1
|
Miao B, Zhang C, Stroh N, Brenner L, Hufnagel K, Hoheisel JD, Bandapalli OR. Transcription factor TFE3 enhances cell cycle and cancer progression by binding to the hTERT promoter. Cancer Commun (Lond) 2021; 41:1423-1426. [PMID: 34523267 PMCID: PMC8696226 DOI: 10.1002/cac2.12216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/19/2021] [Accepted: 09/03/2021] [Indexed: 11/18/2022] Open
Affiliation(s)
- Beiping Miao
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
- Medical Faculty HeidelbergHeidelberg UniversityHeidelbergBaden‐Württemberg69120Germany
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
| | - Chaoyang Zhang
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
- Medical Faculty HeidelbergHeidelberg UniversityHeidelbergBaden‐Württemberg69120Germany
| | - Nadine Stroh
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
| | - Lukas Brenner
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
| | - Katrin Hufnagel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
| | - Jörg D. Hoheisel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
| | - Obul Reddy Bandapalli
- Medical Faculty HeidelbergHeidelberg UniversityHeidelbergBaden‐Württemberg69120Germany
- Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ)HeidelbergBaden‐Württemberg69120Germany
| |
Collapse
|
2
|
Rashid M, Zadeh LR, Baradaran B, Molavi O, Ghesmati Z, Sabzichi M, Ramezani F. Up-down regulation of HIF-1α in cancer progression. Gene 2021; 798:145796. [PMID: 34175393 DOI: 10.1016/j.gene.2021.145796] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/25/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022]
Abstract
Hypoxia induicible factor-1 alpha (HIF-1α) is a key transcription factor in cancer progression and target therapy in cancer. HIF-1α acts differently depending on presence or absence of Oxygen. In an oxygen-immersed environment, HIF-1α completely deactivated and destroyed by the ubiquitin proteasome pathway (UPP). In contrast, in the oxygen-free environment, it escapes destruction and enters to the nucleus of cells then upregulates many genes involved in cancer progression. Overexpressed HIF-1α and downstream genes support cancer progression through various mechanisms including angiogenesis, proliferation and survival of cells, metabolism reprogramming, invasion and metastasis, cancer stem cell maintenance, induction of genetic instability, and treatment resistance. HIF-1α can be provoked by signaling pathways unrelated to hypoxia during cancer progression. Therefore, cancer development and progression can be modulated by targeting HIF-1α and its downstream signaling molecules. In this regard, HIF-1α inhibitors which are categorized into the agents that regulate HIF-1α in gene, mRNA and protein levels used as an efficient way in cancer treatment. Also, HIF-1α expression can be negatively affected by the agents suppressing the activation of mTOR, PI3k/Akt and MAPK pathways.
Collapse
Affiliation(s)
- Mohsen Rashid
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Rostami Zadeh
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Ghesmati
- Department of Medical Biotechnology, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sabzichi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Fatemeh Ramezani
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Plyasova AA, Zhdanov DD. Alternative Splicing of Human Telomerase Reverse Transcriptase (hTERT) and Its Implications in Physiological and Pathological Processes. Biomedicines 2021; 9:526. [PMID: 34065134 PMCID: PMC8150890 DOI: 10.3390/biomedicines9050526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
Alternative splicing (AS) of human telomerase catalytic subunit (hTERT, human telomerase reverse transcriptase) pre-mRNA strongly regulates telomerase activity. Several proteins can regulate AS in a cell type-specific manner and determine the functions of cells. In addition to being involved in telomerase activity regulation, AS provides cells with different splice variants that may have alternative biological activities. The modulation of telomerase activity through the induction of hTERT AS is involved in the development of different cancer types and embryos, and the differentiation of stem cells. Regulatory T cells may suppress the proliferation of target human and murine T and B lymphocytes and NK cells in a contact-independent manner involving activation of TERT AS. This review focuses on the mechanism of regulation of hTERT pre-mRNA AS and the involvement of splice variants in physiological and pathological processes.
Collapse
Affiliation(s)
| | - Dmitry D. Zhdanov
- Institute of Biomedical Chemistry, Pogodinskaya st 10/8, 119121 Moscow, Russia;
| |
Collapse
|
4
|
Natua S, Ashok C, Shukla S. Hypoxia-induced alternative splicing in human diseases: the pledge, the turn, and the prestige. Cell Mol Life Sci 2021; 78:2729-2747. [PMID: 33386889 PMCID: PMC11072330 DOI: 10.1007/s00018-020-03727-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 12/30/2022]
Abstract
Maintenance of oxygen homeostasis is an indispensable criterion for the existence of multicellular life-forms. Disruption of this homeostasis due to inadequate oxygenation of the respiring tissues leads to pathological hypoxia, which acts as a significant stressor in several pathophysiological conditions including cancer, cardiovascular defects, bacterial infections, and neurological disorders. Consequently, the hypoxic tissues develop necessary adaptations both at the tissue and cellular level. The cellular adaptations involve a dramatic alteration in gene expression, post-transcriptional and post-translational modification of gene products, bioenergetics, and metabolism. Among the key responses to oxygen-deprivation is the skewing of cellular alternative splicing program. Herein, we discuss the current concepts of oxygen tension-dependent alternative splicing relevant to various pathophysiological conditions. Following a brief description of cellular response to hypoxia and the pre-mRNA splicing mechanism, we outline the impressive number of hypoxia-elicited alternative splicing events associated with maladies like cancer, cardiovascular diseases, and neurological disorders. Furthermore, we discuss how manipulation of hypoxia-induced alternative splicing may pose promising strategies for novel translational diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Subhashis Natua
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, 462066, Madhya Pradesh, India
| | - Cheemala Ashok
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, 462066, Madhya Pradesh, India
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, 462066, Madhya Pradesh, India.
| |
Collapse
|
5
|
Song H, Chen X, Jiao Q, Qiu Z, Shen C, Zhang G, Sun Z, Zhang H, Luo QY. HIF-1α-Mediated Telomerase Reverse Transcriptase Activation Inducing Autophagy Through Mammalian Target of Rapamycin Promotes Papillary Thyroid Carcinoma Progression During Hypoxia Stress. Thyroid 2021; 31:233-246. [PMID: 32772829 DOI: 10.1089/thy.2020.0023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background: It is important to properly understand the molecular mechanisms of aggressive tumors among papillary thyroid carcinomas (PTCs) that are often the most indolent. Hypoxia inducible factor-1α (HIF-1α), induced by hypoxia, plays pivotal roles in the development and metastasis of the many tumors, including PTCs. Upregulation of telomerase reverse transcriptase (TERT) activity is found in highly invasive PTCs. Further, previous studies have reported that autophagy serves as a protective mechanism to facilitate PTC cell survival. We, therefore, hypothesized that there was a link between HIF-1α, TERT, and autophagy in promoting PTC progression. Methods: Immunohistochemistry staining was conducted to evaluate the expressions of HIF-1α, TERT, and autophagy marker, LC3-II, in matched PTC tumors and corresponding nontumor tissues. Two PTC cell lines (TPC-1 and BCPAP) were used in subsequent cytological function studies. Cell viability, proliferation, apoptosis, migration, and invasion were assessed during hypoxia, genetic enhancement and inhibition of TERT, and chemical and genetic inhibition of autophagy. The protein expression levels of the corresponding biomarkers were determined by Western blotting, and autophagy flow was detected. We characterized the molecular mechanism of PTC cell progression. Results: The protein expression levels of HIF-1α, TERT, and LC3-II were upregulated in PTCs and were significantly correlated with high tumor-node-metastasis stage. Further, an in vitro study indicated that HIF-1α induced by hypoxia functioned as a transcriptional activator by binding with sequences potentially located in the TERT promoter and was positively correlated with the malignant behavior of PTC cell lines. Overexpression of TERT inhibited the kinase activity of mammalian target of rapamycin (mTOR), resulting in the activation of autophagy. Functionally, TERT-induced autophagy provided a survival advantage to PTC cells during hypoxia stress. Conclusions: We identified a novel molecular mechanism involving the HIF-1α/TERT axis, which promoted PTC progression by inducing autophagy through mTOR during hypoxia stress. These findings may provide a basis for the new treatment of aggressive PTCs.
Collapse
Affiliation(s)
- Hongjun Song
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaoyue Chen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhongling Qiu
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chentian Shen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guoqiang Zhang
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhenkui Sun
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huizhen Zhang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
6
|
Gala K, Khattar E. Long non-coding RNAs at work on telomeres: Functions and implications in cancer therapy. Cancer Lett 2021; 502:120-132. [PMID: 33450357 DOI: 10.1016/j.canlet.2020.12.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/13/2020] [Accepted: 12/29/2020] [Indexed: 12/27/2022]
Abstract
Long non-coding RNAs (lncRNAs) are known to regulate various biological processes including cancer. Cancer cells possess limitless replicative potential which is attained by telomere length maintenance while normal somatic cells have a limited lifespan because their telomeres shorten with every cell division ultimately triggering replicative senescence. Two lncRNAs have been observed to play a key role in telomere length maintenance. First is the lncRNA TERC (telomerase RNA component) which functions as a template for telomeric DNA synthesis in association with telomerase reverse transcriptase (TERT) which serves as the catalytic component. Together they constitute the telomerase complex which functions as a reverse transcriptase to elongate telomeres. Second lncRNA that helps in regulating telomere length is the telomeric repeat-containing RNA (TERRA) which is transcribed from the subtelomeric region and extends to the telomeric region. TERC and TERRA exhibit important functions in cancer with implications in precision oncology. In this review, we discuss various aspects of these important lncRNAs in humans and their role in cancer along with recent advancements in their anticancer therapeutic application.
Collapse
Affiliation(s)
- Kavita Gala
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, 400056, Maharashtra, India
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, 400056, Maharashtra, India.
| |
Collapse
|
7
|
Cangelosi D, Morini M, Zanardi N, Sementa AR, Muselli M, Conte M, Garaventa A, Pfeffer U, Bosco MC, Varesio L, Eva A. Hypoxia Predicts Poor Prognosis in Neuroblastoma Patients and Associates with Biological Mechanisms Involved in Telomerase Activation and Tumor Microenvironment Reprogramming. Cancers (Basel) 2020; 12:E2343. [PMID: 32825087 PMCID: PMC7563184 DOI: 10.3390/cancers12092343] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/09/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022] Open
Abstract
The biological and clinical heterogeneity of neuroblastoma (NB) demands novel biomarkers and therapeutic targets in order to drive the most appropriate treatment for each patient. Hypoxia is a condition of low-oxygen tension occurring in poorly vascularized tumor tissues. In this study, we aimed to assess the role of hypoxia in the pathogenesis of NB and at developing a new clinically relevant hypoxia-based predictor of outcome. We analyzed the gene expression profiles of 1882 untreated NB primary tumors collected at diagnosis and belonging to four existing data sets. Analyses took advantage of machine learning methods. We identified NB-hop, a seven-gene hypoxia biomarker, as a predictor of NB patient prognosis, which is able to discriminate between two populations of patients with unfavorable or favorable outcome on a molecular basis. NB-hop retained its prognostic value in a multivariate model adjusted for established risk factors and was able to additionally stratify clinically relevant groups of patients. Tumors with an unfavorable NB-hop expression showed a significant association with telomerase activation and a hypoxic, immunosuppressive, poorly differentiated, and apoptosis-resistant tumor microenvironment. NB-hop defines a new population of NB patients with hypoxic tumors and unfavorable prognosis and it represents a critical factor for the stratification and treatment of NB patients.
Collapse
Affiliation(s)
- Davide Cangelosi
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.M.); (N.Z.); (L.V.); (A.E.)
| | - Martina Morini
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.M.); (N.Z.); (L.V.); (A.E.)
| | - Nicolò Zanardi
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.M.); (N.Z.); (L.V.); (A.E.)
| | - Angela Rita Sementa
- Laboratory of Pathology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Marco Muselli
- Institute of Electronics, Computer and Telecommunication Engineering, Italian National Research Council, 16149 Genova, Italy;
| | - Massimo Conte
- Pediatric Oncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.C.); (A.G.)
| | - Alberto Garaventa
- Pediatric Oncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.C.); (A.G.)
| | - Ulrich Pfeffer
- Integrated Oncology Therapies Department, Molecular Pathology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Maria Carla Bosco
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.M.); (N.Z.); (L.V.); (A.E.)
| | - Luigi Varesio
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.M.); (N.Z.); (L.V.); (A.E.)
| | - Alessandra Eva
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (M.M.); (N.Z.); (L.V.); (A.E.)
| |
Collapse
|
8
|
Farina AR, Cappabianca L, Sebastiano M, Zelli V, Guadagni S, Mackay AR. Hypoxia-induced alternative splicing: the 11th Hallmark of Cancer. J Exp Clin Cancer Res 2020; 39:110. [PMID: 32536347 PMCID: PMC7294618 DOI: 10.1186/s13046-020-01616-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/03/2020] [Indexed: 12/16/2022] Open
Abstract
Hypoxia-induced alternative splicing is a potent driving force in tumour pathogenesis and progression. In this review, we update currents concepts of hypoxia-induced alternative splicing and how it influences tumour biology. Following brief descriptions of tumour-associated hypoxia and the pre-mRNA splicing process, we review the many ways hypoxia regulates alternative splicing and how hypoxia-induced alternative splicing impacts each individual hallmark of cancer. Hypoxia-induced alternative splicing integrates chemical and cellular tumour microenvironments, underpins continuous adaptation of the tumour cellular microenvironment responsible for metastatic progression and plays clear roles in oncogene activation and autonomous tumour growth, tumor suppressor inactivation, tumour cell immortalization, angiogenesis, tumour cell evasion of programmed cell death and the anti-tumour immune response, a tumour-promoting inflammatory response, adaptive metabolic re-programming, epithelial to mesenchymal transition, invasion and genetic instability, all of which combine to promote metastatic disease. The impressive number of hypoxia-induced alternative spliced protein isoforms that characterize tumour progression, classifies hypoxia-induced alternative splicing as the 11th hallmark of cancer, and offers a fertile source of potential diagnostic/prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Lucia Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Stefano Guadagni
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
9
|
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: 16] [Impact Index Per Article: 2.7] [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
|
10
|
Polonis K, Sompalli S, Becari C, Xie J, Covassin N, Schulte PJ, Druliner BR, Johnson RA, Narkiewicz K, Boardman LA, Singh P, Somers VK. Telomere Length and Risk of Major Adverse Cardiac Events and Cancer in Obstructive Sleep Apnea Patients. Cells 2019; 8:E381. [PMID: 31027347 PMCID: PMC6562838 DOI: 10.3390/cells8050381] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
Abstract
Telomere length (TL) is associated with cardiovascular disease (CVD) and cancer. Obstructive sleep apnea (OSA) is also linked to higher risk of CVD and cancer, and to TL. We investigated the association between TL and risk of major adverse cardiac events (MACE) and cancer in OSA patients. We studied 210 individuals undergoing sleep-related studies between 2000 and 2007. Baseline characteristics and follow-up data (available in 164 subjects) were obtained from clinic records. Incidence rates were calculated for the entire group and by OSA status. Hazard ratios were calculated to estimate effects of OSA and TL on risk of MACE and cancer. In total, 32 individuals (20%) developed MACE and/or cancer during 12.7-year follow-up. The OSA group had a higher likelihood of cancer (16.0 vs. 4.9 events per 1000 person-years, P = 0.044) but no clear evidence of an elevated incidence of MACE (10.8 vs. 4.8 events per 1000 person-years, P = 0.293) compared to the non-OSA group. There was no association between TL and MACE- (HR = 1.01, 95% CI 0.78-1.28), or cancer-risk (HR = 1.18, 95% CI 0.96-1.43). Our study warrants further investigation of any modulating effect of OSA on TL and the risk of MACE and cancer.
Collapse
Affiliation(s)
- Katarzyna Polonis
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
- Department of Hypertension and Diabetology, Faculty of Medicine, Medical University of Gdansk, 80 210 Gdansk, Poland.
| | - Sreeja Sompalli
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| | - Christiane Becari
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, Ribeirão Preto-SP 14049-900, Brazil.
| | - Jiang Xie
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| | - Naima Covassin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| | - Phillip J Schulte
- Division of Biomedical Statistics and Informatics, Mayo Clinic, MN55 905, USA.
| | - Brooke R Druliner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| | - Ruth A Johnson
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Faculty of Medicine, Medical University of Gdansk, 80 210 Gdansk, Poland.
| | - Lisa A Boardman
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| | - Prachi Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55 905, USA.
| |
Collapse
|
11
|
de Punder K, Heim C, Wadhwa PD, Entringer S. Stress and immunosenescence: The role of telomerase. Psychoneuroendocrinology 2019; 101:87-100. [PMID: 30445409 PMCID: PMC6458519 DOI: 10.1016/j.psyneuen.2018.10.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/27/2018] [Accepted: 10/22/2018] [Indexed: 01/04/2023]
Abstract
Chronic stress is associated with the accelerated aging of the immune system and represents a potent risk factor for the development and progression of a wide range of physical and mental disorders. The elucidation of molecular pathways and mechanisms underlying the link between stress and cellular aging is an area of considerable interest and investigation. In this context, telomere biology has emerged as a particularly attractive candidate mechanism. Several studies have linked immune cell telomere length with stress-related conditions and states, and also with several physical and mental disorders. Because the cellular reverse transcriptase enzyme telomerase is the primary regulator of telomere length (by adding telomeric DNA to telomeres and thereby attenuating telomere shortening), the understanding of its regulation and regulatory functions constitutes a prime target for developing strategies to prevent, attenuate or reverse the adverse consequences of immune system aging (immunosenescence). In this review we provide an overview of the mechanistic pathways linking telomerase with stress and cellular aging, with an emphasis on the immune system. We summarize and synthesize the current state of the literature on immune cell telomerase in different stress- and aging-related disease states and provide recommendations for future research directions.
Collapse
Affiliation(s)
- Karin de Punder
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany.
| | - Christine Heim
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany; Department of Biobehavioral Health, College of Health and Human Development, Pennsylvania State University, USA
| | - Pathik D Wadhwa
- Department of Psychiatry & Human Behavior, University of California, Irvine, School of Medicine, Irvine, CA, USA; Department of Obstetrics & Gynecology, University of California, Irvine, School of Medicine, Irvine, CA, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, CA, USA; Department of Epidemiology, University of California, Irvine, School of Medicine, Irvine, CA, USA
| | - Sonja Entringer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, CA, USA; Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|
12
|
Minassian LM, Cotechini T, Huitema E, Graham CH. Hypoxia-Induced Resistance to Chemotherapy in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:123-139. [PMID: 31201721 DOI: 10.1007/978-3-030-12734-3_9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A major barrier to the successful management of cancer is the development of resistance to therapy. Chemotherapy resistance can either be an intrinsic property of malignant cells developed prior to therapy, or acquired following exposure to anti-cancer drugs. Given the impact of drug resistance to the overall poor survival of cancer patients, there is an urgent need to better understand the molecular pathways regulating this malignant phenotype. In this chapter we describe some of the molecular pathways that contribute to drug resistance in cancer, the role of a microenvironment deficient in oxygen (hypoxia) in malignant progression, and how hypoxia can be a significant factor in the development of drug resistance. We conclude by proposing potential therapeutic approaches that take advantage of a hypoxic microenvironment to chemosensitize therapy-resistant tumours.
Collapse
Affiliation(s)
- Lori M Minassian
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Erin Huitema
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
13
|
Polonis K, Somers VK, Becari C, Covassin N, Schulte PJ, Druliner BR, Johnson RA, Narkiewicz K, Boardman LA, Singh P. Moderate-to-severe obstructive sleep apnea is associated with telomere lengthening. Am J Physiol Heart Circ Physiol 2017; 313:H1022-H1030. [PMID: 28822964 PMCID: PMC5792204 DOI: 10.1152/ajpheart.00197.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/27/2017] [Accepted: 08/10/2017] [Indexed: 12/15/2022]
Abstract
Obstructive sleep apnea (OSA) is associated with cardiometabolic diseases. Telomere shortening is linked to hypertension, diabetes mellitus, and cardiovascular diseases. Because these conditions are highly prevalent in OSA, we hypothesized that telomere length (TL) would be reduced in OSA patients. We identified 106 OSA and 104 non-OSA subjects who underwent polysomnography evaluation. Quantitative PCR was used to measure telomere length in genomic DNA isolated from peripheral blood samples. The association between OSA and TL was determined using unadjusted and adjusted linear models. There was no difference in TL between the OSA and non-OSA (control) group. However, we observed a J-shaped relationship between TL and OSA severity: the longest TL in moderate-to-severe OSA [4,918 ± 230 (SD) bp] and the shortest TL in mild OSA (4,735 ± 145 bp). Mean TL in moderate-to-severe OSA was significantly longer than in the control group after adjustment for age, sex, body mass index, hypertension, dyslipidemia, and depression (β = 96.0, 95% confidence interval: 15.4-176.6, P = 0.020). In conclusion, moderate-to-severe OSA is associated with telomere lengthening. Our findings support the idea that changes in TL are not unidirectional processes, such that telomere shortening occurs with age and disease but may be prolonged in moderate-to-severe OSA.NEW & NOTEWORTHY Here, we show that moderate-to-severe obstructive sleep apnea is associated with longer telomeres, independent of age and cardiovascular risk factors, challenging the hypothesis that telomere shortening is a unidirectional process related to age/disease. A better understanding of the mechanisms underlying telomere dynamics may identify targets for therapeutic intervention in cardiovascular aging/other chronic diseases.
Collapse
Affiliation(s)
- Katarzyna Polonis
- 1Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; ,4Department of Hypertension and Diabetology, Medical University of Gdansk, Gdansk, Poland
| | - Virend K. Somers
- 1Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota;
| | - Christiane Becari
- 1Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota;
| | - Naima Covassin
- 1Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota;
| | - Phillip J. Schulte
- 2Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota;
| | - Brooke R. Druliner
- 3Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Ruth A. Johnson
- 3Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Krzysztof Narkiewicz
- 4Department of Hypertension and Diabetology, Medical University of Gdansk, Gdansk, Poland
| | - Lisa A. Boardman
- 3Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota; and
| | - Prachi Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota;
| |
Collapse
|
14
|
Gastrointestinal cancer cells treatment with bevacizumab activates a VEGF autoregulatory mechanism involving telomerase catalytic subunit hTERT via PI3K-AKT, HIF-1α and VEGF receptors. PLoS One 2017; 12:e0179202. [PMID: 28594907 PMCID: PMC5466359 DOI: 10.1371/journal.pone.0179202] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/25/2017] [Indexed: 11/29/2022] Open
Abstract
Background Targeting angiogenesis has been considered a promising treatment of choice for a large number of malignancies, including gastrointestinal cancers. Bevacizumab is an anti-vascular endothelial growth factor (anti-VEGF) being used for this purpose. However, treatment efficacy is largely questioned. Telomerase activity, responsible for cancer cell immortality, is detected in 85–95% of human cancers and is considered a potential regulator of VEGF. The aim of our study was to investigate the interrelationship between VEGF and hTERT in gastrointestinal cancers and to explore cell response to a combined inhibition of telomerase and VEGF. Methods AGS (gastric cancer), Caco-2 (colorectal cancer) and HepG2/C3A (hepatocellular carcinoma), were treated with telomerase inhibitors BIBR-1232 (10μM) and costunolide (10μM), with bevacizumab (Avastin® at 5 ng/ml or 100μg/ml) or with a combination of both types of inhibitors. VEGF and hTERT mRNA levels, and telomerase activity were detected by RT-PCR. VEGF levels were quantified by ELISA. Telomerase was knocked down using hTERT siRNA and hTERT was overexpressed in the telomerase negative cell line, Saos-2 (osteosarcoma), using constructs expressing either wild type hTERT (hTERT-WT) or dominant negative hTERT (hTERT-DN). Tube formation by HUVECs was assessed using ECMatrix™ (EMD Millipore). Results Our results showed that telomerase regulates VEGF expression and secretion through its catalytic subunit hTERT in AGS, Caco2, and HepG2/C3A, independent of its catalytic activity. Interestingly, VEGF inhibition with bevacizumab (100μg/ml) increased hTERT expression 42.3% in AGS, 94.1% in Caco2, and 52.5% in HepG2/C3A, and increased telomerase activity 30-fold in AGS, 10.3-fold in Caco2 and 8-fold in HepG2/C3A. A further investigation showed that VEGF upregulates hTERT expression in a mechanism that implicates the PI3K/AKT/mTOR pathway and HIF-1α. Moreover, bevacizumab treatment increased VEGFR1 and VEGFR2 expression in cancer cells and human umbilical vein endothelial cells (HUVECs) through hTERT. Thus, the combination of bevacizumab with telomerase inhibitors decreased VEGF expression and secretion by cancer cells, inhibited VEGFR1 and VEGFR2 upregulation, and reduced tube formation by HUVECs. Conclusions Taken together, our results suggest that bevacizumab treatment activates a VEGF autoregulatory mechanism involving hTERT and VEGF receptors and that an inhibition of this pathway could improve tumor cell response to anti-VEGF treatment.
Collapse
|
15
|
Ozturk MB, Li Y, Tergaonkar V. Current Insights to Regulation and Role of Telomerase in Human Diseases. Antioxidants (Basel) 2017; 6:antiox6010017. [PMID: 28264499 PMCID: PMC5384180 DOI: 10.3390/antiox6010017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 12/31/2022] Open
Abstract
The telomerase ribonucleoprotein complex has a pivotal role in regulating the proliferation and senescence of normal somatic cells as well as cancer cells. This complex is comprised mainly of telomerase reverse transcriptase (TERT), telomerase RNA component (TERC) and other associated proteins that function to elongate telomeres localized at the end of the chromosomes. While reactivation of telomerase is a major hallmark of most cancers, together with the synergistic activation of other oncogenic signals, deficiency in telomerase and telomeric proteins might lead to aging and senescence-associated disorders. Therefore, it is critically important to understand the canonical as well as non-canonical functions of telomerase through TERT to develop a therapeutic strategy against telomerase-related diseases. In this review, we shed light on the regulation and function of telomerase, and current therapeutic strategies against telomerase in cancer and age-related diseases.
Collapse
Affiliation(s)
- Mert Burak Ozturk
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.
| | - Yinghui Li
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
| | - Vinay Tergaonkar
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide SA 5000, Australia.
| |
Collapse
|
16
|
Khattar E, Tergaonkar V. Transcriptional Regulation of Telomerase Reverse Transcriptase (TERT) by MYC. Front Cell Dev Biol 2017; 5:1. [PMID: 28184371 PMCID: PMC5266692 DOI: 10.3389/fcell.2017.00001] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022] Open
Abstract
Telomerase elongates telomeres and is crucial for maintaining genomic stability. While stem cells and cancer cells display high telomerase activity, normal somatic cells lack telomerase activity primarily due to transcriptional repression of telomerase reverse transcriptase (TERT), the catalytic component of telomerase. Transcription factor binding, chromatin status as well as epigenetic modifications at the TERT promoter regulates TERT transcription. Myc is an important transcriptional regulator of TERT that directly controls its expression by promoter binding and associating with other transcription factors. In this review, we discuss the current understanding of the molecular mechanisms behind regulation of TERT transcription by Myc. We also discuss future perspectives in investigating the regulation of Myc at TERT promoter during cancer development.
Collapse
Affiliation(s)
- Ekta Khattar
- Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology, ASTAR Singapore, Singapore
| | - Vinay Tergaonkar
- Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology, ASTARSingapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore; Centre for Cancer Biology, University of South Australia and SA PathologyAdelaide, SA, Australia
| |
Collapse
|
17
|
Avin BA, Umbricht CB, Zeiger MA. Human telomerase reverse transcriptase regulation by DNA methylation, transcription factor binding and alternative splicing (Review). Int J Oncol 2016; 49:2199-2205. [PMID: 27779655 DOI: 10.3892/ijo.2016.3743] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 10/17/2016] [Indexed: 12/31/2022] Open
Abstract
The catalytic subunit of telomerase, human telomerase reverse transcriptase (hTERT), plays an essential role in telomere maintenance to oppose cellular senescence and, is highly regulated in normal and cancerous cells. Regulation of hTERT occurs through multiple avenues, including a unique pattern of CpG promoter methylation and alternative splicing. Promoter methylation affects the binding of transcription factors, resulting in changes in expression of the gene. In addition to expression level changes, changes in promoter binding can affect alternative splicing in a cotranscriptional manner. The alternative splicing of hTERT results in either the full length transcript which can form the active telomerase complex with hTR, or numerous inactive isoforms. Both regulation strategies are exploited in cancer to activate telomerase, however, the exact mechanism is unknown. Therefore, unraveling the link between promoter methylation status and alternative splicing for hTERT could expose yet another level of hTERT regulation. In an attempt to provide insight into the cellular control of active telomerase in cancer, this review will discuss our current perspective on CpG methylation of the hTERT promoter region, summarize the different forms of alternatively spliced variants, and examine examples of transcription factor binding that affects splicing.
Collapse
Affiliation(s)
- Brittany A Avin
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christopher B Umbricht
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Martha A Zeiger
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
Cancer-Specific Telomerase Reverse Transcriptase (TERT) Promoter Mutations: Biological and Clinical Implications. Genes (Basel) 2016; 7:genes7070038. [PMID: 27438857 PMCID: PMC4962008 DOI: 10.3390/genes7070038] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/05/2016] [Accepted: 07/11/2016] [Indexed: 12/11/2022] Open
Abstract
The accumulated evidence has pointed to a key role of telomerase in carcinogenesis. As a RNA-dependent DNA polymerase, telomerase synthesizes telomeric DNA at the end of linear chromosomes, and attenuates or prevents telomere erosion associated with cell divisions. By lengthening telomeres, telomerase extends cellular life-span or even induces immortalization. Consistent with its functional activity, telomerase is silent in most human normal somatic cells while active only in germ-line, stem and other highly proliferative cells. In contrast, telomerase activation widely occurs in human cancer and the enzymatic activity is detectable in up to 90% of malignancies. Recently, hotspot point mutations in the regulatory region of the telomerase reverse transcriptase (TERT) gene, encoding the core catalytic component of telomerase, was identified as a novel mechanism to activate telomerase in cancer. This review discusses the cancer-specific TERT promoter mutations and potential biological and clinical significances.
Collapse
|
19
|
Telomerase Activity is Downregulated Early During Human Brain Development. Genes (Basel) 2016; 7:genes7060027. [PMID: 27322326 PMCID: PMC4929426 DOI: 10.3390/genes7060027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/25/2016] [Accepted: 06/06/2016] [Indexed: 12/14/2022] Open
Abstract
Changes in hTERT splice variant expression have been proposed to facilitate the decrease of telomerase activity during fetal development in various human tissues. Here, we analyzed the expression of telomerase RNA (hTR), wild type and α-spliced hTERT in developing human fetal brain (post conception weeks, pcw, 6-19) and in young and old cortices using qPCR and correlated it to telomerase activity measured by TRAP assay. Decrease of telomerase activity occurred early during brain development and correlated strongest to decreased hTR expression. The expression of α-spliced hTERT increased between pcw 10 and 19, while that of wild type hTERT remained unchanged. Lack of expression differences between young and old cortices suggests that most changes seem to occur early during human brain development. Using in vitro differentiation of neural precursor stem cells (NPSCs) derived at pcw 6 we found a decrease in telomerase activity but no major expression changes in telomerase associated genes. Thus, they do not seem to model the mechanisms for the decrease in telomerase activity in fetal brains. Our results suggest that decreased hTR levels, as well as transient increase in α-spliced hTERT, might both contribute to downregulation of telomerase activity during early human brain development between 6 and 17 pcw.
Collapse
|
20
|
Akincilar SC, Unal B, Tergaonkar V. Reactivation of telomerase in cancer. Cell Mol Life Sci 2016; 73:1659-70. [PMID: 26846696 PMCID: PMC4805692 DOI: 10.1007/s00018-016-2146-9] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 02/06/2023]
Abstract
Activation of telomerase is a critical step in the development of about 85 % of human cancers. Levels of Tert, which encodes the reverse transcriptase subunit of telomerase, are limiting in normal somatic cells. Tert is subjected to transcriptional, post-transcriptional and epigenetic regulation, but the precise mechanism of how telomerase is re-activated in cancer cells is poorly understood. Reactivation of the Tert promoter involves multiple changes which evolve during cancer progression including mutations and chromosomal re-arrangements. Newly described non-coding mutations in the Tert promoter region of many cancer cells (19 %) in two key positions, C250T and C228T, have added another layer of complexity to telomerase reactivation. These mutations create novel consensus sequences for transcription factors which can enhance Tert expression. In this review, we will discuss gene structure and function of Tert and provide insights into the mechanisms of Tert reactivation in cancers, highlighting the contribution of recently identified Tert promoter mutations.
Collapse
Affiliation(s)
- Semih Can Akincilar
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Proteos, 61, Biopolis Drive, Singapore, 138673, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore
| | - Bilal Unal
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Proteos, 61, Biopolis Drive, Singapore, 138673, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore
| | - Vinay Tergaonkar
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Proteos, 61, Biopolis Drive, Singapore, 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore.
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia.
| |
Collapse
|
21
|
Teichroeb JH, Kim J, Betts DH. The role of telomeres and telomerase reverse transcriptase isoforms in pluripotency induction and maintenance. RNA Biol 2016; 13:707-19. [PMID: 26786236 DOI: 10.1080/15476286.2015.1134413] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Telomeres are linear guanine-rich DNA structures at the ends of chromosomes. The length of telomeric DNA is actively regulated by a number of mechanisms in highly proliferative cells such as germ cells, cancer cells, and pluripotent stem cells. Telomeric DNA is synthesized by way of the ribonucleoprotein called telomerase containing a reverse transcriptase (TERT) subunit and RNA component (TERC). TERT is highly conserved across species and ubiquitously present in their respective pluripotent cells. Recent studies have uncovered intricate associations between telomeres and the self-renewal and differentiation properties of pluripotent stem cells. Interestingly, the past decade's work indicates that the TERT subunit also has the capacity to modulate mitochondrial function, to remodel chromatin structure, and to participate in key signaling pathways such as the Wnt/β-catenin pathway. Many of these non-canonical functions do not require TERT's catalytic activity, which hints at possible functions for the extensive number of alternatively spliced TERT isoforms that are highly expressed in pluripotent stem cells. In this review, some of the established and potential routes of pluripotency induction and maintenance are highlighted from the perspectives of telomere maintenance, known TERT isoform functions and their complex regulation.
Collapse
Affiliation(s)
- Jonathan H Teichroeb
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada
| | - Joohwan Kim
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada
| | - Dean H Betts
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada.,b Children's Health Research Institute, Lawson Health Research Institute , London , Ontario , Canada
| |
Collapse
|
22
|
Yaswen P, MacKenzie KL, Keith WN, Hentosh P, Rodier F, Zhu J, Firestone GL, Matheu A, Carnero A, Bilsland A, Sundin T, Honoki K, Fujii H, Georgakilas AG, Amedei A, Amin A, Helferich B, Boosani CS, Guha G, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Niccolai E, Aquilano K, Ashraf SS, Nowsheen S, Yang X. Therapeutic targeting of replicative immortality. Semin Cancer Biol 2015; 35 Suppl:S104-S128. [PMID: 25869441 PMCID: PMC4600408 DOI: 10.1016/j.semcancer.2015.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 03/06/2015] [Accepted: 03/13/2015] [Indexed: 12/15/2022]
Abstract
One of the hallmarks of malignant cell populations is the ability to undergo continuous proliferation. This property allows clonal lineages to acquire sequential aberrations that can fuel increasingly autonomous growth, invasiveness, and therapeutic resistance. Innate cellular mechanisms have evolved to regulate replicative potential as a hedge against malignant progression. When activated in the absence of normal terminal differentiation cues, these mechanisms can result in a state of persistent cytostasis. This state, termed "senescence," can be triggered by intrinsic cellular processes such as telomere dysfunction and oncogene expression, and by exogenous factors such as DNA damaging agents or oxidative environments. Despite differences in upstream signaling, senescence often involves convergent interdependent activation of tumor suppressors p53 and p16/pRB, but can be induced, albeit with reduced sensitivity, when these suppressors are compromised. Doses of conventional genotoxic drugs required to achieve cancer cell senescence are often much lower than doses required to achieve outright cell death. Additional therapies, such as those targeting cyclin dependent kinases or components of the PI3K signaling pathway, may induce senescence specifically in cancer cells by circumventing defects in tumor suppressor pathways or exploiting cancer cells' heightened requirements for telomerase. Such treatments sufficient to induce cancer cell senescence could provide increased patient survival with fewer and less severe side effects than conventional cytotoxic regimens. This positive aspect is countered by important caveats regarding senescence reversibility, genomic instability, and paracrine effects that may increase heterogeneity and adaptive resistance of surviving cancer cells. Nevertheless, agents that effectively disrupt replicative immortality will likely be valuable components of new combinatorial approaches to cancer therapy.
Collapse
Affiliation(s)
- Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA, United States.
| | - Karen L MacKenzie
- Children's Cancer Institute Australia, Kensington, New South Wales, Australia.
| | | | | | | | - Jiyue Zhu
- Washington State University College of Pharmacy, Pullman, WA, United States.
| | | | | | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, HUVR, Consejo Superior de Investigaciones Cientificas, Universdad de Sevilla, Seville, Spain.
| | | | | | | | | | | | | | - Amr Amin
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| | | | - Gunjan Guha
- SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust, Guildford, Surrey, United Kingdom
| | | | - Asfar S Azmi
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | | | | | | | | | - S Salman Ashraf
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
23
|
Wang Z, Dabrosin C, Yin X, Fuster MM, Arreola A, Rathmell WK, Generali D, Nagaraju GP, El-Rayes B, Ribatti D, Chen YC, Honoki K, Fujii H, Georgakilas AG, Nowsheen S, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich B, Yang X, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Halicka D, Mohammed SI, Azmi AS, Bilsland A, Keith WN, Jensen LD. Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol 2015; 35 Suppl:S224-S243. [PMID: 25600295 PMCID: PMC4737670 DOI: 10.1016/j.semcancer.2015.01.001] [Citation(s) in RCA: 336] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 12/25/2014] [Accepted: 01/08/2015] [Indexed: 12/20/2022]
Abstract
Deregulation of angiogenesis--the growth of new blood vessels from an existing vasculature--is a main driving force in many severe human diseases including cancer. As such, tumor angiogenesis is important for delivering oxygen and nutrients to growing tumors, and therefore considered an essential pathologic feature of cancer, while also playing a key role in enabling other aspects of tumor pathology such as metabolic deregulation and tumor dissemination/metastasis. Recently, inhibition of tumor angiogenesis has become a clinical anti-cancer strategy in line with chemotherapy, radiotherapy and surgery, which underscore the critical importance of the angiogenic switch during early tumor development. Unfortunately the clinically approved anti-angiogenic drugs in use today are only effective in a subset of the patients, and many who initially respond develop resistance over time. Also, some of the anti-angiogenic drugs are toxic and it would be of great importance to identify alternative compounds, which could overcome these drawbacks and limitations of the currently available therapy. Finding "the most important target" may, however, prove a very challenging approach as the tumor environment is highly diverse, consisting of many different cell types, all of which may contribute to tumor angiogenesis. Furthermore, the tumor cells themselves are genetically unstable, leading to a progressive increase in the number of different angiogenic factors produced as the cancer progresses to advanced stages. As an alternative approach to targeted therapy, options to broadly interfere with angiogenic signals by a mixture of non-toxic natural compound with pleiotropic actions were viewed by this team as an opportunity to develop a complementary anti-angiogenesis treatment option. As a part of the "Halifax Project" within the "Getting to know cancer" framework, we have here, based on a thorough review of the literature, identified 10 important aspects of tumor angiogenesis and the pathological tumor vasculature which would be well suited as targets for anti-angiogenic therapy: (1) endothelial cell migration/tip cell formation, (2) structural abnormalities of tumor vessels, (3) hypoxia, (4) lymphangiogenesis, (5) elevated interstitial fluid pressure, (6) poor perfusion, (7) disrupted circadian rhythms, (8) tumor promoting inflammation, (9) tumor promoting fibroblasts and (10) tumor cell metabolism/acidosis. Following this analysis, we scrutinized the available literature on broadly acting anti-angiogenic natural products, with a focus on finding qualitative information on phytochemicals which could inhibit these targets and came up with 10 prototypical phytochemical compounds: (1) oleanolic acid, (2) tripterine, (3) silibinin, (4) curcumin, (5) epigallocatechin-gallate, (6) kaempferol, (7) melatonin, (8) enterolactone, (9) withaferin A and (10) resveratrol. We suggest that these plant-derived compounds could be combined to constitute a broader acting and more effective inhibitory cocktail at doses that would not be likely to cause excessive toxicity. All the targets and phytochemical approaches were further cross-validated against their effects on other essential tumorigenic pathways (based on the "hallmarks" of cancer) in order to discover possible synergies or potentially harmful interactions, and were found to generally also have positive involvement in/effects on these other aspects of tumor biology. The aim is that this discussion could lead to the selection of combinations of such anti-angiogenic compounds which could be used in potent anti-tumor cocktails, for enhanced therapeutic efficacy, reduced toxicity and circumvention of single-agent anti-angiogenic resistance, as well as for possible use in primary or secondary cancer prevention strategies.
Collapse
Affiliation(s)
- Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Xin Yin
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Mark M Fuster
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, San Diego, CA, USA
| | - Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Daniele Generali
- Molecular Therapy and Pharmacogenomics Unit, AO Isituti Ospitalieri di Cremona, Cremona, Italy
| | - Ganji P Nagaraju
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy; National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Yi Charlie Chen
- Department of Biology, Alderson Broaddus University, Philippi, WV, USA
| | - Kanya Honoki
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Arthroplasty and Regenerative Medicine, Nara Medical University, Nara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirate University, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirate University, United Arab Emirates
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guilford, Surrey, UK
| | | | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Asfar S Azmi
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lasse D Jensen
- Department of Medical, and Health Sciences, Linköping University, Linköping, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Debabrata K, Prasanta B, Vineet N, Anshul G, Arindam S, Satadal D. Aggressive periodontitis: An appraisal of systemic effects on its etiology-genetic aspect. J Indian Soc Periodontol 2015; 19:169-73. [PMID: 26015667 PMCID: PMC4439626 DOI: 10.4103/0972-124x.148647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 10/08/2014] [Indexed: 11/19/2022] Open
Abstract
Background: Myeloperoxidase (MPO) is a lysosomal enzyme found in the azurophilic granules of polymorphonuclear leukocytes and is able to mediate inflammatory tissue destruction in aggressive and chronic periodontitis (CP). Human telomerase is a multi subunit ribonucleoprotein enzyme concerned with telomeric lengthening and homeostasis in man and has been found to be elevated in inflammatory conditions such as rheumatoid arthritis and periodontitis. The aim of this study was to explore in aggressive periodontitis (AP) subjects: (i) The role of MPO-463G/A gene polymorphism and (ii) the level of telomerase expression. These parameters have been compared with the subjects of CP and that of the healthy controls. Materials and Methods: A total of 45 subjects of the age group 20–50 years and free from any known systemic disease were included in the study. They were divided into three groups – Group I-periodontally healthy control (n = 15), Group II-CP (n = 15) and Group III-AP (n = 15). Peripheral blood samples and gingival tissue samples were collected for MPO gene polymorphism and telomerase expression, respectively, for detection by reverse transcriptase polymerase chain reaction. Results: The frequencies of AG and AA genotypes in the MPO gene polymorphism were more common in the AP subjects when compared to the controls. The m-RNA expression of human telomerase reverse transcriptase (hTERT) was undetectable in the gingival tissue of the control group. Its expression in AP subjects was significantly higher than that of CP group (83.61 ± 2.94 in CP and 104.27 ± 6.06 in AP) (P < 0.0001). Conclusions: Our data suggest that MPO-463G/A may be associated with increased risk of AP. The level of tissue hTERT was elevated in AP subjects as compared to CP and healthy control groups.
Collapse
Affiliation(s)
- Kundu Debabrata
- Department of Periodontia, Dr. R. Ahmed Dental College and Hospital, Kolkata, India
| | | | - Nair Vineet
- Department of Periodontia, Dr. R. Ahmed Dental College and Hospital, Kolkata, India
| | - Garg Anshul
- Department of Periodontia, Dr. R. Ahmed Dental College and Hospital, Kolkata, India
| | - Saha Arindam
- Private practice, Peerless Hospital and B.K Roy Research Centre, Kolkata, India
| | - Das Satadal
- Institute of Haematology and Transfusion Medicine, Peerless Hospital and B.K Roy Research Centre, Kolkata, India
| |
Collapse
|
25
|
Zhao Y, Cheng D, Wang S, Zhu J. Dual roles of c-Myc in the regulation of hTERT gene. Nucleic Acids Res 2014; 42:10385-98. [PMID: 25170084 PMCID: PMC4176324 DOI: 10.1093/nar/gku721] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 12/04/2022] Open
Abstract
Human telomerase gene hTERT is important for cancer and aging. hTERT promoter is regulated by multiple transcription factors (TFs) and its activity is dependent on the chromatin environment. However, it remains unsolved how the interplay between TFs and chromatin environment controls hTERT transcription. In this study, we employed the recombinase-mediated BAC targeting and BAC recombineering techniques to dissect the functions of two proximal E-box sites at -165 and +44 nt in regulating the hTERT promoter in the native genomic contexts. Our data showed that mutations of these sites abolished promoter binding by c-Myc/Max, USF1 and USF2, decreased hTERT promoter activity, and prevented its activation by overexpressed c-Myc. Upon inhibition of histone deacetylases, mutant and wildtype promoters were induced to the same level, indicating that the E-boxes functioned to de-repress the hTERT promoter and allowed its transcription in a repressive chromatin environment. Unexpectedly, knockdown of endogenous c-Myc/Max proteins activated hTERT promoter. This activation did not require the proximal E-boxes but was accompanied by increased promoter accessibility, as indicated by augmented active histone marks and binding of multiple TFs at the promoter. Our studies demonstrated that c-Myc/Max functioned in maintaining chromatin-dependent repression of the hTERT gene in addition to activating its promoter.
Collapse
Affiliation(s)
- Yuanjun Zhao
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - De Cheng
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| | - Shuwen Wang
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| | - Jiyue Zhu
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| |
Collapse
|
26
|
Doloff JC, Waxman DJ. Adenoviral vectors for prodrug activation-based gene therapy for cancer. Anticancer Agents Med Chem 2014; 14:115-26. [PMID: 23869779 DOI: 10.2174/18715206113139990309] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 01/09/2013] [Accepted: 04/10/2013] [Indexed: 11/22/2022]
Abstract
Cancer cell heterogeneity is a common feature - both between patients diagnosed with the same cancer and within an individual patient's tumor - and leads to widely different response rates to cancer therapies and the potential for the emergence of drug resistance. Diverse therapeutic approaches have been developed to combat the complexity of cancer, including individual treatment modalities designed to target tumor heterogeneity. This review discusses adenoviral vectors and how they can be modified to replicate in a cancer-specific manner and deliver therapeutic genes under multi-tiered regulation to target tumor heterogeneity, including heterogeneity associated with cancer stem cell-like subpopulations. Strategies that allow for combination of prodrug-activation gene therapy with a novel replication-conditional, heterogeneous tumor-targeting adenovirus are discussed, as are the benefits of using adenoviral vectors as tumor-targeting oncolytic vectors. While the anticancer activity of many adenoviral vectors has been well established in preclinical studies, only limited successes have been achieved in the clinic, indicating a need for further improvements in activity, specificity, tumor cell delivery and avoidance of immunogenicity.
Collapse
Affiliation(s)
| | - David J Waxman
- Department of Cell and Molecular, Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
27
|
Radan L, Hughes CS, Teichroeb JH, Vieira Zamora FM, Jewer M, Postovit LM, Betts DH. Microenvironmental regulation of telomerase isoforms in human embryonic stem cells. Stem Cells Dev 2014; 23:2046-66. [PMID: 24749509 DOI: 10.1089/scd.2013.0373] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recent evidence points to extra-telomeric, noncanonical roles for telomerase in regulating stem cell function. In this study, human embryonic stem cells (hESCs) were cultured in 20% or 2% O2 microenvironments for up to 5 days and evaluated for telomerase reverse transcriptase (TERT) expression and telomerase activity. Results showed increased cell survival and maintenance of the undifferentiated state with elevated levels of nuclear TERT in 2% O2-cultured hESCs despite no significant difference in telomerase activity compared with their high-O2-cultured counterparts. Pharmacological inhibition of telomerase activity using a synthetic tea catechin resulted in spontaneous hESC differentiation, while telomerase inhibition with a phosphorothioate oligonucleotide telomere mimic did not. Reverse transcription polymerase chain reaction (RT-PCR) analysis revealed variations in transcript levels of full-length and alternate splice variants of TERT in hESCs cultured under varying O2 atmospheres. Steric-blocking of Δα and Δβ hTERT splicing using morpholino oligonucleotides altered the hTERT splicing pattern and rapidly induced spontaneous hESC differentiation that appeared biased toward endomesodermal and neuroectodermal cell fates, respectively. Together, these results suggest that post-transcriptional regulation of TERT under varying O2 microenvironments may help regulate hESC survival, self-renewal, and differentiation capabilities through expression of extra-telomeric telomerase isoforms.
Collapse
Affiliation(s)
- Lida Radan
- 1 Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario , London, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
28
|
Hu X, Wu R, Shehadeh LA, Zhou Q, Jiang C, Huang X, Zhang L, Gao F, Liu X, Yu H, Webster KA, Wang J. Severe hypoxia exerts parallel and cell-specific regulation of gene expression and alternative splicing in human mesenchymal stem cells. BMC Genomics 2014; 15:303. [PMID: 24758227 PMCID: PMC4234502 DOI: 10.1186/1471-2164-15-303] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 04/16/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The endosteum of the bone marrow provides a specialized hypoxic niche that may serve to preserve the integrity, pluripotency, longevity and stemness of resident mesenchymal stem cells (MSCs). To explore the molecular genetic consequences of such a niche we subjected human (h) MSCs to a pO2 of 4 mmHg and analyzed global gene expression and alternative splicing (AS) by genome-exon microarray and RT-qPCR, and phenotype by western blot and immunostaining. RESULTS Out of 446 genes differentially regulated by >2.5-fold, down-regulated genes outnumbered up-regulated genes by 243:203. Exon analyses revealed 60 hypoxia-regulated AS events with splice indices (SI) >1.0 from 53 genes and a correlation between high SI and degree of transcript regulation. Parallel analyses of a publicly available AS study on human umbilical vein endothelial cells (HUVECs) showed that there was a strong cell-specific component with only 11 genes commonly regulated in hMSCs and HUVECs and 17 common differentially spliced genes. Only 3 genes were differentially responsive to hypoxia at the gene (>2.0) and AS levels in both cell types. Functional assignments revealed unique profiles of gene expression with complex regulation of differentiation, extracellular matrix, intermediate filament and metabolic marker genes. Antioxidant genes, striated muscle genes and insulin/IGF-1 signaling intermediates were down-regulated. There was a coordinate induction of 9 out of 12 acidic keratins that along with other epithelial and cell adhesion markers implies a partial mesenchymal to epithelial transition. CONCLUSIONS We conclude that severe hypoxia confers a quiescent phenotype in hMSCs that is reflected by both the transcriptome profile and gene-specific changes of splicosome actions. The results reveal that severe hypoxia imposes markedly different patterns of gene regulation of MSCs compared with more moderate hypoxia. This is the first study to report hypoxia-regulation of AS in stem/progenitor cells and the first molecular genetic characterization of MSC in a hypoxia-induced quiescent immobile state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Keith A Webster
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, P,R, China.
| | | |
Collapse
|
29
|
Wang Z, Zhang X, Li T, Li J, Tang Y, Le W. Valproic acid reduces neuritic plaque formation and improves learning deficits in APP(Swe) /PS1(A246E) transgenic mice via preventing the prenatal hypoxia-induced down-regulation of neprilysin. CNS Neurosci Ther 2014; 20:209-217. [PMID: 24289518 PMCID: PMC6493036 DOI: 10.1111/cns.12186] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 09/10/2013] [Accepted: 09/14/2013] [Indexed: 01/19/2023] Open
Abstract
AIMS Previously, we have documented that prenatal hypoxia can aggravate the cognitive impairment and Alzheimer's disease (AD) neuropathology in APP(Swe) /PS1(A246E) (APP/PS1) transgenic mice, and valproic acid (VPA) can prevent hypoxia-induced down-regulation of β-amyloid (Aβ) degradation enzyme neprilysin (NEP) in primary neurons. In this study, we have investigated the molecular mechanisms of VPA's anti-AD effects and found that VPA can reduce the prenatal hypoxia-induced neuritic plaque formation and improve the learning deficits in the AD mouse model. METHODS The pregnant APP/PS1 transgenic mice were exposed in a hypobaric chamber. Neuritic plaque staining, Morris water maze, and enzyme-linked immunosorbent assay (ELISA) were used to detect the effects of VPA on Aβ neuropathology, learning, and memory. Chromatin immunoprecipitation (ChIP) assays and real-time PCR (RT-PCR) were used to determine the effect of VPA on the histone3 acetylation (H3-Ace). RESULTS We found that VPA can inhibit neuritic plaque formation and improve the learning and memory in the prenatal hypoxic APP/PS1 transgenic mice. In addition, VPA treatment can decrease the soluble and insoluble Aβ42 levels and increase the NEP expression via up-regulation of H3-Ace in the APP/PS1 transgenic mice. CONCLUSION Valproic acid is able to attenuate the prenatal hypoxia-induced Aβ neuropathology and learning and memory deficits via inhibiting the activation of histone deacetylase 1 (HDAC1), preventing the decrease in H3-Ace in the NEP promoter regions and reducing the down-regulation of NEP.
Collapse
Affiliation(s)
- Zheng Wang
- Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Medical NeurobiologyDepartment of NeurologyHuashan HospitalFudan UniversityShanghaiChina
| | - Xiao‐Jie Zhang
- Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ting Li
- Institute of Health SciencesShanghai Institutes for Biological SciencesChinese Academy of Sciences, & Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jia Li
- Institute of Health SciencesShanghai Institutes for Biological SciencesChinese Academy of Sciences, & Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yu Tang
- Institute of Health SciencesShanghai Institutes for Biological SciencesChinese Academy of Sciences, & Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weidong Le
- Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Health SciencesShanghai Institutes for Biological SciencesChinese Academy of Sciences, & Shanghai Jiao Tong University School of MedicineShanghaiChina
- 1 Affiliated HospitalDalian Medical UniversityDalianChina
| |
Collapse
|
30
|
Mathematical model of a telomerase transcriptional regulatory network developed by cell-based screening: analysis of inhibitor effects and telomerase expression mechanisms. PLoS Comput Biol 2014; 10:e1003448. [PMID: 24550717 PMCID: PMC3923661 DOI: 10.1371/journal.pcbi.1003448] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 11/30/2013] [Indexed: 12/16/2022] Open
Abstract
Cancer cells depend on transcription of telomerase reverse transcriptase (TERT). Many transcription factors affect TERT, though regulation occurs in context of a broader network. Network effects on telomerase regulation have not been investigated, though deeper understanding of TERT transcription requires a systems view. However, control over individual interactions in complex networks is not easily achievable. Mathematical modelling provides an attractive approach for analysis of complex systems and some models may prove useful in systems pharmacology approaches to drug discovery. In this report, we used transfection screening to test interactions among 14 TERT regulatory transcription factors and their respective promoters in ovarian cancer cells. The results were used to generate a network model of TERT transcription and to implement a dynamic Boolean model whose steady states were analysed. Modelled effects of signal transduction inhibitors successfully predicted TERT repression by Src-family inhibitor SU6656 and lack of repression by ERK inhibitor FR180204, results confirmed by RT-QPCR analysis of endogenous TERT expression in treated cells. Modelled effects of GSK3 inhibitor 6-bromoindirubin-3′-oxime (BIO) predicted unstable TERT repression dependent on noise and expression of JUN, corresponding with observations from a previous study. MYC expression is critical in TERT activation in the model, consistent with its well known function in endogenous TERT regulation. Loss of MYC caused complete TERT suppression in our model, substantially rescued only by co-suppression of AR. Interestingly expression was easily rescued under modelled Ets-factor gain of function, as occurs in TERT promoter mutation. RNAi targeting AR, JUN, MXD1, SP3, or TP53, showed that AR suppression does rescue endogenous TERT expression following MYC knockdown in these cells and SP3 or TP53 siRNA also cause partial recovery. The model therefore successfully predicted several aspects of TERT regulation including previously unknown mechanisms. An extrapolation suggests that a dominant stimulatory system may programme TERT for transcriptional stability. Tumour cells acquire the ability to divide and multiply indefinitely whereas normal cells can undergo only a limited number of divisions. The switch to immortalisation of the tumour cell is dependent on maintaining the integrity of telomere DNA which forms chromosome ends and is achieved through activation of the telomerase enzyme by turning on synthesis of the TERT gene, which is usually silenced in normal cells. Suppressing telomerase is toxic to cancer cells and it is widely believed that understanding TERT regulation could lead to potential cancer therapies. Previous studies have identified many of the factors which individually contribute to activate or repress TERT levels in cancer cells. However, transcription factors do not behave in isolation in cells, but rather as a complex co-operative network displaying inter-regulation. Therefore, full understanding of TERT regulation will require a broader view of the transcriptional network. In this paper we take a computational modelling approach to study TERT regulation at the network level. We tested interactions between 14 TERT-regulatory factors in an ovarian cancer cell line using a screening approach and developed a model to analyse which network interventions were able to silence TERT.
Collapse
|
31
|
Bollmann FM. Physiological and pathological significance of human telomerase reverse transcriptase splice variants. Biochimie 2013; 95:1965-70. [DOI: 10.1016/j.biochi.2013.07.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 07/29/2013] [Indexed: 12/22/2022]
|
32
|
The growing complexity of HIF-1α’s role in tumorigenesis: DNA repair and beyond. Oncogene 2012; 32:3569-76. [DOI: 10.1038/onc.2012.510] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 09/28/2012] [Accepted: 09/27/2012] [Indexed: 12/13/2022]
|
33
|
Upstream stimulatory factor 2 and hypoxia-inducible factor 2α (HIF2α) cooperatively activate HIF2 target genes during hypoxia. Mol Cell Biol 2012; 32:4595-610. [PMID: 22966206 DOI: 10.1128/mcb.00724-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
While the functions of hypoxia-inducible factor 1α (HIF1α)/aryl hydrocarbon receptor nuclear translocator (ARNT) and HIF2α/ARNT (HIF2) proteins in activating hypoxia-inducible genes are well established, the role of other transcription factors in the hypoxic transcriptional response is less clear. We report here for the first time that the basic helix-loop-helix-leucine-zip transcription factor upstream stimulatory factor 2 (USF2) is required for the hypoxic transcriptional response, specifically, for hypoxic activation of HIF2 target genes. We show that inhibiting USF2 activity greatly reduces hypoxic induction of HIF2 target genes in cell lines that have USF2 activity, while inducing USF2 activity in cells lacking USF2 activity restores hypoxic induction of HIF2 target genes. Mechanistically, USF2 activates HIF2 target genes by binding to HIF2 target gene promoters, interacting with HIF2α protein, and recruiting coactivators CBP and p300 to form enhanceosome complexes that contain HIF2α, USF2, CBP, p300, and RNA polymerase II on HIF2 target gene promoters. Functionally, the effect of USF2 knockdown on proliferation, motility, and clonogenic survival of HIF2-dependent tumor cells in vitro is phenocopied by HIF2α knockdown, indicating that USF2 works with HIF2 to activate HIF2 target genes and to drive HIF2-depedent tumorigenesis.
Collapse
|
34
|
Hashimoto Y, Tazawa H, Teraishi F, Kojima T, Watanabe Y, Uno F, Yano S, Urata Y, Kagawa S, Fujiwara T. The hTERT promoter enhances the antitumor activity of an oncolytic adenovirus under a hypoxic microenvironment. PLoS One 2012; 7:e39292. [PMID: 22720091 PMCID: PMC3376103 DOI: 10.1371/journal.pone.0039292] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 05/18/2012] [Indexed: 12/11/2022] Open
Abstract
Hypoxia is a microenvironmental factor that contributes to the invasion, progression and metastasis of tumor cells. Hypoxic tumor cells often show more resistance to conventional chemoradiotherapy than normoxic tumor cells, suggesting the requirement of novel antitumor therapies to efficiently eliminate the hypoxic tumor cells. We previously generated a tumor-specific replication-competent oncolytic adenovirus (OBP-301: Telomelysin), in which the human telomerase reverse transcriptase (hTERT) promoter drives viral E1 expression. Since the promoter activity of the hTERT gene has been shown to be upregulated by hypoxia, we hypothesized that, under hypoxic conditions, the antitumor effect of OBP-301 with the hTERT promoter would be more efficient than that of the wild-type adenovirus 5 (Ad5). In this study, we investigated the antitumor effects of OBP-301 and Ad5 against human cancer cells under a normoxic (20% oxygen) or a hypoxic (1% oxygen) condition. Hypoxic condition induced nuclear accumulation of the hypoxia-inducible factor-1α and upregulation of hTERT promoter activity in human cancer cells. The cytopathic activity of OBP-301 was significantly higher than that of Ad5 under hypoxic condition. Consistent with their cytopathic activity, the replication of OBP-301 was significantly higher than that of Ad5 under the hypoxic condition. OBP-301-mediated E1A was expressed within hypoxic areas of human xenograft tumors in mice. These results suggest that the cytopathic activity of OBP-301 against hypoxic tumor cells is mediated through hypoxia-mediated activation of the hTERT promoter. Regulation of oncolytic adenoviruses by the hTERT promoter is a promising antitumor strategy, not only for induction of tumor-specific oncolysis, but also for efficient elimination of hypoxic tumor cells.
Collapse
Affiliation(s)
- Yuuri Hashimoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Center for Gene and Cell Therapy, Okayama University Hospital, Okayama, Japan
| | - Fuminori Teraishi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Kojima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuichi Watanabe
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Futoshi Uno
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shuya Yano
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- * E-mail:
| |
Collapse
|
35
|
|
36
|
hTERT promoter methylation and telomere length in childhood acute lymphoblastic leukemia: associations with immunophenotype and cytogenetic subgroup. Exp Hematol 2011; 39:1144-51. [PMID: 21914494 DOI: 10.1016/j.exphem.2011.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Revised: 08/17/2011] [Accepted: 08/22/2011] [Indexed: 01/11/2023]
Abstract
Telomere maintenance, important for long-term cell survival and malignant transformation, is directed by a multitude of factors, including epigenetic mechanisms, and has been implicated in outcomes for patients with leukemia. In the present study, the objective was to investigate the biological and clinical significance of telomere length and promoter methylation of the human telomerase reverse transcriptase gene in childhood acute lymphoblastic leukemia. A cohort of 169 childhood acute lymphoblastic leukemias was investigated for telomere length, human telomerase reverse transcriptase gene promoter methylation status, genomic aberrations, immunophenotype, and clinical outcomes. Methylation of the core promoter of the human telomerase reverse transcriptase (hTERT) gene was demonstrated in 24% of diagnostic samples, with a significant difference between B-cell precursor (n = 130) and T-cell acute lymphoblastic leukemia (ALL) (n = 17) cases (18% and 72%, respectively; p < 0.001). No remission sample demonstrated hTERT promoter methylation (n = 40). Within the B-cell precursor group, t(12;21)(p13;q22) [ETV6/RUNX1] cases (n = 19) showed a much higher frequency of hTERT methylation than high-hyperdiploid (51-61 chromosomes) ALL (n = 44) (63% and 7%, respectively; p < 0.001). hTERT messenger RNA levels were negatively associated with methylation status and, in the t(12;21) group, methylated cases had shorter telomeres (p = 0.017). In low-risk B-cell precursor patients (n = 101), long telomeres indicated a worse prognosis. The collected data from the present study indicate that the telomere biology in childhood ALL has clinical implications and reflects molecular differences between diverse ALL subgroups.
Collapse
|
37
|
Joshua AM, Shen E, Yoshimoto M, Marrano P, Zielenska M, Evans AJ, Van der Kwast T, Squire JA. Topographical analysis of telomere length and correlation with genomic instability in whole mount prostatectomies. Prostate 2011; 71:778-90. [PMID: 21031437 DOI: 10.1002/pros.21294] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 09/26/2010] [Indexed: 11/06/2022]
Abstract
BACKGROUND Many critical events in prostatic carcinogenesis appear to relate to the emergence of genomic instability. Characteristic genomic abnormalities such as 8p loss, 8q gain, trisomy 7, and PTEN microdeletions may provide selective advantages to increase neoplastic transformation. Evidence suggests that telomere dysfunction is a plausible mechanism for some of these abnormalities on the basis of the break-fusion-bridge cycle that can lead to manifestations of genomic instability. METHODS In this study, we correlate telomere length measured by quantitative FISH in various prostatic histologies with markers of genomic instability and immunohistochemical measures of proliferation and oxidative stress. RESULTS We find that telomere shortening is correlated with abnormalities on chromosome 8, but not with trisomy 7 or abnormalities of the PTEN locus. There are associations with C-MYC aberrations in stroma with greater proximity to cancer and a correlation between telomere length in a number of prostatic histologies and the adjacent stroma, suggesting the importance of microenvironmental effects on telomere maintenance in the prostate. This finding was also supported by the finding of the correlation between telomere attrition and the levels of oxidative stress as measured by malondialdehyde staining in HPIN lesions close to cancer. CONCLUSIONS Telomere attrition in the prostate gland is associated with particular genomic aberrations that contribute to the genomic instability characteristic of prostatic carcinogenesis. Correlations between various histologies and adjacent stroma telomere length suggest it is also may reveal microenvironmental effects within the prostate gland. Oxidative stress may contribute to telomere attrition in HPIN close to cancer.
Collapse
Affiliation(s)
- A M Joshua
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang Z, Yang D, Zhang X, Li T, Li J, Tang Y, Le W. Hypoxia-induced down-regulation of neprilysin by histone modification in mouse primary cortical and hippocampal neurons. PLoS One 2011; 6:e19229. [PMID: 21559427 PMCID: PMC3084787 DOI: 10.1371/journal.pone.0019229] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 03/23/2011] [Indexed: 01/12/2023] Open
Abstract
Amyloid β-peptide (Aβ) accumulation leads to neurodegeneration and Alzheimer's disease (AD). Aβ metabolism is a dynamic process in the Aβ production and clearance that requires neprilysin (NEP) and other enzymes to degrade Aβ. It has been reported that NEP expression is significantly decreased in the brain of AD patients. Previously we have documented hypoxia is a risk factor for Aβ generation in vivo and in vitro through increasing Aβ generation by altering β-cleavage and γ-cleavage of APP and down-regulating NEP, and causing tau hyperphosphorylation. Here, we investigated the molecular mechanisms of hypoxia-induced down-regulation of NEP. We found a significant decrease in NEP expression at the mRNA and protein levels after hypoxic treatment in mouse primary cortical and hippocampal neurons. Chromatin immunoprecipitation (ChIP) assays and relative quantitative PCR (q-PCR) revealed an increase of histone H3-lysine9 demethylation (H3K9me2) and a decrease of H3 acetylation (H3-Ace) in the NEP promoter regions following hypoxia. In addition, we found that hypoxia caused up-regulation of histone methyl transferase (HMT) G9a and histone deacetylases (HDACs) HDAC-1. Decreased expression of NEP during hypoxia can be prevented by application with the epigenetic regulators 5-Aza-2'-deoxycytidine (5-Aza), HDACs inhibitor sodium valproate (VA), and siRNA-mediated knockdown of G9a or HDAC1. DNA methylation PCR data do not support that hypoxia affects the methylation of NEP promoters. This study suggests that hypoxia may down-regulate NEP by increasing H3K9me2 and decreasing H3-Ace modulation.
Collapse
Affiliation(s)
- Zheng Wang
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dehua Yang
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojie Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Li
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Li
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Tang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weidong Le
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
39
|
Rastmanesh R. Potential of melatonin to treat or prevent age-related macular degeneration through stimulation of telomerase activity. Med Hypotheses 2011; 76:79-85. [DOI: 10.1016/j.mehy.2010.08.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Accepted: 08/07/2010] [Indexed: 12/15/2022]
|
40
|
Heeg S, Hirt N, Queisser A, Schmieg H, Thaler M, Kunert H, Quante M, Goessel G, von Werder A, Harder J, Beijersbergen R, Blum HE, Nakagawa H, Opitz OG. EGFR overexpression induces activation of telomerase via PI3K/AKT-mediated phosphorylation and transcriptional regulation through Hif1-alpha in a cellular model of oral-esophageal carcinogenesis. Cancer Sci 2010; 102:351-60. [PMID: 21156006 DOI: 10.1111/j.1349-7006.2010.01796.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Telomerase plays an important role during immortalization and malignant transformation as crucial steps in the development of human cancer. In a cellular model of oral-esophageal carcinogenesis, recapitulating the human disease, immortalization occurred independent of the activation of telomerase but through the recombination-based alternative lengthening of telomeres (ALT). In this stepwise model, additional overexpression of EGFR led to in vitro transformation and activation of telomerase with homogeneous telomere elongation in already immortalized oral squamous epithelial cells (OKF6-D1_dnp53). More interestingly, EGFR overexpression activated the PI3K/AKT pathway. This strongly suggested a role for telomerase in tumor progression in addition to just elongating telomeres and inferring an immortalized state. Therefore, we sought to identify the regulatory mechanisms involved in this activation of telomerase and in vitro transformation induced by EGFR. In the present study we demonstrate that telomerase expression and activity are induced through both direct phosphorylation of hTERT by phospho-AKT as well as PI3K-dependent transcriptional regulation involving Hif1-alpha as a key transcription factor. Furthermore, EGFR overexpression enhanced cell cycle progression and proliferation via phosphorylation and translocation of p21. Whereas immortalization was induced by ALT, in vitro transformation was associated with telomerase activation, supporting an additional role for telomerase in tumor progression besides elongating telomeres.
Collapse
Affiliation(s)
- Steffen Heeg
- Department of Medicine, Institute for Molecular Medicine and Cell Research, Tumorzentrum Ludwig Heilmeyer-Comprehensive Cancer Center Freiburg, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Petrenko A, Korolenkova L, Skvortsov D, Fedorova M, Skoblov M, Baranova A, Zvereva M, Rubtsova M, Kisseljov F. Cervical intraepithelial neoplasia: Telomerase activity and splice pattern of hTERT mRNA. Biochimie 2010; 92:1827-31. [DOI: 10.1016/j.biochi.2010.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 07/27/2010] [Indexed: 11/26/2022]
|
42
|
Kobliakov VA. Mechanisms of tumor promotion by reactive oxygen species. BIOCHEMISTRY (MOSCOW) 2010; 75:675-85. [PMID: 20636258 DOI: 10.1134/s0006297910060015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review analyzes the available information concerning mechanisms of non-genotoxic action of reactive oxygen species (ROS) during tumor promotion and pathways of their generation under the influence of chemical compounds. Special attention is given to the ability of ROS to induce pseudohypoxia through inhibition of prolyl oxidase, which is an oxygen sensor in the cell. Functions of HIF-1alpha as a main contributor to the ROS-induced promotion are analyzed. Data suggest that an unregulated high level of HIF-1alpha in the cell could induce the development of tumors. Hypothetical possibilities of ROS production under the influence of different environmental pollutants, which are promoters of tumorigenesis, include functioning of cytochrome P450 during oxidation of substrates, functioning of the mitochondrial respiratory chain, and action of peroxisome proliferators.
Collapse
Affiliation(s)
- V A Kobliakov
- Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russia.
| |
Collapse
|
43
|
Mild hyperoxia limits hTR levels, telomerase activity, and telomere length maintenance in hTERT-transduced bone marrow endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:1142-53. [DOI: 10.1016/j.bbamcr.2010.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 06/28/2010] [Accepted: 06/28/2010] [Indexed: 01/01/2023]
|
44
|
Coussens M, Davy P, Brown L, Foster C, Andrews WH, Nagata M, Allsopp R. RNAi screen for telomerase reverse transcriptase transcriptional regulators identifies HIF1alpha as critical for telomerase function in murine embryonic stem cells. Proc Natl Acad Sci U S A 2010; 107:13842-7. [PMID: 20643931 PMCID: PMC2922273 DOI: 10.1073/pnas.0913834107] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In various types of stem cells, including embryonic stem (ES) cells and hematopoietic stem cells, telomerase functions to ensure long-term self-renewal capacity via maintenance of telomere reserve. Expression of the catalytic component of telomerase, telomerase reverse transcriptase (Tert), which is essential for telomerase activity, is limiting in many types of cells and therefore plays an important role in establishing telomerase activity levels. However, the mechanisms regulating expression of Tert in cells, including stem cells, are presently poorly understood. In the present study, we sought to identify genes involved in the regulation of Tert expression in stem cells by performing a screen in murine ES (mES) cells using a shRNA expression library targeting murine transcriptional regulators. Of 18 candidate transcriptional regulators of Tert expression identified in this screen, only one candidate, hypoxia inducible factor 1 alpha (Hif1alpha), did not have a significant effect on mES cell morphology, survival, or growth rate. Direct shRNA-mediated knockdown of Hif1alpha expression confirmed that suppression of Hif1alpha levels was accompanied by a reduction in both Tert mRNA and telomerase activity levels. Furthermore, gradual telomere attrition was observed during extensive proliferation of Hif1alpha-targeted mES cells. Switching Hif1alpha-targeted mES cells to a hypoxic environment largely restored Hif1alpha levels, as well as Tert expression, telomerase activity levels, and telomere length. Together, these findings suggest a direct effect of Hif1alpha on telomerase regulation in mES cells, and imply that Hif1alpha may have a physiologically relevant role in maintenance of functional levels of telomerase in stem cells.
Collapse
Affiliation(s)
- Matthew Coussens
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI 96813; and
| | - Philip Davy
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI 96813; and
| | | | | | | | - Melissa Nagata
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI 96813; and
| | - Richard Allsopp
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI 96813; and
| |
Collapse
|
45
|
Meeran SM, Patel SN, Tollefsbol TO. Sulforaphane causes epigenetic repression of hTERT expression in human breast cancer cell lines. PLoS One 2010; 5:e11457. [PMID: 20625516 PMCID: PMC2897894 DOI: 10.1371/journal.pone.0011457] [Citation(s) in RCA: 282] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 06/11/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Sulforaphane (SFN), an isothiocyanate found in cruciferous vegetables, is a common dietary component that has histone deacetylase inhibition activity and exciting potential in cancer prevention. The mechanisms by which SFN imparts its chemopreventive properties are of considerable interest and little is known of its preventive potential for breast cancer. PRINCIPAL FINDINGS We found that SFN significantly inhibits the viability and proliferation of breast cancer cells in vitro while it has negligible effects on normal breast cells. Inhibition of telomerase has received considerable attention because of its high expression in cancer cells and extremely low level of expression in normal cells. SFN treatment dose- and time-dependently inhibited human telomerase reverse transcriptase (hTERT), the catalytic regulatory subunit of telomerase, in both MCF-7 and MDA-MB-231 human breast cancer cells. DNA methyltransferases (DNMTs), especially DNMT1 and DNMT3a, were also decreased in SFN-treated breast cancer cells suggesting that SFN may repress hTERT by impacting epigenetic pathways. Down-regulation of DNMTs in response to SFN induced site-specific CpG demethylation occurring primarily in the first exon of the hTERT gene thereby facilitating CTCF binding associated with hTERT repression. Chromatin immunoprecipitation (ChIP) analysis of the hTERT promoter revealed that SFN increased the level of active chromatin markers acetyl-H3, acetyl-H3K9 and acetyl-H4, whereas the trimethyl-H3K9 and trimethyl-H3K27 inactive chromatin markers were decreased in a dose-dependent manner. SFN-induced hyperacetylation facilitated the binding of many hTERT repressor proteins such as MAD1 and CTCF to the hTERT regulatory region. Depletion of CTCF using siRNA reduced the SFN-induced down-regulation of hTERT mRNA transcription in these breast cancer cells. In addition, down-regulation of hTERT expression facilitated the induction of cellular apoptosis in human breast cancer cells. SIGNIFICANCE Collectively, our results provide novel insights into SFN-mediated epigenetic down-regulation of telomerase in breast cancer prevention and may open new avenues for approaches to SFN-mediated cancer prevention.
Collapse
Affiliation(s)
- Syed M. Meeran
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Shweta N. Patel
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Aging, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
46
|
Kim J, Lee S, Bhattacharjee R, Khalyfa A, Kheirandish-Gozal L, Gozal D. Leukocyte telomere length and plasma catestatin and myeloid-related protein 8/14 concentrations in children with obstructive sleep apnea. Chest 2010; 138:91-9. [PMID: 20299626 PMCID: PMC2897695 DOI: 10.1378/chest.09-2832] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 02/05/2010] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is common in children and leads to multiple end-organ morbidities induced by the cumulative burden of oxidative stress and inflammation. Leukocyte telomere length (LTL) reflects not only chronologic age but also the burden of disease. We hypothesized that LTL would be decreased in children with OSA. METHODS Two hundred thirteen children (mean age, 7.7 +/- 1.4 years) were included after a sleep study and a morning blood sample. LTL was examined by quantitative polymerase chain reaction in a case-control setting involving 111 OSA cases and 102 controls. Myeloid-related protein (MRP) 8/14 and catestatin plasma levels also were assayed using enzyme-linked immunosorbent assay. RESULTS Log LTL was significantly increased and OSA severity dependently increased in children (P = .012), was positively associated with apnea-hypopnea index (AHI) (r = 0.236; P < .01), and was inversely correlated with age (r = -0.145; P < .05). In a multivariate regression model, LTL was independently associated with AHI (beta = 0.28; P = .002) after adjusting for age, sex, BMI z score, and race. Children with OSA exhibited higher BP (P < .05), lower plasma catestatin (P = .009), and higher MRP 8/14 levels (P < .001) than controls. Of note, children with the lowest plasma catestatin levels (< 1.39 ng/mL) had 5.2-fold increased odds of moderate-to-severe OSA (95% CI, 1.19-23.4 ng/mL; P < .05) after adjusting for confounding variables. CONCLUSIONS In pediatric OSA, LTL is longer rather than shorter. Children with OSA have reduced plasma catestatin levels and increased BP along with increased MRP 8/14 levels that exhibit AHI dependencies. Thus, catestatin and MRP 8/14 levels may serve as biomarkers for cardiovascular risk in the context of pediatric OSA. However, the implications of increased LTL in children with OSA remain to be defined.
Collapse
Affiliation(s)
- Jinkwan Kim
- Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, 5721 S Maryland Ave, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
47
|
Epel ES, Lin J, Dhabhar FS, Wolkowitz OM, Puterman E, Karan L, Blackburn EH. Dynamics of telomerase activity in response to acute psychological stress. Brain Behav Immun 2010; 24:531-9. [PMID: 20018236 PMCID: PMC2856774 DOI: 10.1016/j.bbi.2009.11.018] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 11/17/2009] [Accepted: 11/26/2009] [Indexed: 12/26/2022] Open
Abstract
Telomerase activity plays an essential role in cell survival, by lengthening telomeres and promoting cell growth and longevity. It is now possible to quantify the low levels of telomerase activity in human leukocytes. Low basal telomerase activity has been related to chronic stress in people and to chronic glucocorticoid exposure in vitro. Here we test whether leukocyte telomerase activity changes under acute psychological stress. We exposed 44 elderly women, including 22 high stress dementia caregivers and 22 matched low stress controls, to a brief laboratory psychological stressor, while examining changes in telomerase activity of peripheral blood mononuclear cells (PBMCs). At baseline, caregivers had lower telomerase activity levels than controls, but during stress telomerase activity increased similarly in both groups. Across the entire sample, subsequent telomerase activity increased by 18% one hour after the end of the stressor (p<0.01). The increase in telomerase activity was independent of changes in numbers or percentages of monocytes, lymphocytes, and specific T cell types, although we cannot fully rule out some potential contribution from immune cell redistribution in the change in telomerase activity. Telomerase activity increases were associated with greater cortisol increases in response to the stressor. Lastly, psychological response to the tasks (greater threat perception) was also related to greater telomerase activity increases in controls. These findings uncover novel relationships of dynamic telomerase activity with exposure to an acute stressor, and with two classic aspects of the stress response - perceived psychological stress and neuroendocrine (cortisol) responses to the stressor.
Collapse
Affiliation(s)
- Elissa S. Epel
- University of California, San Francisco, Dept of Psychiatry, 3333 Calif St, Suite 465, San Francisco, CA 94143,Corresponding authors, Elissa Epel, 3333 Calif. St, Ste 465, San Francisco, CA 94143, , 415-476-7648, Fax: 415-476-7744, Elizabeth Blackburn, 600 16th St, Room S-312F, San Francisco, CA, 94107, , 415-476-4912, FAX: 415-514-2913
| | - Jue Lin
- University of California, San Francisco, Dept of Biochemistry and Biophysics
| | - Firdaus S. Dhabhar
- Dept. of Psychiatry & Behavioral Sciences, and Institute for Immunity, Transplantation, and Infection, Stanford University, 300 Pasteur Drive, MC 5135, Stanford, CA 94305
| | - Owen M. Wolkowitz
- University of California, San Francisco, Dept of Psychiatry, 3333 Calif St, Suite 465, San Francisco, CA 94143
| | - E Puterman
- University of California, San Francisco, Dept of Psychiatry, 3333 Calif St, Suite 465, San Francisco, CA 94143
| | - Lori Karan
- University of California, San Francisco, Dept of Psychiatry, 3333 Calif St, Suite 465, San Francisco, CA 94143
| | - Elizabeth H. Blackburn
- University of California, San Francisco, Dept of Biochemistry and Biophysics,Corresponding authors, Elissa Epel, 3333 Calif. St, Ste 465, San Francisco, CA 94143, , 415-476-7648, Fax: 415-476-7744, Elizabeth Blackburn, 600 16th St, Room S-312F, San Francisco, CA, 94107, , 415-476-4912, FAX: 415-514-2913
| |
Collapse
|
48
|
Doloff JC, Waxman DJ, Jounaidi Y. Human telomerase reverse transcriptase promoter-driven oncolytic adenovirus with E1B-19 kDa and E1B-55 kDa gene deletions. Hum Gene Ther 2009; 19:1383-400. [PMID: 18771358 DOI: 10.1089/hum.2008.056] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We constructed an oncolytic adenovirus, Adeno-hTERT-E1A, with deletions of the viral E1B, E3A, and E3B regions and insertion of a human telomerase reverse transcriptase (hTERT) promoter-driven early viral 1A (E1A) cassette that confers high transcriptional activity in multiple human tumor cell lines. The oncolytic potential of Adeno-hTERT-E1A was characterized in comparison with that of the E1B-55 kDa- and E3B-region-deleted oncolytic adenovirus ONYX-015. Tumor cells infected with Adeno-hTERT-E1A expressed dramatically higher levels of E1A oncoprotein, underwent enhanced lysis, and displayed an earlier and higher apoptotic index than cells infected with ONYX-015. Despite the increase in virus-induced apoptotic death, Adeno-hTERT-E1A replicated and produced functional progeny leading to viral spread, but with reduced efficiency compared with ONYX-015, in particular in A549 cells. Virus-induced E1A expression, host cell apoptosis, viral hexon protein production, and DNA synthesis were markedly reduced in primary human hepatocytes after infection with Adeno-hTERT-E1A as compared with ONYX-015. The strong oncolytic activity of Adeno-hTERT-E1A in tumor cell culture translated into superior antitumor activity in vivo in an MDA-MB-231 solid tumor xenograft model. Adeno-hTERT-E1A thus has strong therapeutic potential and an improved safety profile compared with ONYX-015, which may lead to reduced toxicity in the clinic.
Collapse
Affiliation(s)
- Joshua C Doloff
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215, USA
| | | | | |
Collapse
|
49
|
Sitaram RT, Cairney CJ, Grabowski P, Keith WN, Hallberg B, Ljungberg B, Roos G. The PTEN regulator DJ-1 is associated with hTERT expression in clear cell renal cell carcinoma. Int J Cancer 2009; 125:783-90. [DOI: 10.1002/ijc.24335] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
50
|
Vavassori S, Covey LR. Post-transcriptional regulation in lymphocytes: the case of CD154. RNA Biol 2009; 6:259-65. [PMID: 19395873 DOI: 10.4161/rna.6.3.8581] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The control of mRNA decay is emerging as an important control point and a major contributor to gene expression in both immune and non-immune cells. The identification of protein factors and cis-acting elements responsible for transcript degradation has illuminated a comprehensive picture of precisely orchestrated events required to both regulate and establish the decay process. One gene that is highly regulated at the post-transcriptional level is CD40 ligand (CD154 or CD40L). CD154 on CD4(+) T cells is tightly controlled by an interacting network of transcriptional and post-transcriptional processes that result in precise surface levels of protein throughout an extended time course of antigen stimulation. The activation-induced stabilization of the CD154 transcript by a polypyrimidine tract-binding protein (PTB)-complex is a key event that corresponds to the temporal expression of CD154. In this review, we discuss known and potential roles of major mRNA decay pathways in lymphocytes and focus on the unique post-transcriptional mechanisms leading to CD154 expression by activated CD4(+) T cells.
Collapse
Affiliation(s)
- Stefano Vavassori
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.
| | | |
Collapse
|