1
|
Aborode AT, Abass OA, Nasiru S, Eigbobo MU, Nefishatu S, Idowu A, Tiamiyu Z, Awaji AA, Idowu N, Busayo BR, Mehmood Q, Onifade IA, Fakorede S, Akintola AA. RNA binding proteins (RBPs) on genetic stability and diseases. Glob Med Genet 2025; 12:100032. [PMID: 39925443 PMCID: PMC11803229 DOI: 10.1016/j.gmg.2024.100032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 02/11/2025] Open
Abstract
RNA-binding proteins (RBPs) are integral components of cellular machinery, playing crucial roles in the regulation of gene expression and maintaining genetic stability. Their interactions with RNA molecules govern critical processes such as mRNA splicing, stability, localization, and translation, which are essential for proper cellular function. These proteins interact with RNA molecules and other proteins to form ribonucleoprotein complexes (RNPs), hence controlling the fate of target RNAs. The interaction occurs via RNA recognition motif, the zinc finger domain, the KH domain and the double stranded RNA binding motif (all known as RNA-binding domains (RBDs). These domains are found within the coding sequences (intron and exon domains), 5' untranslated regions (5'UTR) and 3' untranslated regions (3'UTR). Dysregulation of RBPs can lead to genomic instability, contributing to various pathologies, including cancer neurodegenerative diseases, and metabolic disorders. This study comprehensively explores the multifaceted roles of RBPs in genetic stability, highlighting their involvement in maintaining genomic integrity through modulation of RNA processing and their implications in cellular signalling pathways. Furthermore, it discusses how aberrant RBP function can precipitate genetic instability and disease progression, emphasizing the therapeutic potential of targeting RBPs in restoring cellular homeostasis. Through an analysis of current literature, this study aims to delineate the critical role of RBPs in ensuring genetic stability and their promise as targets for innovative therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Shaibu Nasiru
- Department of Research and Development, Healthy Africans Platform, Ibadan, Nigeria
- Department of Biochemistry, Ambrose Alli University Ekpoma, Nigeria
| | | | - Sumana Nefishatu
- Department of Biochemistry, Ambrose Alli University Ekpoma, Nigeria
| | - Abdullahi Idowu
- Department of Biological Sciences, Purdue University Fort Wayne, USA
| | - Zainab Tiamiyu
- Department of Biochemistry and Cancer Biology, Medical College of Georgia, Augusta University, USA
| | - Aeshah A. Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Nike Idowu
- Department of Chemistry, University of Nebraska-Lincoln, USA
| | | | - Qasim Mehmood
- Shifa Clinical Research Center, Shifa International Hospital, Islamabad, Pakistan
| | - Isreal Ayobami Onifade
- Department of Division of Family Health, Health Research Incorporated, New York State Department of Health, USA
| | - Sodiq Fakorede
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ashraf Akintayo Akintola
- Department of Biology Education, Teachers College & Institute for Phylogenomics and Evolution, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
2
|
Lacalle-Gonzalez C, Florez-Cespedes M, Sanz-Criado L, Ochieng’ Otieno M, Ramos-Muñoz E, Fernandez-Aceñero MJ, Ortega-Medina L, Garcia-Foncillas J, Martinez-Useros J. DLL3 Is a Prognostic and Potentially Predictive Biomarker for Immunotherapy Linked to PD/PD-L Axis and NOTCH1 in Pancreatic Cancer. Biomedicines 2023; 11:2812. [PMID: 37893184 PMCID: PMC10604228 DOI: 10.3390/biomedicines11102812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive neoplasm with very poor patient survival outcomes despite available treatments. There is an urgent need for new potential treatment options and novel biomarkers for these patients. Delta-like canonical Notch ligand 3 (DLL3) interacts with the Notch receptor and causes inhibition of Notch signaling, which confers a survival advantage to PDAC cells. Thus, DLL3 expression could affect cell survival, and its inhibition could increase a patient's survival. To test this hypothesis, a survival analysis was conducted using the progression-free and overall survival from two independent datasets of PDAC patients, with one using mRNA z-score levels and the other using the Hscore protein expression level; both were carried out using a log-rank test and plotted using Kaplan-Meier curves. DLL3 at the mRNA expression level showed an association between high mRNA expression and both a longer progression-free survival (PFS) and overall survival (OS) of patients. Then, we designed a retrospective study with resected PDAC samples. Our primary objective with this dataset was to assess the relationship between PFS and OS and DLL3 protein expression. The secondary assessment was to provide a rationale for the use of anti-DLL3-based treatments in combination with immunotherapy that is supported by the link between DLL3 and other factors that are involved in immune checkpoints. The survival analyses revealed a protective effect of high DLL3 protein expression levels in both PFS and OS. Interestingly, high DLL3 protein expression levels were significantly correlated with PD-L1/2 and negatively correlated with NOTCH1. Therefore, DLL3 could be considered a biomarker for better prognosis in resectable PDAC patients as well as a therapeutic biomarker for immunotherapy response. These facts set a rationale for testing anti-DLL3-based treatments either alone or combined with immunotherapy or other NOTCH1 inhibitors.
Collapse
Affiliation(s)
- Carlos Lacalle-Gonzalez
- Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain;
| | | | - Lara Sanz-Criado
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, 28040 Madrid, Spain; (L.S.-C.); (M.O.O.)
| | - Michael Ochieng’ Otieno
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, 28040 Madrid, Spain; (L.S.-C.); (M.O.O.)
| | - Edurne Ramos-Muñoz
- Biomarkers and Therapeutic Targets Group and Core Facility, RICORS2040, EATRIS, Ramón y Cajal Health Research Institute, (IRYCIS), C/Carretera Colmenar Km 9,100, 28034 Madrid, Spain;
| | - Maria Jesus Fernandez-Aceñero
- Pathology Department, Clinico San Carlos University Hospital, C/Profesor Martin Lagos, 28040 Madrid, Spain; (M.J.F.-A.); (L.O.-M.)
| | - Luis Ortega-Medina
- Pathology Department, Clinico San Carlos University Hospital, C/Profesor Martin Lagos, 28040 Madrid, Spain; (M.J.F.-A.); (L.O.-M.)
| | - Jesus Garcia-Foncillas
- Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, 28040 Madrid, Spain; (L.S.-C.); (M.O.O.)
| | - Javier Martinez-Useros
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, 28040 Madrid, Spain; (L.S.-C.); (M.O.O.)
- Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, 28922 Madrid, Spain
| |
Collapse
|
3
|
Lianyuan T, Deyu L, Haibo Y, Yadong D, Guanjing T. Clinical features and prognostic factors of elderly patients with metastatic pancreatic cancer: a population-based study. Aging (Albany NY) 2021; 13:7133-7146. [PMID: 33639615 PMCID: PMC7993726 DOI: 10.18632/aging.202570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
The aim of this study was to evaluate the clinical features and prognostic factors of elderly patients with metastatic pancreatic cancer. Patients diagnosed with metastatic pancreatic cancer between 2004 and 2014 were identified from the Surveillance Epidemiology and End Results database. Clinical characteristics and prognostic factors in elderly patients with metastatic pancreatic cancer were examined. A total of 10784 metastatic pancreatic cancer patients between 65 and 80 years old were included and divided into three age groups. Elderly metastatic pancreatic cancer patients differed from younger patients in many aspects, including marital status, race, sex, T stage, N stage, treatment regimen, prognosis, cause of death, and metastatic characteristics (P<0.001). An analysis of prognostic factors showed that chemotherapy, as the main treatment for elderly patients, can significantly improve prognosis, while surgery can improve the prognosis of patients between 65 and 80 years old. Other factors, including sex, marital status, T stage, and site of metastasis, had different effects on patients in different age groups. Elderly patients with metastatic pancreatic cancer are a special group of individuals whose clinical characteristics and prognostic factors are different from those of younger patients, and these patients require special treatment and attention.
Collapse
Affiliation(s)
- Tao Lianyuan
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| | - Li Deyu
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| | - Yu Haibo
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| | - Dong Yadong
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| | - Tian Guanjing
- Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| |
Collapse
|
4
|
The Regulatory Properties of the Ccr4-Not Complex. Cells 2020; 9:cells9112379. [PMID: 33138308 PMCID: PMC7692201 DOI: 10.3390/cells9112379] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
The mammalian Ccr4–Not complex, carbon catabolite repression 4 (Ccr4)-negative on TATA-less (Not), is a large, highly conserved, multifunctional assembly of proteins that acts at different cellular levels to regulate gene expression. In the nucleus, it is involved in the regulation of the cell cycle, chromatin modification, activation and inhibition of transcription initiation, control of transcription elongation, RNA export, nuclear RNA surveillance, and DNA damage repair. In the cytoplasm, the Ccr4–Not complex plays a central role in mRNA decay and affects protein quality control. Most of our original knowledge of the Ccr4–Not complex is derived, primarily, from studies in yeast. More recent studies have shown that the mammalian complex has a comparable structure and similar properties. In this review, we summarize the evidence for the multiple roles of both the yeast and mammalian Ccr4–Not complexes, highlighting their similarities.
Collapse
|
5
|
Pizzamiglio L, Focchi E, Antonucci F. ATM Protein Kinase: Old and New Implications in Neuronal Pathways and Brain Circuitry. Cells 2020; 9:E1969. [PMID: 32858941 PMCID: PMC7564642 DOI: 10.3390/cells9091969] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
Despite that the human autosomal recessive disease ataxia telangiectasia (A-T) is a rare pathology, interest in the function of ataxia-telangiectasia mutated protein (ATM) is extensive. From a clinical point of view, the role of ATM in the central nervous system (CNS) is the most impacting, as motor disability is the predominant symptom affecting A-T patients. Coherently, spino-cerebellar neurodegeneration is the principal hallmark of A-T and other CNS regions such as dentate and olivary nuclei and brain stem are implicated in A-T pathophysiology. Recently, several preclinical studies also highlighted the involvement of ATM in the cerebral cortex and hippocampus, thus extending A-T symptomatology to new brain areas and pathways. Here, we review old and recent evidence that largely demonstrates not only the historical ATM account in DNA damage response and cell cycle regulation, but the multiple pathways through which ATM controls oxidative stress homeostasis, insulin signalling pathways, epigenetic regulation, synaptic transmission, and excitatory-inhibitory balance. We also summarise recent evidence on ATM implication in neurological and cognitive diseases beyond A-T, bringing out ATM as new pathological substrate and potential therapeutic target.
Collapse
Affiliation(s)
- Lara Pizzamiglio
- Institute of Molecular and Cellular Pharmacology (IPMC), Université Côte d’Azur (UCA), CNRS UMR7275, 06560 Valbonne-Sophia Antipolis, France;
| | - Elisa Focchi
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, 20100 Milan, Italy;
| | - Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, 20100 Milan, Italy;
| |
Collapse
|
6
|
Wang ST, Huang SW, Liu KT, Lee TY, Shieh JJ, Wu CY. Atorvastatin-induced senescence of hepatocellular carcinoma is mediated by downregulation of hTERT through the suppression of the IL-6/STAT3 pathway. Cell Death Discov 2020; 6:17. [PMID: 32257389 PMCID: PMC7105491 DOI: 10.1038/s41420-020-0252-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 01/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC), a hepatic malignancy, has a poor prognosis and contributes to cancer-related death worldwide. Cellular senescence is an anticancer therapeutic strategy that causes irreversible cell cycle arrest and enables immune-mediated clearance of cancer cells. Atorvastatin, an HMG-CoA reductase inhibitor, has been shown to inhibit tumor growth and induce apoptosis or autophagy in malignant tumors. However, whether atorvastatin can induce HCC cell senescence and the mechanisms involved are poorly understood. The effects of atorvastatin-induced senescence were examined in both HCC cells and mouse xenograft models. The phenomenon and mechanism of senescence were examined by cell cycle analysis, senescence-associated β-galactosidase (SA-β-gal) staining and western blotting in HCC cells, and HCC tissues from mice were analyzed by immunohistochemical (IHC) staining. We demonstrated that atorvastatin induced cell growth inhibition and G0/G1 phase cell cycle arrest, leading to senescence in HCC cells. Atorvastatin-induced senescence was independent of p53, p14, and p16, and atorvastatin not only decreased the secretion of IL-6, a major senescence-associated secretory phenotype (SASP) factor, and the phosphorylation of STAT3 but also inhibited the expression of hTERT, a catalytic subunit of telomerase. Supplementation with exogenous IL-6 reversed both atorvastatin-induced suppression of STAT3 phosphorylation and hTERT expression and atorvastatin-induced senescence. Overexpression of constitutively activated STAT3 rescued HCC cells from atorvastatin-induced hTERT suppression and senescence. Moreover, atorvastatin decreased tumor growth in mouse xenograft models. Consistent with these results, atorvastatin decreased the IL-6, p-STAT3, and hTERT levels and increased β-gal expression in tumor sections. Taken together, these data indicate that atorvastatin can induce atypical cellular senescence in HCC cells to inhibit tumor growth, an effect mediated by downregulation of hTERT through suppression of the IL-6/STAT3 pathway.
Collapse
Affiliation(s)
- Sin-Ting Wang
- Division of Translational Research and Center of Excellence for Cancer Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shi-Wei Huang
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung, Taiwan
| | - Kuang-Ting Liu
- Department of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Pathology & Laboratory Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Teng-Yu Lee
- Division of Gastroenterology and Hepatology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jeng-Jer Shieh
- Department of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Life Sciences and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chun-Ying Wu
- Division of Translational Research and Center of Excellence for Cancer Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Biomedical Informatics, Institute of Clinical Medicine, and Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
- Department of Public Health, China Medical University, Taichung, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
- Taiwan Microbiota Consortium, Taipei, Taiwan
| |
Collapse
|
7
|
Wang Y, Huang T, Li Y, Sha X. The self-organization model reveals systematic characteristics of aging. Theor Biol Med Model 2020; 17:4. [PMID: 32197622 PMCID: PMC7082995 DOI: 10.1186/s12976-020-00120-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/25/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aging is a fundamental biological process, where key bio-markers interact with each other and synergistically regulate the aging process. Thus aging dysfunction will induce many disorders. Finding aging markers and re-constructing networks based on multi-omics data (i.e. methylation, transcriptional and so on) are informative to study the aging process. However, optimizing the model to predict aging have not been performed systemically, although it is critical to identify potential molecular mechanism of aging related diseases. METHODS This paper aims to model the aging self-organization system using a series of supervised learning methods, and study complex molecular mechanisms of aging at system level: i.e. optimizing the aging network; summarizing interactions between aging markers; accumulating patterns of aging markers within module; finding order-parameters in the aging self-organization system. RESULTS In this work, the normal aging process is modeled based on multi-omics profiles across different tissues. In addition, the computational pipeline aims to model aging self-organizing systems and study the relationship between aging and related diseases (i.e. cancers), thus provide useful indicators of aging related diseases and could help to improve prediction abilities of diagnostics. CONCLUSIONS The aging process could be studied thoroughly by modelling the self-organization system, where key functions and the crosstalk between aging and cancers were identified.
Collapse
Affiliation(s)
- Yin Wang
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110012, Liaoning Province, China.,Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Yixue Li
- Bio-Med Big Data Center, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, 200433, China. .,Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China.
| | - Xianzheng Sha
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110012, Liaoning Province, China.
| |
Collapse
|
8
|
Wang S, Wang Q, Wang H, Qin C, Cui X, Li L, Liu Y, Chang H. Induction of ROS and DNA damage-dependent senescence by icaritin contributes to its antitumor activity in hepatocellular carcinoma cells. PHARMACEUTICAL BIOLOGY 2019; 57:424-431. [PMID: 31407933 PMCID: PMC8871611 DOI: 10.1080/13880209.2019.1628073] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/06/2019] [Accepted: 05/21/2019] [Indexed: 05/30/2023]
Abstract
Context: Icaritin (ICT), a prenylflavonoid derivative extracted from the Epimedium (Berberidaceae) genus, has been identified to exhibit antitumor effect in hepatocellular carcinoma (HCC) cells by inducing apoptosis. However, its effect on cellular senescence has not been elucidated. Objective: To investigate the mechanism for low concentrations of ICT exerting antitumor activity through induction of cellular senescence. Materials and methods: Human HepG2 and Huh7 cells were treated with low concentrations of ICT (1 and 2 μM) once per day for a week. Cellular senescence was evaluated through cell viability and senescence-associated-β-galactosidase activity. Cell cycle distribution and ROS levels were measured with flow cytometry. Gene expression was detected using qRT-PCR and western blotting. Fluorescent punctuates formation of γH2AX was analyzed by immunofluorescence. Results: ICT (1 and 2 μM) promoted cellular senescence in HepG2 and Huh7 cells, as observed by enlarged and flattened morphology and increased senescence-associated-β-galactosidase activity (∼7-8-fold and ∼11-12-fold of vehicle controls, respectively), accompanied by significant cell cycle arrest and decrease in DNA synthesis. Mechanistically, ICT-induced senescence occurred through accumulation of ROS (∼1.3-fold and ∼1.8-fold of vehicle controls in response to 1 and 2 μM ICT, respectively), which further resulted in DNA damage response, as evidenced by strong induction of γH2AX through immunofluorescence and western blotting assays. Pharmacological inhibition of ROS production with N-acetylcysteine attenuated ICT-induced γH2AX and senescence-associated-β-galactosidase activity (∼0.28-0.30-fold decrease, p < 0.05). Discussion and conclusions: Induction of cellular senescence by ICT defines a novel anticancer mechanism of ICT and provides a rationale for generalizing the study design to a broader study population to further developing ICT as a novel therapeutic agent for treatment of HCC.
Collapse
Affiliation(s)
- Shikang Wang
- Emergency Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Qian Wang
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Huijun Wang
- Department of Internal Medicine, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, China
| | - Chengkun Qin
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xianping Cui
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lei Li
- Emergency Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yongqing Liu
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, China
| | - Hong Chang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
9
|
Chen J, Huang X, Tao C, Xiao T, Li X, Zeng Q, Ma M, Wu Z. Artemether Attenuates the Progression of Non-small Cell Lung Cancer by Inducing Apoptosis, Cell Cycle Arrest and Promoting Cellular Senescence. Biol Pharm Bull 2019; 42:1720-1725. [PMID: 31378747 DOI: 10.1248/bpb.b19-00391] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Lung cancer is the most common cause of cancer death, approximately 85% of which are non-small cell lung cancer (NSCLC). Here we found that artemether (ART), a natural derivative of artemisinin, significantly inhibits the proliferation of NSCLC cells in a dose- and time-dependent manner. We also demonstrated that high concentration of ART induces apoptosis in NSCLC cells through down-regulating the level of anti-apoptotic protein B-cell lymphoma-2 (Bcl-2), cellular inhibitor of apoptosis protein 1 (cIAP1) and cellular inhibitor of apoptosis protein 2 (cIAP2). While low concentration of ART inhibits the mRNA level of cell cycle related genes including cyclin-dependent kinase 1 (CDK1), cyclin-dependent kinase 2 (CDK2), cyclin-dependent kinase 6 (CDK6), cyclin A2, cyclin B1 and cyclin D1, leading to cell cycle arrest in NSCLC cells. Moreover, we confirmed that low concentration of ART induces DNA double-stranded breaks (DSBs), as well as promoting cellular senescence in NSCLC cells by up-regulating the mRNA and protein level of p16. Taken together, ART represents a promising new anti-NSCLC drug candidate that could attenuate progression of NSCLC cells in a p53-independent manner through inducing apoptosis, cell cycle arrest and promoting cellular senescence.
Collapse
Affiliation(s)
- Jian Chen
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital
- Shenzhen Institute of Geriatrics
| | - Xiaofei Huang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine
| | - Cheng Tao
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital
- Shenzhen Institute of Geriatrics
- Dongguan Institute of Jinan University
| | - Ting Xiao
- Shunde Hospital of Guangzhou University of Chinese Medicine
| | | | - Qiang Zeng
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital
- Shenzhen Institute of Geriatrics
| | - Min Ma
- College of Traditional Chinese Medicine, Jinan University
- The First Affiliated Hospital of Jinan University
| | - Zhengzhi Wu
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital
- Shenzhen Institute of Geriatrics
- The Eighth Affiliated Hospital of Sun Yat-sen University
| |
Collapse
|
10
|
Wang S, Wang H, Sun B, Li D, Wu J, Li J, Tian X, Qin C, Chang H, Liu Y. Acetyl-11-keto-β-boswellic acid triggers premature senescence via induction of DNA damage accompanied by impairment of DNA repair genes in hepatocellular carcinoma cells in vitro and in vivo. Fundam Clin Pharmacol 2019; 34:65-76. [PMID: 31141202 DOI: 10.1111/fcp.12488] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/28/2019] [Accepted: 05/24/2019] [Indexed: 01/25/2023]
Abstract
Cellular senescence, a state of irreversible growth arrest, occurs in all somatic cells and causes the cells to exhaust replicative capacity. Recently, cellular senescence has been emerging as one of the principal mechanisms of tumor suppression, which can be induced by low doses of therapeutic drugs in cancer cells. Acetyl-11-keto-β-boswellic acid (AKBA), an active ingredient isolated from the plant Boswellia serrata, has been identified to induce apoptosis in hepatocellular carcinoma (HCC) cells. In this study, we found that low concentrations of AKBA treatment triggered cell growth arrest at G0/G1 phase with features of premature cellular senescence phenotype in both HCC cell lines HepG2 and SMMC7721, as observed by enlarged and flattened morphology, significant increase in cells with senescence-associated β-galactosidase staining, and decrease in cell proliferation and DNA synthesis. Furthermore, cellular senescence induced by AKBA occurred via activation of DNA damage response and impairment of DNA repair, as evidenced by strong induction of γH2AX and p53, and downregulated expressions of multiple DNA repair associated genes. Induction of p53 by AKBA caused a significant increase in p21CIP1 , which had a critical involvement in the induction of cellular senescence. Additionally, in vivo study demonstrated that induction of senescence contributed to the anticancer efficacy of AKBA. Therefore, our findings suggested that induction of premature senescence by AKBA through DNA damage response accompanied by impairment of DNA repair genes defines a novel mechanism contributing to its growth suppression in HCC cells.
Collapse
Affiliation(s)
- Shikang Wang
- Emergency Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China.,Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Huijun Wang
- Department of Internal Medicine, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Baoyou Sun
- Emergency Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Duanfeng Li
- Emergency Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Jing Wu
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Juan Li
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Xiaona Tian
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| | - Chengkun Qin
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Hong Chang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Yongqing Liu
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, 250033, Shandong, China
| |
Collapse
|
11
|
Hofman P, Ayache N, Barbry P, Barlaud M, Bel A, Blancou P, Checler F, Chevillard S, Cristofari G, Demory M, Esnault V, Falandry C, Gilson E, Guérin O, Glaichenhaus N, Guigay J, Ilié M, Mari B, Marquette CH, Paquis-Flucklinger V, Prate F, Saintigny P, Seitz-Polsky B, Skhiri T, Van Obberghen-Schilling E, Van Obberghen E, Yvan-Charvet L. The OncoAge Consortium: Linking Aging and Oncology from Bench to Bedside and Back Again. Cancers (Basel) 2019; 11:E250. [PMID: 30795607 PMCID: PMC6406685 DOI: 10.3390/cancers11020250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/17/2019] [Accepted: 02/19/2019] [Indexed: 01/04/2023] Open
Abstract
It is generally accepted that carcinogenesis and aging are two biological processes, which are known to be associated. Notably, the frequency of certain cancers (including lung cancer), increases significantly with the age of patients and there is now a wealth of data showing that multiple mechanisms leading to malignant transformation and to aging are interconnected, defining the so-called common biology of aging and cancer. OncoAge, a consortium launched in 2015, brings together the multidisciplinary expertise of leading public hospital services and academic laboratories to foster the transfer of scientific knowledge rapidly acquired in the fields of cancer biology and aging into innovative medical practice and silver economy development. This is achieved through the development of shared technical platforms (for research on genome stability, (epi)genetics, biobanking, immunology, metabolism, and artificial intelligence), clinical research projects, clinical trials, and education. OncoAge focuses mainly on two pilot pathologies, which benefit from the expertise of several members, namely lung and head and neck cancers. This review outlines the broad strategic directions and key advances of OncoAge and summarizes some of the issues faced by this consortium, as well as the short- and long-term perspectives.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology/Biobank 0033-00025, CHU Nice, FHU OncoAge, Université Côte d'Azur, 06001 Nice, France.
- Inserm U1081, CNRS UMR7284, Institut de Recherche sur le Cancer et le Vieillissement (IRCAN), FHU OncoAge, Université Côte d'Azur, 06107 Nice, France.
| | - Nicholas Ayache
- Epione Team, Inria, FHU OncoAge, Université Côte d'Azur, 06902 Sophia Antipolis, France.
| | - Pascal Barbry
- CNRS UMR7275, Institut de Pharmacologie Cellulaire et Moléculaire, FHU OncoAge, Université Côte d'Azur, 06560 Valbonne, France.
| | - Michel Barlaud
- i3S Sophia Antipolis, FHU OncoAge, Université Côte d'Azur, 06560 Sophia Antipolis, France.
| | - Audrey Bel
- Centre d'Innovation et d'Usages en Santé (CIUS), FHU OncoAge, Université Côte d'Azur, 06000 Nice, France.
| | - Philippe Blancou
- CNRS UMR7275, Institut de Pharmacologie Cellulaire et Moléculaire, FHU OncoAge, Université Côte d'Azur, 06560 Valbonne, France.
| | - Frédéric Checler
- CNRS UMR7275, Institut de Pharmacologie Cellulaire et Moléculaire, FHU OncoAge, Université Côte d'Azur, 06560 Valbonne, France.
| | - Sylvie Chevillard
- Laboratoire de Cancérologie Expérimentale, Institut François Jacob, CEA Direction de la Recherche Fondamentale, FHU OncoAge, Université Côte d'Azur, 92265 Fontenay-aux-Roses, France.
| | - Gael Cristofari
- Inserm U1081, CNRS UMR7284, Institut de Recherche sur le Cancer et le Vieillissement (IRCAN), FHU OncoAge, Université Côte d'Azur, 06107 Nice, France.
| | - Mathilde Demory
- Ville de Nice, Mairie de Nice, FHU OncoAge, Université Côte d'Azur, 06364 Nice, France.
| | - Vincent Esnault
- Nephrology Department, CHU Nice, FHU OncoAge, Université Côte d'Azur, 06001 Nice, France.
| | - Claire Falandry
- Geriatric Unit, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, FHU OncoAge, Université Claude Bernard Lyon 1, 69310 Pierre-Benite, France.
- Laboratoire CarMeN, Inserm U1060, INRA U139, INSA Lyon, Ecole de Médecine Charles Mérieux, Université Claude Bernard Lyon 1, 69921 Oullins, France.
| | - Eric Gilson
- Inserm U1081, CNRS UMR7284, Institut de Recherche sur le Cancer et le Vieillissement (IRCAN), FHU OncoAge, Université Côte d'Azur, 06107 Nice, France.
| | - Olivier Guérin
- Geriatric Coordination Unit for Geriatric Oncology (UCOG) PACA Est, CHU Nice, FHU OncoAge, Université Côte d'Azur, 06000 Nice, France.
| | - Nicolas Glaichenhaus
- CNRS UMR7275, Institut de Pharmacologie Cellulaire et Moléculaire, FHU OncoAge, Université Côte d'Azur, 06560 Valbonne, France.
| | - Joel Guigay
- Oncology Department, Centre Antoine Lacassagne, FHU OncoAge, Université Côté d'Azur, 06189 Nice, France.
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology/Biobank 0033-00025, CHU Nice, FHU OncoAge, Université Côte d'Azur, 06001 Nice, France.
- Inserm U1081, CNRS UMR7284, Institut de Recherche sur le Cancer et le Vieillissement (IRCAN), FHU OncoAge, Université Côte d'Azur, 06107 Nice, France.
| | - Bernard Mari
- CNRS UMR7275, Institut de Pharmacologie Cellulaire et Moléculaire, FHU OncoAge, Université Côte d'Azur, 06560 Valbonne, France.
| | - Charles-Hugo Marquette
- Department of Pulmonary Medicine and Oncology, CHU Nice, FHU OncoAge, Université Côte d'Azur, 06000 Nice, France.
| | - Véronique Paquis-Flucklinger
- Inserm U1081, CNRS UMR7284, Institut de Recherche sur le Cancer et le Vieillissement (IRCAN), FHU OncoAge, Université Côte d'Azur, 06107 Nice, France.
| | - Frédéric Prate
- Geriatric Coordination Unit for Geriatric Oncology (UCOG) PACA Est, CHU Nice, FHU OncoAge, Université Côte d'Azur, 06000 Nice, France.
| | - Pierre Saintigny
- Département de Médecine, INSERM 1052, CNRS 5286, Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, FHU OncoAge, Université Claude Bernard Lyon 1, 69008 Lyon, France.
| | - Barbara Seitz-Polsky
- CNRS UMR7275, Institut de Pharmacologie Cellulaire et Moléculaire, FHU OncoAge, Université Côte d'Azur, 06560 Valbonne, France.
- Laboratory of Immunology, CHU Nice, FHU OncoAge, Université Côte d'Azur, 06200 Nice, France.
| | - Taycir Skhiri
- Centre d'Innovation et d'Usages en Santé (CIUS), FHU OncoAge, Université Côte d'Azur, 06000 Nice, France.
| | | | | | - Laurent Yvan-Charvet
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), FHU OncoAge, Université Côte d'Azur, 06200 Nice, France.
| |
Collapse
|
12
|
Skórkowska-Telichowska K, Kulma A, Gębarowski T, Wojtasik W, Kostyn K, Moreira H, Szyjka A, Boba A, Preisner M, Mierziak J, Arendt M, Kostyn A, Szatkowski M, Szopa J, Gąsiorowski K. V79 Fibroblasts Are Protected Against Reactive Oxygen Species by Flax Fabric. Appl Biochem Biotechnol 2017; 184:366-385. [PMID: 28689336 DOI: 10.1007/s12010-017-2552-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/27/2017] [Indexed: 01/22/2023]
Abstract
Chinese hamster pulmonary fibroblasts (V79 cells) pre-treated with flax fabrics derived from non-modified or genetically engineered flax fibres and treated with H2O2 revealed a markedly lower level of intracellular reactive oxygen species (ROS) than control, non-pre-treated cells. The fabrics were prepared from fibres derived from two kinds of transgenic plants: W92 plants, which overproduce flavonoids, and M type plants, which produce hydroxybutyrate polymer in their vascular bundles and thus in fibres. Incubating the V79 cells with the flax fabrics prior to H2O2 treatment also reduced the amount of DNA damage, as established using the comet assay (also known as alkaline single-cell gel electrophoresis) and pulsed-field electrophoresis of intact cellular DNA. Selected gene expression analysis revealed the activator impact of fabrics on the apoptotic (BCL2 family, caspases) gene expression. This promoting activity was also detected for histone acetyltransferase (HAT; MYST2) gene expression. The flax fabric derived from both GM flax plants exhibited a protective effect against oxidative stress and ROS-mediated genotoxic damage, but the W92 fabric was the strongest. It is thus suggested that these fabrics might be useful as a basis for new biomedical products (e.g. wound dressings) that actively protect cells against inflammation and degeneration.
Collapse
Affiliation(s)
- Katarzyna Skórkowska-Telichowska
- Angiology Department, Regional Specialist Hospital in Wroclaw, Research and Development Centre, Kamienskiego 73a St, 51-124, Wroclaw, Poland
| | - Anna Kulma
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland
| | - Tomasz Gębarowski
- Department of Basic Medical Sciences, Wroclaw Medical University, Wroclaw, Poland
| | - Wioleta Wojtasik
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland
| | - Kamil Kostyn
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland.
| | - Helena Moreira
- Department of Basic Medical Sciences, Wroclaw Medical University, Wroclaw, Poland
| | - Anna Szyjka
- Department of Basic Medical Sciences, Wroclaw Medical University, Wroclaw, Poland
| | - Aleksandra Boba
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland
| | - Marta Preisner
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland
| | - Justyna Mierziak
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland
| | - Malgorzata Arendt
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland
| | - Anna Kostyn
- Institute of Genetics and Microbiology, University of Wroclaw, Przybyszewskiego 63/77, 51-148, Wroclaw, Poland
| | - Michał Szatkowski
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland
| | - Jan Szopa
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63, 51-137, Wroclaw, Poland.,Department of Genetics, Plant Breeding and Seed Production, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | | |
Collapse
|
13
|
Hochberg ME, Noble RJ. A framework for how environment contributes to cancer risk. Ecol Lett 2017; 20:117-134. [DOI: 10.1111/ele.12726] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/03/2016] [Accepted: 12/01/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Michael E. Hochberg
- Intstitut des Sciences de l'Evolution de Montpellier; Université de Montpellier; Place E. Bataillon, CC065 34095 Montpellier Cedex 5 France
- Santa Fe Institute; 1399 Hyde Park Rd. Santa Fe NM 87501 USA
| | - Robert J. Noble
- Intstitut des Sciences de l'Evolution de Montpellier; Université de Montpellier; Place E. Bataillon, CC065 34095 Montpellier Cedex 5 France
| |
Collapse
|
14
|
Wang Y, Huang T, Xie L, Liu L. Integrative analysis of methylation and transcriptional profiles to predict aging and construct aging specific cross-tissue networks. BMC SYSTEMS BIOLOGY 2016; 10:132. [PMID: 28155676 PMCID: PMC5260078 DOI: 10.1186/s12918-016-0354-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Aging is a complex process relating multi-scale omics data. Finding key age markers in normal tissues could help to provide reliable aging predictions in human. However, predicting age based on multi-omics data with both accuracy and informative biological function has not been performed systematically, thus relative cross-tissue analysis has not been investigated entirely, either. RESULTS Here we have developed an improved prediction pipeline, the Integrating and Stepwise Age-Prediction (ISAP) method, to regress age and find key aging markers effectively. Furthermore, we have performed a serious of network analyses, such as the PPI network, cross-tissue networks and pathway interaction networks. CONCLUSION Our results find important coordinated aging patterns between different tissues. Both co-profiling and cross-pathway analyses identify more thorough functions of aging, and could help to find aging markers, pathways and relative aging disease researches.
Collapse
Affiliation(s)
- Yin Wang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Lu Xie
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China
| | - Lei Liu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai, 201203, China.
| |
Collapse
|
15
|
Chakraborty S, Rasool RU, Kumar S, Nayak D, Rah B, Katoch A, Amin H, Ali A, Goswami A. Cristacarpin promotes ER stress-mediated ROS generation leading to premature senescence by activation of p21(waf-1). AGE (DORDRECHT, NETHERLANDS) 2016; 38:62. [PMID: 27246693 PMCID: PMC5005927 DOI: 10.1007/s11357-016-9922-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 05/09/2016] [Indexed: 06/05/2023]
Abstract
Stress-induced premature senescence (SIPS) is quite similar to replicative senescence that is committed by cells exposed to various stress conditions viz. ultraviolet radiation (DNA damage), hydrogen peroxide (oxidative stress), chemotherapeutic agents (cytotoxic threat), etc. Here, we report that cristacarpin, a natural product obtained from the stem bark of Erythrina suberosa, promotes endoplasmic reticulum (ER) stress, leading to sub-lethal reactive oxygen species (ROS) generation and which eventually terminates by triggering senescence in pancreatic and breast cancer cells through blocking the cell cycle in the G1 phase. The majority of cristacarpin-treated cells responded to conventional SA-β-gal stains; showed characteristic p21(waf1) upregulation along with enlarged and flattened morphology; and increased volume, granularity, and formation of heterochromatin foci-all of these features are the hallmarks of senescence. Inhibition of ROS generation by N-acetyl-L-cysteine (NAC) significantly reduced the expression of p21(waf1), confirming that the modulation in p21(waf1) by anti-proliferative cristacarpin was ROS dependent. Further, the elevation in p21(waf1) expression in PANC-1 and MCF-7 cells was consistent with the decrease in the expression of Cdk-2 and cyclinD1. Here, we provide evidence that cristacarpin promotes senescence in a p53-independent manner. Moreover, cristacarpin treatment induced p38MAPK, indicating the ROS-dependent activation of the MAP kinase pathway, and thus abrogates the tumor growth in mouse allograft tumor model.
Collapse
Affiliation(s)
- Souneek Chakraborty
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India
| | - Reyaz Ur Rasool
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India
| | - Sunil Kumar
- Natural Product Chemistry Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India
| | - Debasis Nayak
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India
| | - Bilal Rah
- University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Archana Katoch
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India
| | - Hina Amin
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India
| | - Asif Ali
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Natural Product Chemistry Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India
| | - Anindya Goswami
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India.
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K, 180001, India.
| |
Collapse
|
16
|
Higuera O, Ghanem I, Nasimi R, Prieto I, Koren L, Feliu J. Management of pancreatic cancer in the elderly. World J Gastroenterol 2016; 22:764-75. [PMID: 26811623 PMCID: PMC4716075 DOI: 10.3748/wjg.v22.i2.764] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/03/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023] Open
Abstract
Currently, pancreatic adenocarcinoma mainly occurs after 60 years of age, and its prognosis remains poor despite modest improvements in recent decades. The aging of the population will result in a rise in the incidence of pancreatic adenocarcinoma within the next years. Thus, the management of pancreatic cancer in the elderly population is gaining increasing relevance. Older cancer patients represent a heterogeneous group with different biological, functional and psychosocial characteristics that can modify the usual management of this disease, including pharmacokinetic and pharmacodynamic changes, polypharmacy, performance status, comorbidities and organ dysfunction. However, the biological age, not the chronological age, of the patient should be the limiting factor in determining the most appropriate treatment for these patients. Unfortunately, despite the increased incidence of this pathology in older patients, there is an underrepresentation of these patients in clinical trials, and the management of older patients is thus determined by extrapolation from the results of studies performed in younger patients. In this review, the special characteristics of the elderly, the multidisciplinary management of localized and advanced ductal adenocarcinoma of the pancreas and the most recent advances in the management of this condition will be discussed, focusing on surgery, chemotherapy, radiation and palliative care.
Collapse
|
17
|
Abstract
Aging is a progressive process determined by genetic and acquired factors. Among the latter are the chemical reactions referred to as nonenzymatic posttranslational modifications (NEPTMs), such as glycoxidation, which are responsible for protein molecular aging. Carbamylation is a more recently described NEPTM that is caused by the nonenzymatic binding of isocyanate derived from urea dissociation or myeloperoxidase-mediated catabolism of thiocyanate to free amino groups of proteins. This modification is considered an adverse reaction, because it induces alterations of protein and cell properties. It has been shown that carbamylated proteins increase in plasma and tissues during chronic kidney disease and are associated with deleterious clinical outcomes, but nothing is known to date about tissue protein carbamylation during aging. To address this issue, we evaluated homocitrulline rate, the most characteristic carbamylation-derived product (CDP), over time in skin of mammalian species with different life expectancies. Our results show that carbamylation occurs throughout the whole lifespan and leads to tissue accumulation of carbamylated proteins. Because of their remarkably long half-life, matrix proteins, like type I collagen and elastin, are preferential targets. Interestingly, the accumulation rate of CDPs is inversely correlated with longevity, suggesting the occurrence of still unidentified protective mechanisms. In addition, homocitrulline accumulates more intensely than carboxymethyl-lysine, one of the major advanced glycation end products, suggesting the prominent role of carbamylation over glycoxidation reactions in age-related tissue alterations. Thus, protein carbamylation may be considered a hallmark of aging in mammalian species that may significantly contribute in the structural and functional tissue damages encountered during aging.
Collapse
|
18
|
Biomolecular bases of the senescence process and cancer. A new approach to oncological treatment linked to ageing. Ageing Res Rev 2015; 23:125-38. [PMID: 25847820 DOI: 10.1016/j.arr.2015.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/30/2015] [Indexed: 01/07/2023]
Abstract
Human ageing is associated with a gradual decline in the physiological functions of the body at multiple levels and it is a key risk factor for many diseases, including cancer. Ageing process is intimately related to widespread cellular senescence, characterised by an irreversible loss of proliferative capacity and altered functioning associated with telomere attrition, accumulation of DNA damage and compromised mitochondrial and metabolic function. Tumour and senescent cells may be generated in response to the same stimuli, where either cellular senescence or transformation would constitute two opposite outcomes of the same degenerative process. This paper aims to review the state of knowledge on the biomolecular relationship between cellular senescence, ageing and cancer. Importantly, many of the cell signalling pathways that are found to be altered during both cellular senescence and tumourigenesis are regulated through shared epigenetic mechanisms and, therefore, they are potentially reversible. MicroRNAs are emerging as pivotal players linking ageing and cancer. These small RNA molecules have generated great interest from the point of view of future clinical therapy for cancer because successful experimental results have been obtained in animal models. Micro-RNA therapies for cancer are already being tested in clinical phase trials. These findings have potential importance in cancer treatment in aged people although further research-based knowledge is needed to convert them into an effective molecular therapies for cancer linked to ageing.
Collapse
|
19
|
Zhang XW, Wang XF, Ni SJ, Qin W, Zhao LQ, Hua RX, Lu YW, Li J, Dimri GP, Guo WJ. UBTD1 induces cellular senescence through an UBTD1-Mdm2/p53 positive feedback loop. J Pathol 2015; 235:656-67. [PMID: 25382750 DOI: 10.1002/path.4478] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 09/22/2014] [Accepted: 11/04/2014] [Indexed: 01/13/2023]
Abstract
The tumour suppressor p53 plays an important role in tumourigenesis. Besides inducing apoptosis, it regulates cellular senescence, which constitutes an important barrier to tumourigenesis. The mechanism of regulation of cellular senescence by p53 and its downstream pathway are poorly understood. Here, we report that the ubiquitin domain-containing 1 (UBTD1) gene, a new downstream target of p53, induces cellular senescence and acts as a novel tumour suppressor by a mechanism that depends on p53. Expression of UBTD1 increased upon cellular senescence induced by serial passageing of cultures, as well as by exposure to DNA-damageing drugs that induce premature senescence. Over-expression of UBTD1 induces senescence in human fibroblasts and cancer cells and attenuation of the transformed phenotype in cancer cells. UBTD1 is down-regulated in gastric and colorectal cancer tissues, and its lower expression correlates with a more aggressive phenotype and worse prognosis. Multivariate analysis revealed that UBTD1 expression was an independent prognostic factor for gastric cancer patients. Furthermore, UBTD1 increased the stability of p53 protein, by promoting the degradation of Mdm2 protein. Importantly, UBTD1 and p53 function mutually depend on each other in regulating cellular senescence and proliferation. Thus, our data suggest that, upon DNA damage, p53 induction by UBTD1 creates a positive feedback mechanism to further increase p53 expression. Our results establish UBTD1 as a regulator of cellular senescence that mediates p53 function, and provide insights into the mechanism of Mdm2 inhibition that impacts p53 dynamics during cellular senescence and tumourigenesis.
Collapse
Affiliation(s)
- Xiao-Wei Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ortiz-Lazareno PC, Bravo-Cuellar A, Lerma-Díaz JM, Jave-Suárez LF, Aguilar-Lemarroy A, Domínguez-Rodríguez JR, González-Ramella O, De Célis R, Gómez-Lomelí P, Hernández-Flores G. Sensitization of U937 leukemia cells to doxorubicin by the MG132 proteasome inhibitor induces an increase in apoptosis by suppressing NF-kappa B and mitochondrial membrane potential loss. Cancer Cell Int 2014; 14:13. [PMID: 24495648 PMCID: PMC3927225 DOI: 10.1186/1475-2867-14-13] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 01/21/2014] [Indexed: 12/18/2022] Open
Abstract
Background The resistance of cancerous cells to chemotherapy remains the main limitation for cancer treatment at present. Doxorubicin (DOX) is a potent antitumor drug that activates the ubiquitin-proteasome system, but unfortunately it also activates the Nuclear factor kappa B (NF-кB) pathway leading to the promotion of tumor cell survival. MG132 is a drug that inhibits I kappa B degradation by the proteasome-avoiding activation of NF-кB. In this work, we studied the sensitizing effect of the MG132 proteasome inhibitor on the antitumor activity of DOX. Methods U937 human leukemia cells were treated with MG132, DOX, or both drugs. We evaluated proliferation, viability, apoptosis, caspase-3, -8, and −9 activity and cleavage, cytochrome c release, mitochondrial membrane potential, the Bcl-2 and Bcl-XL antiapoptotic proteins, senescence, p65 phosphorylation, and pro- and antiapoptotic genes. Results The greatest apoptosis percentage in U937 cells was obtained with a combination of MG132 + DOX. Likewise, employing both drugs, we observed a decrease in tumor cell proliferation and important caspase-3 activation, as well as mitochondrial membrane potential loss. Therefore, MG132 decreases senescence, p65 phosphorylation, and the DOX-induced Bcl-2 antiapoptotic protein. The MG132 + DOX treatment induced upregulation of proapoptotic genes BAX, DIABLO, NOXA, DR4, and FAS. It also induced downregulation of the antiapoptotic genes BCL-XL and SURVIVIN. Conclusion MG132 sensitizes U937 leukemia cells to DOX-induced apoptosis, increasing its anti-leukemic effectiveness.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Georgina Hernández-Flores
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México.
| |
Collapse
|
21
|
Induction of DNA damage and p21-dependent senescence by Riccardin D is a novel mechanism contributing to its growth suppression in prostate cancer cells in vitro and in vivo. Cancer Chemother Pharmacol 2013; 73:397-407. [DOI: 10.1007/s00280-013-2365-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/29/2013] [Indexed: 12/28/2022]
|
22
|
Yildiz G, Arslan-Ergul A, Bagislar S, Konu O, Yuzugullu H, Gursoy-Yuzugullu O, Ozturk N, Ozen C, Ozdag H, Erdal E, Karademir S, Sagol O, Mizrak D, Bozkaya H, Ilk HG, Ilk O, Bilen B, Cetin-Atalay R, Akar N, Ozturk M. Genome-wide transcriptional reorganization associated with senescence-to-immortality switch during human hepatocellular carcinogenesis. PLoS One 2013; 8:e64016. [PMID: 23691139 PMCID: PMC3655073 DOI: 10.1371/journal.pone.0064016] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/07/2013] [Indexed: 01/11/2023] Open
Abstract
Senescence is a permanent proliferation arrest in response to cell stress such as DNA damage. It contributes strongly to tissue aging and serves as a major barrier against tumor development. Most tumor cells are believed to bypass the senescence barrier (become “immortal”) by inactivating growth control genes such as TP53 and CDKN2A. They also reactivate telomerase reverse transcriptase. Senescence-to-immortality transition is accompanied by major phenotypic and biochemical changes mediated by genome-wide transcriptional modifications. This appears to happen during hepatocellular carcinoma (HCC) development in patients with liver cirrhosis, however, the accompanying transcriptional changes are virtually unknown. We investigated genome-wide transcriptional changes related to the senescence-to-immortality switch during hepatocellular carcinogenesis. Initially, we performed transcriptome analysis of senescent and immortal clones of Huh7 HCC cell line, and identified genes with significant differential expression to establish a senescence-related gene list. Through the analysis of senescence-related gene expression in different liver tissues we showed that cirrhosis and HCC display expression patterns compatible with senescent and immortal phenotypes, respectively; dysplasia being a transitional state. Gene set enrichment analysis revealed that cirrhosis/senescence-associated genes were preferentially expressed in non-tumor tissues, less malignant tumors, and differentiated or senescent cells. In contrast, HCC/immortality genes were up-regulated in tumor tissues, or more malignant tumors and progenitor cells. In HCC tumors and immortal cells genes involved in DNA repair, cell cycle, telomere extension and branched chain amino acid metabolism were up-regulated, whereas genes involved in cell signaling, as well as in drug, lipid, retinoid and glycolytic metabolism were down-regulated. Based on these distinctive gene expression features we developed a 15-gene hepatocellular immortality signature test that discriminated HCC from cirrhosis with high accuracy. Our findings demonstrate that senescence bypass plays a central role in hepatocellular carcinogenesis engendering systematic changes in the transcription of genes regulating DNA repair, proliferation, differentiation and metabolism.
Collapse
Affiliation(s)
- Gokhan Yildiz
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- INSERM - Université Joseph Fourrier, CRI U823, Grenoble, France
| | - Ayca Arslan-Ergul
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Sevgi Bagislar
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- INSERM - Université Joseph Fourrier, CRI U823, Grenoble, France
| | - Ozlen Konu
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Haluk Yuzugullu
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- INSERM - Université Joseph Fourrier, CRI U823, Grenoble, France
| | - Ozge Gursoy-Yuzugullu
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- INSERM - Université Joseph Fourrier, CRI U823, Grenoble, France
| | - Nuri Ozturk
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Cigdem Ozen
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Hilal Ozdag
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Esra Erdal
- Department of Medical Biology, Dokuz Eylul University Medical School, Izmir, Turkey
| | - Sedat Karademir
- Department of Surgery, Dokuz Eylul University Medical School, Izmir, Turkey
| | - Ozgul Sagol
- Department of Pathology, Dokuz Eylul University Medical School, Izmir, Turkey
| | - Dilsa Mizrak
- Department of Gastroenterology, Ankara University, Ankara, Turkey
| | - Hakan Bozkaya
- Department of Gastroenterology, Ankara University, Ankara, Turkey
| | - Hakki Gokhan Ilk
- Department of Electronic Engineering, Ankara University, Ankara, Turkey
| | - Ozlem Ilk
- Department of Statistics, Middle East Technical University, Ankara, Turkey
| | - Biter Bilen
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Rengul Cetin-Atalay
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Nejat Akar
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Mehmet Ozturk
- BilGen Genetics and Biotechnology Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- INSERM - Université Joseph Fourrier, CRI U823, Grenoble, France
- * E-mail:
| |
Collapse
|
23
|
Chang H, Rha SY, Jeung HC, Park KH, Kim TS, Kim YB, Chung HC. Telomerase- and angiogenesis-related gene responses to irradiation in human umbilical vein endothelial cells. Int J Mol Med 2013; 31:1202-8. [PMID: 23503666 DOI: 10.3892/ijmm.2013.1300] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 03/05/2013] [Indexed: 11/06/2022] Open
Abstract
In this study, we investigated the effects of ionizing radiation (IR) on human umbilical vein endothelial cells (HUVECs) in the context of senescence. HUVECs at passage number (PN)1, PN2 and PN3 were exposed to irradiation (2 Gy). The growth rate of the HUVECS was measured by proliferation assay and senescence-associated β-galactosidase assay was used to measure the number of senescent cells. Telomerase activity and the expression of telomerase- and angiogenesis-related genes were measured by telomerase assay and real-time PCR, respectively. The number of senescent cells was significantly increased in the irradiated HUVECs at all PNs. Compared to the controls, telomerase activity, the expression of human telomerase reverse transcriptase (hTERT) and c-Myc in the irradiated HUVECs were downregulated during serial passage. The downregulation of vascular endothelial growth factor (VEGF) was observed in the irradiated HUVECs as the PN increased. The data presented in this study may aid in the understanding of the mechanisms behind IR‑induced EC senescence and telomerase- and angiogenesis‑related gene response.
Collapse
Affiliation(s)
- Hyun Chang
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seodaemun‑gu, Seoul 120-752, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
PURPOSE/OBJECTIVES To introduce a conceptual model detailing the physiologic contributions of malglycemia to cancer formation and increased morbidity and mortality. DATA SOURCES A literature search was conducted using the PubMed, CINAHL®, and Cochrane databases, as well as Surveillance, Epidemiology and End Results (SEER) cancer statistics. DATA SYNTHESIS Multiple complex factors are associated with malignancy formation, proliferation, and outcomes for each individual. The authors present a model, termed the Malglycemia Orbit Model, that is analogous to an atom, centered on a core of individual factors, and surrounded by "orbits" containing cancer and related factors. Highlighted in this model is the role of malglycemia. CONCLUSIONS Cancer formation and sequelae involve numerous multifaceted factors. One factor not well described or understood within the context of malignancies is glycemic status, most notably how malglycemia impacts cancer formation and risks for adverse outcomes. The atomic-structured malglycemia model describes this process. IMPLICATIONS FOR NURSING Among the many uncontrollable factors that contribute to cancer formation and adverse outcomes, malglycemia is one that is modifiable. Nurses are in a prime position to conduct research to enhance understanding and ultimately improve protocols for better glycemic control and, in effect, better outcomes for individuals with cancer.
Collapse
|
25
|
Goldberg AA, Beach A, Davies GF, Harkness TAA, Leblanc A, Titorenko VI. Lithocholic bile acid selectively kills neuroblastoma cells, while sparing normal neuronal cells. Oncotarget 2012; 2:761-82. [PMID: 21992775 PMCID: PMC3248158 DOI: 10.18632/oncotarget.338] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aging is one of the major risk factors of cancer. The onset of cancer can be postponed by pharmacological and dietary anti-aging interventions. We recently found in yeast cellular models of aging that lithocholic acid (LCA) extends longevity. Here we show that, at concentrations that are not cytotoxic to primary cultures of human neurons, LCA kills the neuroblastoma (NB) cell lines BE(2)-m17, SK-n-SH, SK-n-MCIXC and Lan-1. In BE(2)-m17, SK-n-SH and SK-n-MCIXC cells, the LCA anti-tumor effect is due to apoptotic cell death. In contrast, the LCA-triggered death of Lan-1 cells is not caused by apoptosis. While low concentrations of LCA sensitize BE(2)-m17 and SK-n-MCIXC cells to hydrogen peroxide-induced apoptotic cell death controlled by mitochondria, these LCA concentrations make primary cultures of human neurons resistant to such a form of cell death. LCA kills BE(2)-m17 and SK-n-MCIXC cell lines by triggering not only the intrinsic (mitochondrial) apoptotic cell death pathway driven by mitochondrial outer membrane permeabilization and initiator caspase-9 activation, but also the extrinsic (death receptor) pathway of apoptosis involving activation of the initiator caspase-8. Based on these data, we propose a mechanism underlying a potent and selective anti-tumor effect of LCA in cultured human NB cells. Moreover, our finding that LCA kills cultured human breast cancer and rat glioma cells implies that it has a broad anti-tumor effect on cancer cells derived from different tissues and organisms.
Collapse
|
26
|
Ivanković M, Cukušić Kalajžić A, Skrobot Vidaček N, Franić Šimić I, Davidović Mrsić S, Rubelj I. Human Xp/Yp telomere analysis by Southern-STELA. Biogerontology 2011; 13:203-13. [PMID: 22143823 DOI: 10.1007/s10522-011-9368-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/24/2011] [Indexed: 10/15/2022]
Abstract
Telomeres are specialized structures designed to protect the ends of linear chromosomes. They are dynamic structures such that in normal somatic cells they constantly shorten as cell division progresses. There is compelling evidence that telomere shortening leads to cell senescence, a process perceived as the main cause of aging in higher mammals. Therefore, the features of telomere shortening are of great importance in understanding cell senescence and aging in general. By identifying unique subtelomeric regions, large enough to produce strong chemiluminescent signals, we have provided a new tool for Southern blot analysis of individual human Xp/Yp telomeres. We extend these results with quantitative fluorescence in situ hybridization using peptide nucleic acid probe (PNA Q-FISH) analysis of telomeres on the Y chromosome. Our results demonstrates unequal shortening dynamics between the p and q telomeres.
Collapse
Affiliation(s)
- Milena Ivanković
- Laboratory for Molecular and Cell Biology, Department of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | | | | | | | | | | |
Collapse
|
27
|
Zhao H, Halicka HD, Traganos F, Jorgensen E, Darzynkiewicz Z. New biomarkers probing depth of cell senescence assessed by laser scanning cytometry. Cytometry A 2010; 77:999-1007. [PMID: 20939035 PMCID: PMC2977923 DOI: 10.1002/cyto.a.20983] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 09/14/2010] [Accepted: 09/15/2010] [Indexed: 12/31/2022]
Abstract
The imaging analytical capabilities of laser scanning cytometer (LSC) have been used to assess morphological features considered to be typical of the senescent phenotype. The characteristic "flattening" of senescent cells was reflected by the decline in the density of staining (intensity of maximal pixel) of DNA-associated fluorescence [4,6-diamidino-2-phenylindole (DAPI)] paralleled by an increase in nuclear size (area). The decrease in ratio of maximal pixel to nuclear area was even more sensitive senescence biomarker than the change in maximal pixel or nuclear area, each alone. The saturation cell density at plateau phase of growth recorded by LSC was found to be dramatically decreased in cultures of senescent cells, thereby also serving as an additional marker. The induction of cyclin dependent kinase inhibitors p21(WAF1) and p27(KIP1) and γH2AX and activation of ATM markers of DNA damage response were measured in parallel with DNA/DAPI maximal pixel and nuclear area. These biomarker indices were expressed in quantitative terms by reporting them as a fraction of the respective controls. The effect of treatment of A549 and WI-38 cells with different concentrations of mitoxantrone (Mxt) and trichostatin A for various time periods was studied to assess the degree (depth) of cell senescence. Also assessed was the effect of 2-deoxy-D-glucose, the agent attenuating metabolic cell activity, on the depth of senescence induced by Mxt. A relationship between the ability of cells to synthesize RNA (incorporate 5-ethynyluridine) that leads to growth imbalance and induction of cell senescence was also studied. The data show that morphometric analysis of cellular attributes by LSC offers an attractive tool to detect cell senescence and measure its degree particularly in assessing effects of the factors that enhance or attenuate this process. This methodology is of importance in light of the evidence that cellular senescence is not only a biological process that is fundamental for organismal aging but also impedes formation of induced-pluripotent stem cells providing the barrier for neoplastic transformation and is the major mechanism of induction of reproductive cell death during treatment of solid tumors.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | |
Collapse
|
28
|
Kanu N, Penicud K, Hristova M, Wong B, Irvine E, Plattner F, Raivich G, Behrens A. The ATM cofactor ATMIN protects against oxidative stress and accumulation of DNA damage in the aging brain. J Biol Chem 2010; 285:38534-42. [PMID: 20889973 DOI: 10.1074/jbc.m110.145896] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Progressive accumulation of DNA damage is causally involved in cellular senescence and organismal aging. The DNA damage kinase ATM plays a central role in maintaining genomic stability. ATM mutations cause the genetic disorder ataxia telangiectasia, which is primarily characterized by progressive neurodegeneration and cancer susceptibility. Although the importance of ATM function to protect against oxidative DNA damage and during aging is well described, the mechanism of ATM activation by these stimuli is not known. Here we identify ATM interactor (ATMIN) as an essential component of the ATM signaling pathway in response to oxidative stress and aging. Embryos lacking ATMIN (atmin(Δ/Δ)) died in utero and showed increased numbers of cells positive for phosphorylated histone H2aX, indicative of increased DNA damage. atmin(Δ/Δ) mouse embryonic fibroblasts accumulated DNA damage and prematurely entered senescence when cultured at atmospheric oxygen levels (20%), but this defect was rescued by addition of an antioxidant and also by culturing cells at physiological oxygen levels (3%). In response to acute oxidative stress, atmin(Δ/Δ) mouse embryonic fibroblasts showed slightly lower levels of ATM phosphorylation and reduced ATM substrate phosphorylation. Conditional deletion of ATMIN in the murine nervous system (atmin(ΔN)) resulted in reduced numbers of dopaminergic neurons, as does ATM deficiency. ATM activity was observed in old, but not in young, control mice, but aging-induced ATM signaling was impaired by ATMIN deficiency. Consequently, old atmin(ΔN) mice showed accumulation of DNA damage in the cortex accompanied by gliosis, resulting in increased mortality of aging mutant mice. These results suggest that ATMIN mediates ATM activation by oxidative stress, and thereby ATMIN protects the aging brain by preventing accumulation of DNA damage.
Collapse
Affiliation(s)
- Nnennaya Kanu
- Mammalian Genetics Lab, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3Y, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|