1
|
Tripathi S, Gupta E, Galande S. Statins as anti-tumor agents: A paradigm for repurposed drugs. Cancer Rep (Hoboken) 2024; 7:e2078. [PMID: 38711272 PMCID: PMC11074523 DOI: 10.1002/cnr2.2078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Statins, frequently prescribed medications, work by inhibiting the rate-limiting enzyme HMG-CoA reductase (HMGCR) in the mevalonate pathway to reduce cholesterol levels. Due to their multifaceted benefits, statins are being adapted for use as cost-efficient, safe and effective anti-cancer treatments. Several studies have shown that specific types of cancer are responsive to statin medications since they rely on the mevalonate pathway for their growth and survival. RECENT FINDINGS Statin are a class of drugs known for their potent inhibition of cholesterol production and are typically prescribed to treat high cholesterol levels. Nevertheless, there is growing interest in repurposing statins for the treatment of malignant neoplastic diseases, often in conjunction with chemotherapy and radiotherapy. The mechanism behind statin treatment includes targeting apoptosis through the BCL2 signaling pathway, regulating the cell cycle via the p53-YAP axis, and imparting epigenetic modulations by altering methylation patterns on CpG islands and histone acetylation by downregulating DNMTs and HDACs respectively. Notably, some studies have suggested a potential chemo-preventive effect, as decreased occurrence of tumor relapse and enhanced survival rate were reported in patients undergoing long-term statin therapy. However, the definitive endorsement of statin usage in cancer therapy hinges on population based clinical studies with larger patient cohorts and extended follow-up periods. CONCLUSIONS The potential of anti-cancer properties of statins seems to reach beyond their influence on cholesterol production. Further investigations are necessary to uncover their effects on cancer promoting signaling pathways. Given their distinct attributes, statins might emerge as promising contenders in the fight against tumorigenesis, as they appear to enhance the efficacy and address the limitations of conventional cancer treatments.
Collapse
Affiliation(s)
- Sneha Tripathi
- Laboratory of Chromatin Biology & EpigeneticsIndian Institute of Science Education and ResearchPuneIndia
| | - Ekta Gupta
- Laboratory of Chromatin Biology & EpigeneticsIndian Institute of Science Education and ResearchPuneIndia
| | - Sanjeev Galande
- Laboratory of Chromatin Biology & EpigeneticsIndian Institute of Science Education and ResearchPuneIndia
- Centre of Excellence in Epigenetics, Department of Life SciencesShiv Nadar Institution of EminenceGautam Buddha NagarIndia
| |
Collapse
|
2
|
Xie W, Peng M, Liu Y, Zhang B, Yi L, Long Y. Simvastatin induces pyroptosis via ROS/caspase-1/GSDMD pathway in colon cancer. Cell Commun Signal 2023; 21:329. [PMID: 37974278 PMCID: PMC10652480 DOI: 10.1186/s12964-023-01359-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The outcome of patients with colon cancer is still unsatisfied nowadays. Simvastatin is a type of statins with anti-cancer activity, but its effect on colon cancer cells remains unclear. The present study is intended to determine the underlying mechanism of simvastatin in treatment of colon cancer. METHODS The viability and pyroptosis rate of cells treated and untreated with simvastatin were analysed by CCK-8 and flow cytometry assays, respectively. We used DCFH-DA and flow cytometry to detect reactive oxygen species (ROS) production. Levels of pyroptosis markers were detected by western blotting analysis or immunofluorescence staining. Besides, the anticancer properties of simvastatin on colon cancer were further demonstrated using a cell line based xenograft tumor model. RESULTS Simvastatin treatment in HCT116 and SW620 induced pyroptosis and suppressed cell proliferation, with changes in the expression level of NLPR3, ASC, cleaved-caspase-1, mature IL-1β, IL-18 and GSDMD-N. Moreover, inhibition of caspase-1 and ROS attenuated the effects of simvastatin on cancer cell viability. In addition, it was identified that simvastatin has an anti-tumor effect by down-regulating ROS production and inducing downstream caspase-1 dependent pyroptosis in the subcutaneous transplantation tumors of HCT116 cells in BALB/c nude mice. CONCLUSIONS Our in vitro and in vivo results indicated that simvastatin induced pyroptosis through ROS/caspase-1/GSDMD pathway, thereby serving as a potential agent for colon cancer treatment. Video Abstract.
Collapse
Affiliation(s)
- Wei Xie
- Translational medicine centre, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, P. R. China
- Department of Hepatobiliary Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, P. R. China
| | - Mingjing Peng
- Central laboratory, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Ying Liu
- Hunan Provincial Clinical Research Centre for Oncoplastic Surgery, Hunan Cancer hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, 410013, P. R. China
| | - Bocheng Zhang
- Translational medicine centre, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, P. R. China
- Hunan Provincial Clinical Research Centre for Oncoplastic Surgery, Hunan Cancer hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, 410013, P. R. China
| | - Liang Yi
- Hunan Provincial Clinical Research Centre for Oncoplastic Surgery, Hunan Cancer hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, 410013, P. R. China
| | - Ying Long
- Translational medicine centre, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, P. R. China.
- Hunan Provincial Clinical Research Centre for Oncoplastic Surgery, Hunan Cancer hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan, 410013, P. R. China.
| |
Collapse
|
3
|
Riegger J, Brenner RE. Increase of cell surface vimentin is associated with vimentin network disruption and subsequent stress-induced premature senescence in human chondrocytes. eLife 2023; 12:e91453. [PMID: 37855367 PMCID: PMC10622146 DOI: 10.7554/elife.91453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
Accumulation of dysfunctional chondrocytes has detrimental consequences on the cartilagehomeostasis and is thus thought to play a crucial role during the pathogenesis of osteoarthritis(OA). However, the underlying mechanisms of phenotypical alteration in chondrocytes areincompletely understood. Here, we provide evidence that disruption of the intracellularvimentin network and consequent phenotypical alteration in human chondrocytes results in anexternalization of the intermediate filament. The presence of the so-called cell surfacevimentin (CSV) on chondrocytes was associated with the severity of tissue degeneration inclinical OA samples and was enhanced after mechanical injury of cartilage tissue. By meansof a doxorubicine-based in vitro model of stress-induced premature senescence (SIPS), wecould confirm the connection between cellular senescence and amount of CSV. AlthoughsiRNA-mediated silencing of CDKN2A clearly reduced the senescent phenotype as well asCSV levels of human chondrocytes, cellular senescence could not be completely reversed.Interestingly, knockdown of vimentin resulted in a SIPS-like phenotype and consequentlyincreased CSV. Therefore, we concluded that the integrity of the intracellular vimentinnetwork is crucial to maintain cellular function in chondrocytes. This assumption could beconfirmed by chemically- induced collapse of the vimentin network, which resulted in cellularstress and enhanced CSV expression. Regarding its biological function, CSV was found to beassociated with enhanced chondrocyte adhesion and plasticity. While osteogenic capacitiesseemed to be enhanced in chondrocytes expressing high levels of CSV, the chondrogenicpotential was clearly compromised. Overall, our study reinforces the importance of thevimentin network in maintenance of the chondrogenic phenotype and introduces CSV as anovel membrane-bound marker of dysfunctional chondrocytes.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of UlmUlmGermany
| | - Rolf E Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of UlmUlmGermany
| |
Collapse
|
4
|
Quality by design (QbD) assisted Fabrication & evaluation of Simvastatin loaded Nano-Enabled thermogel for melanoma therapy. Int J Pharm 2022; 628:122270. [DOI: 10.1016/j.ijpharm.2022.122270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/22/2022]
|
5
|
miR-197 Participates in Lipopolysaccharide-Induced Cardiomyocyte Injury by Modulating SIRT1. Cardiol Res Pract 2022; 2022:7687154. [PMID: 35223094 PMCID: PMC8872679 DOI: 10.1155/2022/7687154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/11/2022] [Indexed: 11/25/2022] Open
Abstract
Sepsis is a systemic inflammation and is capable of inducing myocarditis, which is a major leading cause of death in patients. Studies have found that miR-197 is correlated with the prognosis of patients with inflammatory heart disease, but its effect on sepsis-induced cardiomyocyte injury remains unclear. We treated H9c2 cells with lipopolysaccharide (LPS), then detected the cell viability via the cell counting kit-8 (CCK-8) assay and quantified miR-197 expression via quantitative real-time polymerase chain reaction (qRT-PCR). Then, we investigated the role of miR-197 in LPS-induced H9c2 cells by CCK-8 assay, flow cytometry, lactate dehydrogenase (LDH) measurement, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, and western blot. Subsequently, silent information regulator 1 (SIRT1) was downregulated in H9c2 cells to explore its interaction with miR-197 under LPS induction. LPS induced miR-197 overexpression in H9c2 cells. LPS restrained viability, the expressions of B-cell lymphoma-2 (Bcl-2) and SIRT1, but promoted apoptosis, LDH release, and levels of interleukin-6 (IL-6), interleukin-1β (IL-1β), acetyl (AC)-p53, BCL2-associated X (Bax), and cleaved caspase-3 in H9c2 cells. miR-197 inhibition reversed the effects of LPS on H9c2 cells. The protective role of miR-197 downregulation in LPS-induced H9c2 cells was reversed by SIRT1 silencing. miR-197 contributed to LPS-induced cardiomyocyte injury by modulating SIRT1, which might be used as a molecular marker in the management of sepsis.
Collapse
|
6
|
Hou X, Shi J, Sun L, Song L, Zhao W, Xiong X, Lu Y. The involvement of ERK1/2 and p38 MAPK in the premature senescence of melanocytes induced by H 2O 2 through a p53-independent p21 pathway. J Dermatol Sci 2022; 105:88-97. [PMID: 35042627 DOI: 10.1016/j.jdermsci.2022.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/30/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND The pathogenesis of vitiligo is still unknown and oxidative stress is an important factor that can damage or incapacitate melanocytes. OBJECTIVE To investigate the role of oxidative stress in the premature senescence of melanocytes and their transfer of melanosomes. METHODS Cultured human melanocytes were treated with H2O2 after which cell viability and apoptosis were assessed. We investigated whether exposure to H2O2 induces premature senescence. RNA sequencing was used to screen aging-related signaling pathways. The expression of dendritic regulatory proteins, adhesion molecules and cell cytoskeletal proteins, as well as melanosome distribution were characterized. The ROS scavenger NAC was used to study the role of ROS in cell senescence and in melanosome transfer. RESULTS Cell viability decreased progressively and cell apoptosis increased after treatment with H2O2. H2O2 treatment tended to induce premature senescence in melanocytes through a p53-independent p21 pathway. RNA sequencing analysis showed that H2O2 treatment induced the differential expression of MAPK signaling pathway components. Western blotting and qRT-PCR confirmed that H2O2 treatment increased the phosphorylation of ERK1/2 and p38 MAPK, which are involved in inducing the senescence of melanocytes, but not JNK. The expression of cell cytoskeleton and adhesion molecules decreased after H2O2 treatment. p21 siRNA treatment reversed these changes. Treatment with NAC improved the premature senescence and the impaired melanosome transfer induced by H2O2. CONCLUSION H2O2 increases ROS levels, which activates the ERK1/2 and p38 MAPK pathways to induce the premature senescence of melanocytes through p21 via a p53-independent pathway and consequently disrupts melanosome transfer.
Collapse
Affiliation(s)
- Xiaoyuan Hou
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Jiaqi Shi
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Li Sun
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Lebin Song
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Wene Zhao
- Nanjing Medical University, Analysis and Testing Center, Nanjing 210029, China.
| | - Xixi Xiong
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Yan Lu
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
7
|
ROS as Regulators of Cellular Processes in Melanoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1208690. [PMID: 34725562 PMCID: PMC8557056 DOI: 10.1155/2021/1208690] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
In this review, we examine the multiple roles of ROS in the pathogenesis of melanoma, focusing on signal transduction and regulation of gene expression. In recent years, different studies have analyzed the dual role of ROS in regulating the redox system, with both negative and positive consequences on human health, depending on cell concentration of these agents. High ROS levels can result from an altered balance between oxidant generation and intracellular antioxidant activity and can produce harmful effects. In contrast, low amounts of ROS are considered beneficial, since they trigger signaling pathways involved in physiological activities and programmed cell death, with protective effects against melanoma. Here, we examine these beneficial roles, which could have interesting implications in melanoma treatment.
Collapse
|
8
|
Yuan D, Liu Y, Li M, Zhou H, Cao L, Zhang X, Li Y. Senescence associated long non-coding RNA 1 regulates cigarette smoke-induced senescence of type II alveolar epithelial cells through sirtuin-1 signaling. J Int Med Res 2021; 49:300060520986049. [PMID: 33535826 PMCID: PMC7869169 DOI: 10.1177/0300060520986049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The primary aim of our study was to explore the mechanisms through which long non-coding RNA (lncRNA)-mediated sirtuin-1 (SIRT1) signaling regulates type II alveolar epithelial cell (AECII) senescence induced by a cigarette smoke-media suspension (CSM). METHODS Pharmacological SIRT1 activation was induced using SRT2104 and senescence-associated lncRNA 1 (SAL-RNA1) was overexpressed. The expression of SIRT1, FOXO3a, p53, p21, MMP-9, and TIMP-1 in different groups was detected by qRT-PCR and Western blotting; the activity of SA-β gal was detected by staining; the binding of SIRT1 to FOXO3a and p53 gene transcription promoters was detected by Chip. RESULTS We found that CSM increased AECII senescence, while SAL-RNA1 overexpression and SIRT1 activation significantly decreased levels of AECII senescence induced by CSM. Using chromatin immunoprecipitation, we found that SIRT1 bound differentially to transcriptional complexes on the FOXO3a and p53 promoters. CONCLUSION Our results suggested that lncRNA-SAL1-mediated SIRT1 signaling reduces senescence of AECIIs induced by CSM. These findings suggest a new therapeutic target to limit the irreversible apoptosis of lung epithelial cells in COPD patients.
Collapse
Affiliation(s)
- Dong Yuan
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College Hangzhou, Zhejiang, P.R. China.,Graduate Department, Bengbu Medical College, Bengbu, Anhui, P. R. China
| | - Yuanshun Liu
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College Hangzhou, Zhejiang, P.R. China
| | - Mengyu Li
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College Hangzhou, Zhejiang, P.R. China.,Graduate Department, Bengbu Medical College, Bengbu, Anhui, P. R. China
| | - Hongbin Zhou
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College Hangzhou, Zhejiang, P.R. China
| | - Liming Cao
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College Hangzhou, Zhejiang, P.R. China
| | - Xiaoqin Zhang
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College Hangzhou, Zhejiang, P.R. China
| | - Yaqing Li
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College Hangzhou, Zhejiang, P.R. China.,Department of Internal Medicine, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, P. R. China
| |
Collapse
|
9
|
Shomali T, Ashrafi M. Statins, cancer, and oxidative stress. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00023-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
10
|
Adult mesenchymal stem cell ageing interplays with depressed mitochondrial Ndufs6. Cell Death Dis 2020; 11:1075. [PMID: 33323934 PMCID: PMC7738680 DOI: 10.1038/s41419-020-03289-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cell (MSC)-based therapy has emerged as a novel strategy to treat many degenerative diseases. Accumulating evidence shows that the function of MSCs declines with age, thus limiting their regenerative capacity. Nonetheless, the underlying mechanisms that control MSC ageing are not well understood. We show that compared with bone marrow-MSCs (BM-MSCs) isolated from young and aged samples, NADH dehydrogenase (ubiquinone) iron-sulfur protein 6 (Ndufs6) is depressed in aged MSCs. Similar to that of Ndufs6 knockout (Ndufs6−/−) mice, MSCs exhibited a reduced self-renewal and differentiation capacity with a tendency to senescence in the presence of an increased p53/p21 level. Downregulation of Ndufs6 by siRNA also accelerated progression of wild-type BM-MSCs to an aged state. In contrast, replenishment of Ndufs6 in Ndufs6−/−-BM-MSCs significantly rejuvenated senescent cells and restored their proliferative ability. Compared with BM-MSCs, Ndufs6−/−-BM-MSCs displayed increased intracellular and mitochondrial reactive oxygen species (ROS), and decreased mitochondrial membrane potential. Treatment of Ndufs6−/−-BM-MSCs with mitochondrial ROS inhibitor Mito-TEMPO notably reversed the cellular senescence and reduced the increased p53/p21 level. We provide direct evidence that impairment of mitochondrial Ndufs6 is a putative accelerator of adult stem cell ageing that is associated with excessive ROS accumulation and upregulation of p53/p21. It also indicates that manipulation of mitochondrial function is critical and can effectively protect adult stem cells against senescence.
Collapse
|
11
|
Gu C, Zhang Q, Ni D, Xiao QF, Cao LF, Fei CY, Ying Y, Li N, Tao F. Therapeutic Effects of SRT2104 on Lung Injury in Rats with Emphysema via Reduction of Type II Alveolar Epithelial Cell Senescence. COPD 2020; 17:444-451. [PMID: 32722945 DOI: 10.1080/15412555.2020.1797657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most prevalent and severe diseases worldwide with high societal and health care costs. The pathogenesis of COPD is very complicated, and no curative treatment is available. Cellular senescence promotes the development of COPD. Type II alveolar epithelial cells (AECII) play a momentous role in lung tissue repair and maintenance of alveolar homeostasis. Sirtuin 1 (SIRT1), an antiaging molecule involved in the response to chronic inflammation and oxidative stress, regulates many pathophysiological changes including stress resistance, apoptosis, inflammation, and cellular senescence. This study aimed to investigate whether the pharmacological SIRT1 activator SRT2104 protects against AECII senescence in rats with emphysema. Our findings confirmed that SRT2104 administration reduced the pathological characteristics of emphysema and improved lung function parameters, including pulmonary resistance, pulmonary dynamic compliance, and peak expiratory flow. Moreover, SRT2104 treatment upregulated the expression of surfactant proteins A and C, SIRT1, and forkhead box O 3a (FoxO3a), decreased senescence-associated-β-galactosidase (SA-β-gal) activity, increased SIRT1 deacetylase activity, and downregulated the levels of p53 and p21. Therefore, SRT2104 administration protected against AECII senescence in rats with emphysema via SIRT1/FoxO3a and SIRT1/p53 signaling pathways and may provide a novel potential therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Chao Gu
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Qi Zhang
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Dan Ni
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Qin-Feng Xiao
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Lin-Feng Cao
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Chun-Yuan Fei
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Ying Ying
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Na Li
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| | - Feng Tao
- Department of Respiratory Medicine, The First Hospital of Jiaxing (the Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang, People's Republic of China
| |
Collapse
|
12
|
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: 4.0] [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
|
13
|
Yang Z, Liu X, Wang L, Wang T, Chen Y, Teng X, Li J, Shao L, Hui J, Ye W, Shen Z. The protective effects of HMGA2 in the senescence process of bone marrow-derived mesenchymal stromal cells. J Tissue Eng Regen Med 2020; 14:588-599. [PMID: 32068957 DOI: 10.1002/term.3023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/25/2019] [Accepted: 02/03/2020] [Indexed: 12/15/2022]
Abstract
Bone marrow-derived mesenchymal stromal cells (MSCs) have been wildly applied to cell-based strategies for tissue engineering and regenerative medicine; however, they have to undergo the senescence process and thus appeared to be less therapeutic effective. HMGA2, a protein belonged to high mobility group A (HMGA) family, exhibits an inverse expression level related to embryonic development and acts as a developmental regulator in stem cell self-renewal progression. Therefore, we performed senescence-associated β-galactosidase (SA-β-gal) staining, transwell assay, to examine the changes of MSCs in different stages and then over-expressed HMGA2 in MSCs by lentivirus transfection. We found the percentage of SA-β-gal staining positive cells in MSCs from 24-month-old Sprague-Dawley (SD) rats (O-MSCs) was significantly higher compared with MSCs from 2-week-old SD rats (Y-MSCs), and the expression levels of P21 and P53, two senescence-related molecules, were also significantly up-regulated in O-MSCs than in Y-MSCs. In contrast, the HMGA2 expression level in O-MSCs was dramatically down-regulated in contrast to Y-MSCs. In additional, the migration ability in O-MSCs was significantly attenuated than in Y-MSCs. After successfully over-expressed HMGA2 in O-MSCs, the percentage of SA-β-gal staining positive cells and the expression levels of P21 and P53 were reduced, and the migration ability was improved compared with O-MSCs without treatment. Further, mRNA sequencing analysis revealed that overexpression of HMGA2 changed the expression of genes related to cell proliferation and senescence, such as Lyz2, Pf4, Rgs2, and Mstn. Knockdown of Rgs2 in HMGA2 overexpression O-MSCs could antagonize the protective effect of HMGA2 in the senescence process of O-MSCs.
Collapse
Affiliation(s)
- Ziying Yang
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xuan Liu
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Longgang Wang
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Tao Wang
- Department of Cardiology, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xiaomei Teng
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Jingjing Li
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Jie Hui
- Department of Cardiology, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Wenxue Ye
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
14
|
Clement M, Luo L. Organismal Aging and Oxidants beyond Macromolecules Damage. Proteomics 2020; 20:e1800400. [DOI: 10.1002/pmic.201800400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Marie‐Veronique Clement
- Department of BiochemistryYong Loo Lin School of MedicineNational University of Singapore Singapore 117596 Singapore
- National University of Singapore Graduate School for Integrative Sciences and Engineering Singapore 117456 Singapore
| | - Le Luo
- Department of BiochemistryYong Loo Lin School of MedicineNational University of Singapore Singapore 117596 Singapore
| |
Collapse
|
15
|
Zdybel M, Chodurek E, Pilawa B. Application of EPR spectroscopy to determine the influence of simvastatin concentration on free radicals in A-375 human melanoma malignum cells. Toxicol In Vitro 2019; 61:104620. [PMID: 31394162 DOI: 10.1016/j.tiv.2019.104620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 08/02/2019] [Indexed: 10/26/2022]
Abstract
Malignant melanoma is the most aggressive and lethal form of skin cancer. Therefore the search for new methods of the treatment for melanoma is needed. In this work simvastatin was tested as the substance of potential anticancer activity. The aim of this study was to determine the effect of simvastatin in different concentration on free radicals in A-375 human melanoma malignum cells. Free radicals and growth of A-375 melanoma cells cultured without and with simvastatin were examined. Free radicals were tested by the use of electron paramagnetic resonance (EPR) spectroscopy. o-Semiquinone free radicals existed in A-375 cells. Free radicals of the control A-375 cells and the cells cultured with simvastatin were homogeneously broadened. The growth of A-375 cells did not change for concentrations of simvastatin 0.1 μM and 0.3 μM, and it slightly decreased for concentration of this substance equal 1 μM, so these concentrations of simvastatin were not preferred for therapy. For the concentrations of simvastatin higher than 1 μM the growth of A-375 cells was considerably weakened. Simvastatin in the concentrations of 0.1 μM and 1 μM decreased free radical concentrations in A-375 cells. The concentrations of simvastatin of 3 μM and 5 μM accompanied by the strong cell damage and strong formation of free radicals in them were recommended for anticancer therapy of A-375 melanoma cells.
Collapse
Affiliation(s)
- Magdalena Zdybel
- Department of Biophysics, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Jedności 8, 41-200 Sosnowiec, Poland.
| | - Ewa Chodurek
- Department of Biopharmacy, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Jedności 8, 41-200 Sosnowiec, Poland
| | - Barbara Pilawa
- Department of Biophysics, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Jedności 8, 41-200 Sosnowiec, Poland
| |
Collapse
|
16
|
Lee YJ, Kim WI, Kim SY, Cho SW, Nam HS, Lee SH, Cho MK. Flavonoid morin inhibits proliferation and induces apoptosis of melanoma cells by regulating reactive oxygen species, Sp1 and Mcl-1. Arch Pharm Res 2019; 42:531-542. [PMID: 31049822 DOI: 10.1007/s12272-019-01158-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/22/2019] [Indexed: 01/21/2023]
Abstract
Reactive oxygen species (ROS) is associated with cancer progression in different cancers, including melanoma. It also affects specificity protein (Sp1), a transcription factor. Flavonoid morin is known to inhibit growth of cancer cells, including lung cancer and breast cancer. Herein, we hypothesized that morin can inhibit cancer activities in melanoma by altering ROS generation. The aim of this study is to determine the effects of morin and its underlying mechanisms in melanoma cells. Effects of morin on cell proliferation and apoptosis were determined using standardized assays. Changes in pro-apoptotic and anti-apoptotic proteins were analyzed by western blot analysis. Cellular ROS levels and mitochondrial function were evaluated by measuring DCF-DA fluorescence and rhodamine-123 fluorescence intensities, respectively. Morin induced ROS production and apoptosis, as presented by increased proportion of cells with Annexin V-PE(+) staining and sub-G0/G1 peak in cell cycle analysis. It also downregulated Sp1, Mcl-1, Bcl-2, and caspase-3 but upregulated cleaved caspase-3, Bax, and PUMA. In immunohistochemical staining, Sp1 was overexpressed in melanoma tissues compared to normal skin tissues. Collectively, our data suggest that morin can induce apoptosis of melanoma cells by regulating pro-apoptotic and anti-apoptotic proteins through ROS, and may be a potential substance for treatment of melanoma.
Collapse
Affiliation(s)
- Yoon Jin Lee
- Molecular Cancer Research, Soonchunhyang University College of Medicine, Cheonan, 31151, Republic of Korea
| | - Woo Il Kim
- Department of Dermatology, Soonchunhyang University Hospital, Seoul, 04401, Republic of Korea
| | - Soo Young Kim
- Department of Dermatology, Soonchunhyang University Hospital, Seoul, 04401, Republic of Korea
| | - Sung Woo Cho
- Molecular Cancer Research, Soonchunhyang University College of Medicine, Cheonan, 31151, Republic of Korea
| | - Hae Seon Nam
- Molecular Cancer Research, Soonchunhyang University College of Medicine, Cheonan, 31151, Republic of Korea
| | - Sang Han Lee
- Molecular Cancer Research, Soonchunhyang University College of Medicine, Cheonan, 31151, Republic of Korea
| | - Moon Kyun Cho
- Department of Dermatology, Soonchunhyang University Hospital, Seoul, 04401, Republic of Korea.
| |
Collapse
|
17
|
Shang D, Wu Y, Ding Y, Lu Z, Shen Y, Zhu F, Liu H, Zhu C, Tu Z. Identification of a pyridine derivative inducing senescence in ovarian cancer cell lines via P21 activation. Clin Exp Pharmacol Physiol 2017; 45:452-460. [DOI: 10.1111/1440-1681.12891] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 10/04/2017] [Accepted: 10/26/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Dongsheng Shang
- School of Pharmacy; Jiangsu University; Zhenjiang China
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Yanfang Wu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Ya Ding
- School of Chemistry & Chemical Engineering; Jiangsu University; Zhenjiang China
| | - Ziwen Lu
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Yanting Shen
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Feifei Zhu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| | - Hanqing Liu
- School of Pharmacy; Jiangsu University; Zhenjiang China
| | - Chunyin Zhu
- School of Chemistry & Chemical Engineering; Jiangsu University; Zhenjiang China
| | - Zhigang Tu
- Institute of Life Sciences; Jiangsu University; Zhenjiang China
| |
Collapse
|
18
|
Hwang ES, Ok JS, Song S. Chemical and Physical Approaches to Extend the Replicative and Differentiation Potential of Stem Cells. Stem Cell Rev Rep 2017; 12:315-26. [PMID: 27085715 DOI: 10.1007/s12015-016-9652-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell therapies using mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) are increasing in regenerative medicine, with applications to a growing number of aging-associated dysfunctions and degenerations. For successful therapies, a certain mass of cells is needed, requiring extensive ex vivo expansion of the cells. However, the proliferation of both MSCs and EPCs is limited as a result of telomere shortening-induced senescence. As cells approach senescence, their proliferation slows down and differentiation potential decreases. Therefore, ways to delay senescence and extend the replicative lifespan these cells are needed. Certain proteins and pathways play key roles in determining the replicative lifespan by regulating ROS generation, damage accumulation, or telomere shortening. And, their agonists and gene activators exert positive effects on lifespan. In many of the treatments, importantly, the lifespan is extended with the retention of differentiation potential. Furthermore, certain culture conditions, including the use of specific atmospheric conditions and culture substrates, exert positive effects on not only the proliferation rate, but also the extent of proliferation and differentiation potential as well as lineage determination. These strategies and known underlying mechanisms are introduced in this review, with an evaluation of their pros and cons in order to facilitate safe and effective MSC expansion ex vivo.
Collapse
Affiliation(s)
- Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemun-gu, Seoulsiripdaero 163, Seoul, 02504, Republic of Korea.
| | - Jeong Soo Ok
- Department of Life Science, University of Seoul, Dongdaemun-gu, Seoulsiripdaero 163, Seoul, 02504, Republic of Korea
| | - SeonBeom Song
- Department of Life Science, University of Seoul, Dongdaemun-gu, Seoulsiripdaero 163, Seoul, 02504, Republic of Korea
| |
Collapse
|
19
|
Bai M, Zhang M, Long F, Yu N, Zeng A, Zhao R. Circulating microRNA-194 regulates human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway. Oncol Rep 2017; 37:2702-2710. [PMID: 28358423 PMCID: PMC5428795 DOI: 10.3892/or.2017.5537] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/07/2017] [Indexed: 12/18/2022] Open
Abstract
In the present study, we analyzed the role of microRNA-194 circulating regulated human melanoma cell growth. We found that microRNA-194 expression was markedly suppressed in human melanoma patients, compared with negative control group. Next, disease-free survival (DFS) and overall survival (OS) of high expression in human melanoma patients was higher than those of low expression in human melanoma patients. MicroRNA-194 overexpression inhibited cell proliferation, induced apoptosis, increased caspase-3/-9 activities and promoted Bax/Bcl-2 of human melanoma cells. Furthermore, microRNA-194 overexpression also suppressed PI3K/AKT/FoxO3a signaling pathway and induced p53/p21 signaling pathway. PI3K inhibitor, suppressed PI3K, phosphorylation-AKT, FoxO3a protein expression and increased the effects of microRNA-194 overexpression on cell growth, apoptosis, caspase-3/-9 activities and Bax/Bcl-2 protein expression of human melanoma cells through the induction of p53/p21 signaling pathway. Taken together, these data indicate that circulating microRNA-194 regulated human melanoma cells via PI3K/AKT/FoxO3a and p53/p21 signaling pathway.
Collapse
Affiliation(s)
- Ming Bai
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Mingzi Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Fei Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Nanze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Ang Zeng
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Ru Zhao
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
20
|
Licarete E, Sesarman A, Rauca VF, Luput L, Patras L, Banciu M. HIF-1α acts as a molecular target for simvastatin cytotoxicity in B16.F10 melanoma cells cultured under chemically induced hypoxia. Oncol Lett 2017; 13:3942-3950. [PMID: 28521491 DOI: 10.3892/ol.2017.5928] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/31/2017] [Indexed: 12/30/2022] Open
Abstract
Our previous studies reported that one of the main mechanisms of the antitumor activity of simvastatin (SIM) in B16.F10 murine melanoma cells was associated with strong suppression of the constitutive cell production of the α subunit of the heterodimeric transcription factor hypoxia-inducible factor (HIF)-1. Thus, the present study aimed to broaden this finding under hypoxic conditions induced by incubation of B16.F10 cells with cobalt chloride, when the constitutive production of HIF-1α in these melanoma cells is amplified by inducible expression of this factor. The data demonstrated that the SIM antiproliferative effects on melanoma cells were mediated mainly via strong suppressive actions on the B16.F10 cell capacity to support tumor angiogenesis and inflammation, as a result of a high inhibition of the inducible expression of HIF-1α. However, the constitutive expression of HIF-1α was not affected by SIM, probably due to the lack of effect of this statin on nuclear factor-κB production in B16.F10 cancer cells at the concentration tested. Additionally, the present study noted slight reducing effects of SIM on tumor oxidative stress, which may contribute to the main inhibitory action of this statin on HIF-1α production in hypoxic tumor cells. Collectively, these data are valuable for future anticancer strategies based on SIM administration in combination with cytotoxic drugs that are able to counteract the constitutive expression of HIF-1α in tumors.
Collapse
Affiliation(s)
- Emilia Licarete
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Valentin Florian Rauca
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Lavinia Luput
- Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| |
Collapse
|
21
|
Gu C, Li Y, Liu J, Ying X, Liu Y, Yan J, Chen C, Zhou H, Cao L, Ma Y. LncRNA‑mediated SIRT1/FoxO3a and SIRT1/p53 signaling pathways regulate type II alveolar epithelial cell senescence in patients with chronic obstructive pulmonary disease. Mol Med Rep 2017; 15:3129-3134. [PMID: 28339038 DOI: 10.3892/mmr.2017.6367] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
Abstract
The loss of alveolar structure and airspace enlargement are major pathological changes in chronic obstructive pulmonary disease (COPD). Type II alveolar epithelial cells (AECII) are involved in maintaining lung tissue repair and alveolar homeostasis. Long non‑coding RNAs (lncRNAs) are involved in multi‑regulating gene transcription, affecting processes including embryonic development, cell differentiation and cellular senescence. The primary aim of the present study was to explore the mechanisms of AECII senescence regulated by lncRNA‑mediated sirtuin 1 (SIRT1) and forkhead box O 3a (FoxO3a) signaling pathways in patients with COPD. Lung tissues from patients with COPD exhibited pathological characteristics and significantly increased senescence‑associated β‑galactosidase activity. Furthermore, the expression levels of senescence‑associated lncRNA1 (SAL‑RNA1), SIRT1 and FoxO3a were reduced, but SAL‑RNA2, SAL‑RNA3, p53 and p21 were upregulated in the lung tissues of patients with COPD compared with control. The results of the present study indicated that lncRNA‑mediated SIRT1/p53 and FoxO3a signaling pathways may regulate AECII senescence in the pathogenesis of COPD, which may provide a novel experimental basis for the treatment of COPD.
Collapse
Affiliation(s)
- Chao Gu
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yaqing Li
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jialiang Liu
- Department of Respiratory Medicine, The First Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiwang Ying
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yuanshun Liu
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jianping Yan
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Chun Chen
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Hongbin Zhou
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Liming Cao
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yingyu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
22
|
Karimian A, Ahmadi Y, Yousefi B. Multiple functions of p21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair (Amst) 2016; 42:63-71. [PMID: 27156098 DOI: 10.1016/j.dnarep.2016.04.008] [Citation(s) in RCA: 728] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/19/2016] [Accepted: 04/19/2016] [Indexed: 12/13/2022]
Abstract
An appropriate control over cell cycle progression depends on many factors. Cyclin-dependent kinase (CDK) inhibitor p21 (also known as p21(WAF1/Cip1)) is one of these factors that promote cell cycle arrest in response to a variety of stimuli. The inhibitory effect of P21 on cell cycle progression correlates with its nuclear localization. P21 can be induced by both p53-dependent and p53-independent mechanisms. Some other important functions attributed to p21 include transcriptional regulation, modulation or inhibition of apoptosis. These functions are largely dependent on direct p21/protein interactions and also on p21 subcellular localizations. In addition, p21 can play a role in DNA repair by interacting with proliferating cell nuclear antigen (PCNA). In this review, we will focus on the multiple functions of p21 in cell cycle regulation, apoptosis and gene transcription after DNA damage and briefly discuss the pathways and factors that have critical roles in p21 expression and activity.
Collapse
Affiliation(s)
- Ansar Karimian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yasin Ahmadi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Abstract
OBJECTIVE Antigen persistence due to HIV is a major source of inflammation and substantial immune activation, both of which are linked to accelerated aging. This illustrates the need to reduce immune activation in these patients and subsequently decrease the risk of cardiovascular diseases and other non-AIDS-defining comorbidities. METHODS CD4 T cells were infected with HIV-1 isolates in the presence or absence of atorvastatin (0.25 to 1 μg/ml) for 24-48 h. Atorvastatin-induced anti-inflammatory functions and anti-viral replication were measured in vitro. RESULTS Atorvastatin, a lipid-lowering medication, exerted a broad spectrum of anti-inflammatory functions by reducing T-cell immune activation markers (e.g. CD38, HLA-DR and Ki67), lowering HIV-1 co-receptor CCR-5, and decreasing proliferative capabilities of CD4 T cells in vitro. In contrast, atorvastatin expanded regulatory T cells (Tregs) and upregulated the expression of T-cell immunoglobulin and ITIM domain (TIGIT), which enhanced the suppressive activity of Tregs. Furthermore, atorvastatin upregulated the cyclin-dependent kinase inhibitor p21, which is also known as cip-1 and waf-1, in the CD4 T cells. Upregulation of p21 in CD4 T cells rendered them less susceptible to HIV-1 infection and replication whereas siRNA-mediated p21 depletion and/or p21 selective inhibitor rescued viral replication. Interestingly, atorvastatin reduced HIV infection in both rested and phytohemagglutinin-activated CD4 T cells in vitro. Finally, atorvastatin mediated p21 upregulation occurred via mevalonate pathway, but independent of p53. CONCLUSION The results demonstrate a novel mechanism by which atorvastatin induced resistance of CD4 T cells to HIV-1 infection via p21 upregulation and suggest that statins may hold particular promise for some HIV-infected individuals.
Collapse
|
24
|
Casey SC, Amedei A, Aquilano K, Azmi AS, Benencia F, Bhakta D, Bilsland AE, Boosani CS, Chen S, Ciriolo MR, Crawford S, Fujii H, Georgakilas AG, Guha G, Halicka D, Helferich WG, Heneberg P, Honoki K, Keith WN, Kerkar SP, Mohammed SI, Niccolai E, Nowsheen S, Vasantha Rupasinghe HP, Samadi A, Singh N, Talib WH, Venkateswaran V, Whelan RL, Yang X, Felsher DW. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin Cancer Biol 2015; 35 Suppl:S199-S223. [PMID: 25865775 PMCID: PMC4930000 DOI: 10.1016/j.semcancer.2015.02.007] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 02/06/2023]
Abstract
Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Asfar S Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Alan E Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chandra S Boosani
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | | | - Sarah Crawford
- Department of Biology, Southern Connecticut State University, New Haven, CT, United States
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | | | - William G Helferich
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sid P Kerkar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture, Dalhousie University, Nova Scotia, Canada
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | | | - Richard L Whelan
- Mount Sinai Roosevelt Hospital, Icahn Mount Sinai School of Medicine, New York City, NY, United States
| | - Xujuan Yang
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
25
|
Licarete E, Sesarman A, Banciu M. Exploitation of pleiotropic actions of statins by using tumour-targeted delivery systems. J Microencapsul 2015; 32:619-31. [PMID: 26299551 DOI: 10.3109/02652048.2015.1073383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Statins are drugs traditionally used to lower cholesterol levels in blood. At concentrations 100- to 500-fold higher than those needed for reaching cholesterol lowering activity, they have anti-tumour activity. This anti-tumour activity is based on statins pleiotropic effects derived from their ability to inhibit the mevalonate synthesis and include anti-proliferative, pro-apoptotic, anti-angiogenic, anti-inflammatory, anti-metastatic actions and modulatory effects on intra-tumour oxidative stress. Thus, in this review, we summarise the possible pleiotropic actions of statins involved in tumour growth inhibition. Since the administration of these high doses of statins is accompanied by severe side effects, targeted delivery of statins seems to be the appropriate strategy for efficient application of statins in oncology. Therefore, we also present an overview of the current status of targeted delivery systems for statins with possible utilisation in oncology.
Collapse
Affiliation(s)
- Emilia Licarete
- a Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology , Babes-Bolyai University , Cluj-Napoca , Romania and.,b Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Alina Sesarman
- a Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology , Babes-Bolyai University , Cluj-Napoca , Romania and.,b Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Manuela Banciu
- a Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology , Babes-Bolyai University , Cluj-Napoca , Romania and.,b Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| |
Collapse
|
26
|
Hambright HG, Meng P, Kumar AP, Ghosh R. Inhibition of PI3K/AKT/mTOR axis disrupts oxidative stress-mediated survival of melanoma cells. Oncotarget 2015; 6:7195-208. [PMID: 25749517 PMCID: PMC4466678 DOI: 10.18632/oncotarget.3131] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/09/2015] [Indexed: 12/17/2022] Open
Abstract
Elevated oxidative stress in cancer cells contributes to hyperactive proliferation and enhanced survival, which can be exploited using agents that increase reactive oxygen species (ROS) beyond a threshold level. Here we show that melanoma cells exhibit an oxidative stress phenotype compared with normal melanocytes, as evidenced by increased total cellular ROS, KEAP1/NRF2 pathway activity, protein damage, and elevated oxidized glutathione. Our overall objective was to test whether augmenting this high oxidative stress level in melanoma cells would inhibit their dependence on oncogenic PI3K/AKT/mTOR-mediated survival. We report that NexrutineR augmented the constitutively elevated oxidative stress markers in melanoma cells, which was abrogated by N-acetyl cysteine (NAC) pre-treatment. NexrutineR disrupted growth homeostasis by inhibiting proliferation, survival, and colony formation in melanoma cells without affecting melanocyte cell viability. Increased oxidative stress in melanoma cells inhibited PI3K/AKT/mTOR pathway through disruption of mTORC1 formation and phosphorylation of downstream targets p70S6K, 4EBP1 and rpS6. NAC pre-treatment reversed inhibition of mTORC1 targets, demonstrating a ROS-dependent mechanism. Overall, our results illustrate the importance of disruption of the intrinsically high oxidative stress in melanoma cells to selectively inhibit their survival mediated by PI3K/AKT/mTOR.
Collapse
Affiliation(s)
- Heather G. Hambright
- Department of Urology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
| | - Peng Meng
- Department of Urology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- Life Sciences Division, Lawrence Berkley National Laboratory, Berkley, California, 94710, USA
| | - Addanki P. Kumar
- Department of Urology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- South Texas Veterans Health Care System, San Antonio, Texas, 78229, USA
| | - Rita Ghosh
- Department of Urology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229, USA
| |
Collapse
|
27
|
TORRES CRISTIANG, OLIVARES ARACELI, STOORE CAROLL. Simvastatin exhibits antiproliferative effects on spheres derived from canine mammary carcinoma cells. Oncol Rep 2015; 33:2235-44. [DOI: 10.3892/or.2015.3850] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/19/2015] [Indexed: 11/05/2022] Open
|
28
|
Altwairgi AK. Statins are potential anticancerous agents (review). Oncol Rep 2015; 33:1019-39. [PMID: 25607255 DOI: 10.3892/or.2015.3741] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/23/2014] [Indexed: 11/05/2022] Open
Abstract
Statins are inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), which is a rate-limiting enzyme in the mevalonate pathway. The pleiotropic effects of statins may be mediated by the inhibition of downstream products such as small GTP-binding proteins, Rho, Ras and Rac whose localization and function are dependent on isoprenylation. Preclinical studies of statins in different cancer cell lines and animal models showed antiproliferative, pro‑apoptotic and anti-invasive effects. Notably, statins showed targeted action in cancerous cell lines compared to normal cells. Previous studies have also shown the synergistic effects of statins with chemotherapeutic agents and radiotherapy. This effect of statins was also observed in chemotherapeutic-resistant tumors. Statins were reported to sensitize the cells to radiation by arresting them in the late G1 phase of the cell cycle. Similarly, population-based studies also demonstrated a chemopreventive and survival benefit of statins in various types of cancers. However, this benefit has yet to be proven in clinical trials. The inter-individual variation in response to statins may be contributed to many genetic and non-genetic factors, including single-nucleotide polymorphisms in HMGCR gene and the overexpression of heterogeneous nuclear ribonucleoprotein A1, which was reported to reduce HMGCR enzyme activity. However, more studies with large phase III randomized controlled trials in cancer patients should be conducted to establish the effect of stains in cancer prevention and treatment.
Collapse
|
29
|
Hwang ES. Senescence suppressors: their practical importance in replicative lifespan extension in stem cells. Cell Mol Life Sci 2014; 71:4207-19. [PMID: 25052377 PMCID: PMC11113678 DOI: 10.1007/s00018-014-1685-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 06/23/2014] [Accepted: 07/14/2014] [Indexed: 02/06/2023]
Abstract
Recent animal and clinical studies report promising results for the therapeutic utilization of stem cells in regenerative medicine. Mesenchymal stem cells (MSCs), with their pluripotent nature, have advantages over embryonic stem cells in terms of their availability and feasibility. However, their proliferative activity is destined to slow by replicative senescence, and the limited proliferative potential of MSCs not only hinders the preparation of sufficient cells for in vivo application, but also draws a limitation on their potential for differentiation. This calls for the development of safe and efficient means to increase the proliferative as well as differentiation potential of MSCs. Recent advances have led to a better understanding of the underlying mechanisms and significance of cellular senescence, facilitating ways to manipulate the replicative lifespan of a variety of primary cells, including MSCs. This paper introduces a class of proteins that function as senescence suppressors. Like tumor suppressors, these proteins are lost in senescence, while their forced expression delays the onset of senescence. Moreover, treatments that increase the expression or the activity of senescence suppressors, therefore, cause expansion of the replicative and differentiation potential of MSCs. The nature of the activities and putative underlying mechanisms of the senescence suppressors will be discussed to facilitate their evaluation.
Collapse
Affiliation(s)
- Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemungu, Jeonnongdong 90, Seoul, 130-743, Republic of Korea,
| |
Collapse
|
30
|
SHEN YUANYUAN, DU YINGYING, ZHANG YING, PAN YUEYIN. Synergistic effects of combined treatment with simvastatin and exemestane on MCF-7 human breast cancer cells. Mol Med Rep 2012; 12:456-62. [DOI: 10.3892/mmr.2015.3406] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 01/27/2015] [Indexed: 11/06/2022] Open
|