study, we found that miR-1178 decreased the translation of the CHIP protein by targeting the 3′-UTR region. We observed that over-expression of miR-1178 facilitated the proliferation, G1/S transition, migration and invasion of pancreatic cancer cells. Conversely, the inhibition of miR-1178 expression significantly suppressed these phenotypes. Furthermore, CHIP over-expression abrogated miR-1178-induced cell proliferation and invasion. Our data suggest that miR-1178 acts as an oncomiR in pancreatic cancer cells by inhibiting CHIP expression.
Collapse
Affiliation(s)
- Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Shen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail: (TZ); (YZ)
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail: (TZ); (YZ)
| |
Collapse
|
46
|
Paul I, Ghosh MK. A CHIPotle in physiology and disease. Int J Biochem Cell Biol 2014; 58:37-52. [PMID: 25448416 DOI: 10.1016/j.biocel.2014.10.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/21/2014] [Accepted: 10/25/2014] [Indexed: 01/06/2023]
Abstract
The carboxy-terminus of Hsc70 interacting protein (CHIP) is known to function as a chaperone associated E3 ligase for several proteins and regulates a variety of physiological processes. Being a connecting link between molecular chaperones and 26S proteasomes, it is widely regarded as the central player in the cellular protein quality control system. Recent analyses have provided new insights on the biochemical and functional dynamics of CHIP. In this review article, we give a comprehensive account of our current knowledge on the biology of CHIP, which apart from shedding light on fundamental biological questions promises to provide a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Indranil Paul
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research - Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja S.C. Mullick Road, Kolkata 700032, India
| | - Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research - Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja S.C. Mullick Road, Kolkata 700032, India.
| |
Collapse
|
47
|
Katsogiannou M, Andrieu C, Baylot V, Baudot A, Dusetti NJ, Gayet O, Finetti P, Garrido C, Birnbaum D, Bertucci F, Brun C, Rocchi P. The functional landscape of Hsp27 reveals new cellular processes such as DNA repair and alternative splicing and proposes novel anticancer targets. Mol Cell Proteomics 2014; 13:3585-601. [PMID: 25277244 PMCID: PMC4256507 DOI: 10.1074/mcp.m114.041228] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously, we identified the stress-induced chaperone, Hsp27, as highly overexpressed in castration-resistant prostate cancer and developed an Hsp27 inhibitor (OGX-427) currently tested in phase I/II clinical trials as a chemosensitizing agent in different cancers. To better understand the Hsp27 poorly-defined cytoprotective functions in cancers and increase the OGX-427 pharmacological safety, we established the Hsp27-protein interaction network using a yeast two-hybrid approach and identified 226 interaction partners. As an example, we showed that targeting Hsp27 interaction with TCTP, a partner protein identified in our screen increases therapy sensitivity, opening a new promising field of research for therapeutic approaches that could decrease or abolish toxicity for normal cells. Results of an in-depth bioinformatics network analysis allying the Hsp27 interaction map into the human interactome underlined the multifunctional character of this protein. We identified interactions of Hsp27 with proteins involved in eight well known functions previously related to Hsp27 and uncovered 17 potential new ones, such as DNA repair and RNA splicing. Validation of Hsp27 involvement in both processes in human prostate cancer cells supports our system biology-predicted functions and provides new insights into Hsp27 roles in cancer cells.
Collapse
Affiliation(s)
- Maria Katsogiannou
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Claudia Andrieu
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Virginie Baylot
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Anaïs Baudot
- ¶Aix-Marseille Université, F-13284, Marseille, France; **Institut de Mathématiques de Marseille, CNRS UMR7373, Marseille, F-13009, France
| | - Nelson J Dusetti
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Odile Gayet
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Pascal Finetti
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France
| | - Carmen Garrido
- ‡‡Inserm U866, Faculty of Medicine, 21000 Dijon, France; §§CGFL Dijon, France
| | - Daniel Birnbaum
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - François Bertucci
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Christine Brun
- ¶Aix-Marseille Université, F-13284, Marseille, France; ¶¶TAGC Inserm U1090, Marseille, F-13009, France; ‖‖CNRS, France
| | - Palma Rocchi
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France;
| |
Collapse
|
48
|
The putative tumor suppressor C53 interacts with the human telomerase reverse transcriptase hTERT and regulates telomerase activity. CHINESE SCIENCE BULLETIN-CHINESE 2014. [DOI: 10.1007/s11434-014-0184-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
49
|
Paul I, Bhattacharya S, Chatterjee A, Ghosh MK. Current Understanding on EGFR and Wnt/β-Catenin Signaling in Glioma and Their Possible Crosstalk. Genes Cancer 2014; 4:427-46. [PMID: 24386505 DOI: 10.1177/1947601913503341] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/31/2013] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiformes (GBMs) are extensively heterogeneous at both cellular and molecular levels. Current therapeutic strategies include targeting of key signaling molecules using pharmacological inhibitors in combination with genotoxic agents such as temozolomide. In spite of all efforts, the prognosis of glioma patients remains dismal. Therefore, a proper understanding of individual molecular pathways responsible for the progression of GBM is necessary. The epidermal growth factor receptor (EGFR) pathway is probably the most significant signaling pathway clinically implicated in glioma. Not surprisingly, anti-EGFR therapies mostly prevail for therapeutic purposes. The Wnt/β-catenin pathway is well implicated in multiple tumors; however, its role in glioma has only recently started to emerge. We give a concise account of the current understanding of the role of both these pathways in glioma. Last, taking evidences from a limited literature, we outline a number of points where these pathways intersect each other and put forward the possibility of combinatorially targeting them for treatment of glioma.
Collapse
Affiliation(s)
- Indranil Paul
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Seemana Bhattacharya
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Anirban Chatterjee
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mrinal K Ghosh
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
50
|
Sun C, Li HL, Shi ML, Liu QH, Bai J, Zheng JN. Diverse roles of C-terminal Hsp70-interacting protein (CHIP) in tumorigenesis. J Cancer Res Clin Oncol 2014; 140:189-97. [PMID: 24370685 DOI: 10.1007/s00432-013-1571-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/12/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND The carboxyl terminus of Hsp70-interacting protein (CHIP) is a member of E3 ubiquitin ligase, functioning as a link between the chaperone (heat shock protein 70/90) and proteasome systems, playing a vital role in maintaining the protein homeostasis in the cytoplasm. CHIP has been demonstrated to be involved in tumorigenesis, proliferation and invasion in several malignancies, regulating a number of oncogenic proteins. However, CHIP has also been implicated in the modulation of tumor suppressor proteins. The pathogenic mechanism of CHIP expression in human malignancy is not yet clear, and a number of studies have suggested that CHIP may have opposing roles in different cancers. Therefore, many studies have focused on the relationship between CHIP and carcinoma. METHODS A literature search focusing on regulation network, biological function and clinical significance of CHIP in connection with its role in cancer development was performed on the MEDLINE databases. RESULTS AND CONCLUSIONS CHIP may be a potential diagnostic biomarker and therapeutic target for human cancer, and may play different roles in different human cancers. This inconsistence might be induced by the diversity of CHIP downstream targeting proteins. Therefore, the phenotypes determined by CHIP should be dependent on the function of its specific targets in a specific type of cancer cells. Whether CHIP contributes to tumor progression or suppression in various human cancers remains unclear, suggesting the necessity of further extensive investigation of its role in tumorigenesis.
Collapse
Affiliation(s)
- Chao Sun
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
| | | | | | | | | | | |
Collapse
|