101
|
Ghosh K, Capell BC. The Senescence-Associated Secretory Phenotype: Critical Effector in Skin Cancer and Aging. J Invest Dermatol 2016; 136:2133-2139. [PMID: 27543988 DOI: 10.1016/j.jid.2016.06.621] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/01/2016] [Accepted: 06/20/2016] [Indexed: 12/11/2022]
Abstract
Cellular senescence, a state of stable cell cycle arrest in response to cellular stress, is an indispensable mechanism to counter tumorigenesis by halting the proliferation of damaged cells. However, through the secretion of an array of diverse cytokines, chemokines, growth factors, and proteases known as the senescence-associated secretory phenotype (SASP), senescent cells can paradoxically promote carcinogenesis. Consistent with this, removal of senescent cells delays the onset of cancer and prolongs lifespan in vivo, potentially in part through SASP reduction. In this review, we consider the evidence for the SASP and "SASP-like" inflammation in driving skin carcinogenesis, emphasizing how further understanding of both the roles and mechanisms of SASP expression may offer new targets for skin cancer prevention and therapy.
Collapse
Affiliation(s)
- Kanad Ghosh
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Brian C Capell
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
102
|
|
103
|
Yao GD, Yang J, Li Q, Zhang Y, Qi M, Fan SM, Hayashi T, Tashiro SI, Onodera S, Ikejima T. Activation of p53 contributes to pseudolaric acid B-induced senescence in human lung cancer cells in vitro. Acta Pharmacol Sin 2016; 37:919-29. [PMID: 27041461 PMCID: PMC4933766 DOI: 10.1038/aps.2016.8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/24/2016] [Indexed: 02/08/2023]
Abstract
AIM Pseudolaric acid B (PAB), a diterpene acid isolated from the root bark of Pseudolarix kaempferi Gordon, has shown to exert anti-tumor effects via inducing cell cycle arrest followed by apoptosis in several cancer cell lines. Here we reported that PAB induced a mitotic catastrophe in human lung cancer A549 cells, which resulted in senescence without apoptosis or necrosis. METHODS Three human lung cancer cell lines (A549, H460 and H1299 cells) were examined. Cell growth inhibition was assessed with MTT assay. Cell cycle distribution was determined using a flow cytometer. Cell nuclear morphology was observed under a fluorescence microscope. Senescent cells were detected using SA-β-Gal staining. Apoptotic and senescent protein expression was examined using Western blot analysis. The expression of p53 and p21 in the cells was downregulated by siRNAs. RESULTS Treatment with PAB (5-80 μmol/L) inhibited the growth of A549 cells in dose- and time-dependent manners. Prolonged treatment with PAB (20 μmol/L) caused G2/M arrest at day 1 followed by mitotic catastrophe from day 2, which eventually resulted in cell senescence between days 3 and 4 without cell death (apoptosis or necrosis). Knockdown of p53 expression with siRNA significantly suppressed PAB-induced senescence in A549 cells (p53 wild). Furthermore, PAB-induced senescence was also observed in human lung cancer H460 cells (p53 wild), but not in human lung cancer H1299 cells (p53 null). CONCLUSION The anti-tumor action of PAB against human lung cancer A549 cells in vitro involves the induction of senescence through activation of the p53 pathway.
Collapse
Affiliation(s)
- Guo-dong Yao
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Yang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiang Li
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ye Zhang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Min Qi
- Tianjin Taiyang Pharmaceutical Co, Ltd, Tianjin 300457, China
| | - Si-miao Fan
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Toshihiko Hayashi
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shin-ichi Tashiro
- Institute for Clinical and Biomedical Sciences, Kyoto 603-8072, Japan
| | - Satoshi Onodera
- Department of Clinical and Pharmaceutical Sciences, Showa Pharmaceutical University, Tokyo 194–8543, Japan
| | - Takashi Ikejima
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
- E-mail
| |
Collapse
|
104
|
Capell BC, Drake AM, Zhu J, Shah PP, Dou Z, Dorsey J, Simola DF, Donahue G, Sammons M, Rai TS, Natale C, Ridky TW, Adams PD, Berger SL. MLL1 is essential for the senescence-associated secretory phenotype. Genes Dev 2016; 30:321-36. [PMID: 26833731 PMCID: PMC4743061 DOI: 10.1101/gad.271882.115] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Capell et al. show that MLL1 inhibition represses expression of critical proproliferative cell cycle regulators required for DNA replication and DNA damage response activation, thus disabling senescence-associated secretory phenotype (SASP) expression. These inhibitory effects of MLL1 on SASP gene expression do not impair oncogene-induced senescence and abolish the ability of the SASP to enhance cancer cell proliferation. Oncogene-induced senescence (OIS) and therapy-induced senescence (TIS), while tumor-suppressive, also promote procarcinogenic effects by activating the DNA damage response (DDR), which in turn induces inflammation. This inflammatory response prominently includes an array of cytokines known as the senescence-associated secretory phenotype (SASP). Previous observations link the transcription-associated methyltransferase and oncoprotein MLL1 to the DDR, leading us to investigate the role of MLL1 in SASP expression. Our findings reveal direct MLL1 epigenetic control over proproliferative cell cycle genes: MLL1 inhibition represses expression of proproliferative cell cycle regulators required for DNA replication and DDR activation, thus disabling SASP expression. Strikingly, however, these effects of MLL1 inhibition on SASP gene expression do not impair OIS and, furthermore, abolish the ability of the SASP to enhance cancer cell proliferation. More broadly, MLL1 inhibition also reduces “SASP-like” inflammatory gene expression from cancer cells in vitro and in vivo independently of senescence. Taken together, these data demonstrate that MLL1 inhibition may be a powerful and effective strategy for inducing cancerous growth arrest through the direct epigenetic regulation of proliferation-promoting genes and the avoidance of deleterious OIS- or TIS-related tumor secretomes, which can promote both drug resistance and tumor progression.
Collapse
Affiliation(s)
- Brian C Capell
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA; Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Adam M Drake
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Jiajun Zhu
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Parisha P Shah
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Zhixun Dou
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Jean Dorsey
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Daniel F Simola
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Greg Donahue
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Morgan Sammons
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Taranjit Singh Rai
- Institute of Cancer Sciences, Beatson Laboratories, University of Glasgow, Glasgow G611BD, United Kingdom; Institute of Biomedical and Environmental Health Research, University of the West of Scotland, Paisley PA12BE, United Kingdom
| | - Christopher Natale
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Todd W Ridky
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Peter D Adams
- Institute of Cancer Sciences, Beatson Laboratories, University of Glasgow, Glasgow G611BD, United Kingdom
| | - Shelley L Berger
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
105
|
Liposome encapsulated curcumin-difluorinated (CDF) inhibits the growth of cisplatin resistant head and neck cancer stem cells. Oncotarget 2016; 6:18504-17. [PMID: 26098778 PMCID: PMC4621906 DOI: 10.18632/oncotarget.4181] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/10/2015] [Indexed: 11/25/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer, with 600,000 new cases every year worldwide. Although chemotherapeutics exist, five-year survival is only 50%. New strategies to overcome drug resistance are required to improve HNSCC treatment. Curcumin-difluorinated (CDF), a synthetic analog of curcumin, was packaged in liposomes and used to evaluate growth inhibition of cisplatin resistant HNSCC cell lines CCL-23R and UM-SCC-1R generated from the parental cell lines CCL-23 and UM-SCC-1 respectively. Growth inhibition in vitro and expression levels of the CD44 (cancer stem cell marker), cytokines, and growth factors were investigated after liposomal CDF treatment. The in vivo growth inhibitory effect of liposomal CDF was evaluated in the nude mice xenograft tumor model of UM-SCC-1R and the inhibition of CD44 was measured. Treatment of the resistant cell lines in vitro with liposomal CDF resulted in a statistically significant growth inhibition (p < 0.05). The nude mice xenograft study showed a statistically significant tumor growth inhibition of UM-SCC-1R cells and a reduction in the expression of CD44 (p < 0.05), indicating an inhibitory effect of liposomal CDF on CSCs. Our results demonstrate that delivery of CDF through liposomes may be an effective method for the treatment of cisplatin resistant HNSCC.
Collapse
|
106
|
Loaiza N, Demaria M. Cellular senescence and tumor promotion: Is aging the key? Biochim Biophys Acta Rev Cancer 2016; 1865:155-67. [PMID: 26845683 DOI: 10.1016/j.bbcan.2016.01.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/29/2016] [Accepted: 01/31/2016] [Indexed: 01/07/2023]
Abstract
The senescence response is a potent tumor suppressor mechanism characterized by an irreversible growth arrest in response to potentially oncogenic signals to prevent the proliferation of damaged cells. Late in life, some of the features of senescent cells seem to mediate the development of age-related pathologies, including cancer. In the present review, we present a summary of the current knowledge regarding the causes, effector pathways and cellular features of senescence. We also discuss how the senescence response, initially a tumor suppressor mechanism, turns into a tumor promoter apparently as a consequence of aging. We argue that three age-related phenomena--senescence-associated secretory phenotype (SASP) dysregulation, decline in the immune system function and genomic instability--could contribute, independently or synergistically, to deteriorate the efficacy of the senescence response in stopping cancer. As a consequence, senescent cells could be considered premalignant cells, and targeting senescent cells could be a preventive and therapeutic strategy against cancer.
Collapse
Affiliation(s)
- Natalia Loaiza
- University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Marco Demaria
- University Medical Center Groningen (UMCG), Groningen, The Netherlands; European Research Institute for the Biology of Aging (ERIBA), Groningen, The Netherlands.
| |
Collapse
|
107
|
Pole A, Dimri M, P. Dimri G. Oxidative stress, cellular senescence and ageing. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.3.300] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
108
|
Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:111. [PMID: 26445347 PMCID: PMC4595070 DOI: 10.1186/s13046-015-0221-y] [Citation(s) in RCA: 483] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/11/2015] [Indexed: 12/15/2022]
Abstract
Tumor tissue is composed of cancer cells and surrounding stromal cells with diverse genetic/epigenetic backgrounds, a situation known as intra-tumoral heterogeneity. Cancer cells are surrounded by a totally different microenvironment than that of normal cells; consequently, tumor cells must exhibit rapidly adaptive responses to hypoxia and hypo-nutrient conditions. This phenomenon of changes of tumor cellular bioenergetics, called “metabolic reprogramming”, has been recognized as one of 10 hallmarks of cancer. Metabolic reprogramming is required for both malignant transformation and tumor development, including invasion and metastasis. Although the Warburg effect has been widely accepted as a common feature of metabolic reprogramming, accumulating evidence has revealed that tumor cells depend on mitochondrial metabolism as well as aerobic glycolysis. Remarkably, cancer-associated fibroblasts in tumor stroma tend to activate both glycolysis and autophagy in contrast to neighboring cancer cells, which leads to a reverse Warburg effect. Heterogeneity of monocarboxylate transporter expression reflects cellular metabolic heterogeneity with respect to the production and uptake of lactate. In tumor tissue, metabolic heterogeneity induces metabolic symbiosis, which is responsible for adaptation to drastic changes in the nutrient microenvironment resulting from chemotherapy. In addition, metabolic heterogeneity is responsible for the failure to induce the same therapeutic effect against cancer cells as a whole. In particular, cancer stem cells exhibit several biological features responsible for resistance to conventional anti-tumor therapies. Consequently, cancer stem cells tend to form minimal residual disease after chemotherapy and exhibit metastatic potential with additional metabolic reprogramming. This type of altered metabolic reprogramming leads to adaptive/acquired resistance to anti-tumor therapy. Collectively, complex and dynamic metabolic reprogramming should be regarded as a reflection of the “robustness” of tumor cells against unfavorable conditions. This review focuses on the concept of metabolic reprogramming in heterogeneous tumor tissue, and further emphasizes the importance of developing novel therapeutic strategies based on drug repositioning.
Collapse
Affiliation(s)
- Go J Yoshida
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan. .,Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
109
|
Castro-Vega LJ, Jouravleva K, Ortiz-Montero P, Liu WY, Galeano JL, Romero M, Popova T, Bacchetti S, Vernot JP, Londoño-Vallejo A. The senescent microenvironment promotes the emergence of heterogeneous cancer stem-like cells. Carcinogenesis 2015; 36:1180-92. [DOI: 10.1093/carcin/bgv101] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 07/08/2015] [Indexed: 11/14/2022] Open
|
110
|
Barakat DJ, Zhang J, Barberi T, Denmeade SR, Friedman AD, Paz-Priel I. CCAAT/Enhancer binding protein β controls androgen-deprivation-induced senescence in prostate cancer cells. Oncogene 2015; 34:5912-22. [PMID: 25772238 PMCID: PMC4573387 DOI: 10.1038/onc.2015.41] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 12/19/2014] [Accepted: 01/06/2015] [Indexed: 12/17/2022]
Abstract
The processes associated with transition to castration independent prostate cancer growth are not well understood. Cellular senescence is a stable cell cycle arrest that occurs in response to sublethal stress. It is often overcome in malignant transformation to confer a survival advantage. CCAAT/Enhancer Binding Protein (C/EBP) β function is frequently deregulated in human malignancies and interestingly, androgen dependent prostate cancer cells express primarily the LIP isoform. We found that C/EBPβ expression is negatively regulated by androgen receptor activity and that treatment of androgen dependent cell lines with anti-androgens increases C/EBPβ mRNA and protein levels. Accordingly, we also find that C/EBPβ levels are significantly elevated in primary prostate cancer samples from castration resistant compared with therapy naive patients. Chromatin immunoprecipitation demonstrated enhanced binding of the androgen receptor to the proximal promoter of the CEBPB gene in the presence of dihydroxytestosterone. Upon androgen deprivation, induction of C/EBPβ is facilitated by active transcription as evident by increased histone 3 acetylation at the C/EBPβ promoter. Also, the androgen agonist R1881 suppresses the activity of a CEBPB promoter reporter. Loss of C/EBPβ expression prevents growth arrest following androgen deprivation or anti-androgen challenge. Accordingly, suppression of C/EBPβ under low androgen conditions results in reduced expression of senescence-associated secretory genes, significantly decreased number of cells displaying heterochromatin foci, and increased numbers of Ki67 positive cells. Ectopic expression of C/EBPβ caused pronounced morphological changes, reduced PC cell growth, and increased the number of senescent LNCaP cells. Lastly, we found that senescence contributes to prostate cancer cell survival under androgen deprivation, and C/EBPβ deficient cells were significantly more susceptible to killing by cytotoxic chemotherapy following androgen deprivation. Our data demonstrate that up-regulation of C/EBPβ is critical for complete maintenance of androgen deprivation induced senescence and that targeting C/EBPβ expression may synergize with anti-androgen or chemotherapy in eradicating prostate cancer.
Collapse
Affiliation(s)
- D J Barakat
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - J Zhang
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - T Barberi
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - S R Denmeade
- Division of Medical Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - A D Friedman
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - I Paz-Priel
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
111
|
Regulation and function of Myb-binding protein 1A (MYBBP1A) in cellular senescence and pathogenesis of head and neck cancer. Cancer Lett 2014; 358:191-199. [PMID: 25543088 DOI: 10.1016/j.canlet.2014.12.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/16/2014] [Accepted: 12/17/2014] [Indexed: 11/20/2022]
Abstract
Myb-binding protein 1A (MYBBP1A) is a nucleolar protein implicated in stress response and carcinogenesis; however, its functional contribution to senescence remains elusive. In this study we show decreased MYBBP1A protein levels in tumor cells after treatment with etoposide, a potent inducer of DNA damage. Although silencing of MYBBP1A expression was not sufficient to induce senescence, it significantly increased the relative abundance of senescent cells after DNA damage. We found an inverse regulation of MYBBP1A and AKT phosphorylation (pAKT(Ser473)), which was characteristic for the pre-senescent state after etoposide administration in vitro. Tissue microarrays with tumor specimens from primary oropharyngeal squamous cell carcinoma (OPSCC) patients (n = 61) by immunohistochemistry revealed a significant correlation between MYBBP1A(low)pAKT(Ser473)(high) staining pattern and shorter progression-free (p = 0.007) or overall survival (p < 0.001). Multivariate analysis showed that MYBBP1A(low)pAKT(Ser473)(high) staining pattern is an independent prognosticator for OPSCC. Taken together, our study points to a critical role of MYBBP1A in the regulation of senescence under genotoxic stress and that a MYBBP1A(low)AKT(Ser473)(high) staining pattern serves not only as a marker for the pre-senescent stage but also as an indicator of OPSCC patients at high risk for treatment failure.
Collapse
|
112
|
Li L, Liu Y. Aging-related gene signature regulated by Nlrp3 predicts glioma progression. Am J Cancer Res 2014; 5:442-449. [PMID: 25628952 PMCID: PMC4300695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/15/2014] [Indexed: 06/04/2023] Open
Abstract
Aging is the strongest risk factor for glioma development, suggesting that molecular crosstalks between aging and tumorigenesis exist in many cellular pathways. Recently, Nlrp3 inflammasome have been shown to modulate several major cellular pathways such as inflammation and cell death and have been demonstrated to be an upstream target that controlled the process of brain aging. We proposed Nlrp3 inflammasome may serve as a possible molecular link between aging and glioma progression. In this study, we generated a aging-related gene signature that regulated by Nlrp3 in mouse hippocampus and demonstrated that this gene signature can distinguish subsets of glioma samples and predicts clinical outcome in radiotherapy-treated patients. In addition, using U87 and GL261 xenograft mouse glioblastoma model, we found that Nlrp3 inflammasome contributed to radiotherapy resistance in glioma. Ionizing radiation can induce Nlrp3 inflammasome expression; Nlrp3 inhibition reduced tumor growth and prolonged the survival of mouse following IR treatment; Nlrp3 inhibition reduced number of senescent cells induced by IR. These results above suggest that Nlrp3 inflammasome is an important molecular link between brain aging and glioma progression; the Nlrp3 gene signature may serve as a predictive biomarker for glioma patients.
Collapse
Affiliation(s)
- Lianling Li
- Department of Neurosurgery, Qilu Hospital of Shandong UniversityJinan 250012, China
- Brain Science Research Institute of Shandong UniversityJinan 250012, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong UniversityJinan 250012, China
| | - Yuguang Liu
- Department of Neurosurgery, Qilu Hospital of Shandong UniversityJinan 250012, China
- Brain Science Research Institute of Shandong UniversityJinan 250012, China
| |
Collapse
|
113
|
[Emergence of cancer stem cells or tumor-initiating/propagating cells and relapse in multiple myeloma]. Bull Cancer 2014; 101:1074-9. [PMID: 25467977 DOI: 10.1684/bdc.2014.2027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
114
|
Gaudichon J, Milano F, Cahu J, DaCosta L, Martens AC, Renoir JM, Sola B. Deazaneplanocin a is a promising drug to kill multiple myeloma cells in their niche. PLoS One 2014; 9:e107009. [PMID: 25255316 PMCID: PMC4177844 DOI: 10.1371/journal.pone.0107009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 08/05/2014] [Indexed: 02/07/2023] Open
Abstract
Tumoral plasma cells has retained stemness features and in particular, a polycomb-silenced gene expression signature. Therefore, epigenetic therapy could be a mean to fight for multiple myeloma (MM), still an incurable pathology. Deazaneplanocin A (DZNep), a S-adenosyl-L-homocysteine hydrolase inhibitor, targets enhancer of zest homolog 2 (EZH2), a component of polycomb repressive complex 2 (PRC2) and is capable to induce the death of cancer cells. We show here that, in some MM cell lines, DZNep induced both caspase-dependent and -independent apoptosis. However, the induction of cell death was not mediated through its effect on EZH2 and the trimethylation on lysine 27 of histone H3 (H3K27me3). DZNep likely acted through non-epigenetic mechanisms in myeloma cells. In vivo, in xenograft models, and in vitro DZNep showed potent antimyeloma activity alone or in combination with bortezomib. These preclinical data let us to envisage new therapeutic strategies for myeloma.
Collapse
Affiliation(s)
- Jérémie Gaudichon
- Equipe Associée 4652, Université de Caen, Normandie Univ, Caen, France
| | - Francesco Milano
- Equipe Associée 4652, Université de Caen, Normandie Univ, Caen, France
| | - Julie Cahu
- Equipe Associée 4652, Université de Caen, Normandie Univ, Caen, France
| | - Lætitia DaCosta
- Institut National de la Santé et de la Recherche Médicale U749, Institut Gustave Roussy, Villejuif, France
| | - Anton C. Martens
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jack-Michel Renoir
- Institut National de la Santé et de la Recherche Médicale U749, Institut Gustave Roussy, Villejuif, France
| | - Brigitte Sola
- Equipe Associée 4652, Université de Caen, Normandie Univ, Caen, France
- * E-mail:
| |
Collapse
|
115
|
T cells and their cytokines in persistent stimulation of the immune system. Curr Opin Immunol 2014; 29:79-85. [DOI: 10.1016/j.coi.2014.05.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/10/2014] [Accepted: 05/04/2014] [Indexed: 12/31/2022]
|
116
|
Pawelec G, Goldeck D, Derhovanessian E. Inflammation, ageing and chronic disease. Curr Opin Immunol 2014; 29:23-8. [DOI: 10.1016/j.coi.2014.03.007] [Citation(s) in RCA: 255] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/27/2014] [Accepted: 03/29/2014] [Indexed: 12/15/2022]
|
117
|
Abstract
Hayflick and Moorhead first described senescence in the late 1960's as a permanent growth arrest that primary cells underwent after a defined number of cellular divisions in culture. This observation gave rise to the hypothesis that cells contained an internal counting mechanism that limited cellular division and that this limit was an important barrier to cellular transformation. What began as an in vitro observation has led to an immense body of work that reaches into all fields of biology and is of particular interest in the areas of aging, tissue regeneration, and tumorigenesis. The initially simplistic view that senescence limits cellular division and contributes to aging while stymying tumorigenesis has now evolved into an important and complex biological process that has numerous caveats and often opposing effects on tumorigenesis. In this review, we limit our discussion to the complex role senescence plays in tumorigenesis. Throughout the review we attempt to draw many parallels to other systems including the role senescent cells play in the tumor microenvironment and their significant molecular and phenotypic similarities to cancer associated fibroblasts (CAFs).
Collapse
Affiliation(s)
- Elise Alspach
- Department of Cell Biology and Physiology; BRIGHT Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Yujie Fu
- Department of Cell Biology and Physiology, BRIGHT Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Sheila A Stewart
- Department of Cell Biology and Physiology, BRIGHT Institute, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
118
|
Affiliation(s)
- Julie Cahu
- MILPAT (EA 4652), Faculté de Médecine, CHU, Caen, France
| |
Collapse
|
119
|
Alspach E, Flanagan KC, Luo X, Ruhland MK, Huang H, Pazolli E, Donlin MJ, Marsh T, Piwnica-Worms D, Monahan J, Novack DV, McAllister SS, Stewart SA. p38MAPK plays a crucial role in stromal-mediated tumorigenesis. Cancer Discov 2014; 4:716-29. [PMID: 24670723 DOI: 10.1158/2159-8290.cd-13-0743] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Neoplastic cells rely on the tumor microenvironment (TME) for survival and progression factors. Indeed, senescent and cancer-associated fibroblasts (CAF) express factors that promote tumorigenesis that are collectively referred to as the senescence-associated secretory phenotype (SASP). Despite their importance in tumorigenesis, the mechanisms that control TME-derived factor expression remain poorly understood. Here, we address a key unanswered question: how the SASP is sustained in senescent fibroblasts and CAFs. We find that the mitogen-activated protein kinase p38 (p38MAPK) controls AUF1 occupancy on SASP mRNAs and thus controls their stability. The importance of this regulatory mechanism is underscored by our findings that stromal-specific p38MAPK inhibition abrogates the tumor-promoting activities of CAFs and senescent fibroblasts. Our data suggest that targeting SASP mRNA stability through inhibition of p38MAPK will significantly aid the development of clinical strategies to target the TME. SIGNIFICANCE The TME plays a key role in tumorigenesis. We demonstrate that p38MAPK governs a posttranscriptional mechanism that sustains the protumorigenic SASP. Inhibition of p38MAPK abrogates the tumor-promoting activities of CAFs and senescent fibroblasts. Thus, p38MAPK is a TME-specific Achilles' heel that may be exploited as a new therapeutic target.
Collapse
Affiliation(s)
- Elise Alspach
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kevin C Flanagan
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xianmin Luo
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Megan K Ruhland
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Huang
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ermira Pazolli
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maureen J Donlin
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy Marsh
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Piwnica-Worms
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph Monahan
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deborah V Novack
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra S McAllister
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sheila A Stewart
- Authors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TexasAuthors' Affiliations: Departments of Cell Biology and Physiology, Medicine, and Pathology and Immunology; BRIGHT Institute, Washington University School of Medicine; Departments of Biochemistry & Molecular Biology and Molecular Microbiology & Immunology, Saint Louis University School of Medicine; Confluence Life Sciences, Inc., St. Louis, Missouri; Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston; Harvard Stem Cell Institute and Broad Institute of Harvard and MIT, Cambridge, Massachusetts; and Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas sheila.stewart@wust
| |
Collapse
|
120
|
Abstract
The p53 tumor suppressor governs multiple cell-intrinsic programs, including cell-cycle arrest and apoptosis, to curb neoplastic growth. A new study reveals that p53 also acts through a novel non-cell-autonomous mechanism, by stimulating the innate immune system to maintain tissue homeostasis and suppress tumorigenesis.
Collapse
|
121
|
TOR-centric view on insulin resistance and diabetic complications: perspective for endocrinologists and gerontologists. Cell Death Dis 2013; 4:e964. [PMID: 24336084 PMCID: PMC3877573 DOI: 10.1038/cddis.2013.506] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 11/11/2013] [Accepted: 11/13/2013] [Indexed: 01/06/2023]
Abstract
This article is addressed to endocrinologists treating patients with diabetic complications as well as to basic scientists studying an elusive link between diseases and aging. It answers some challenging questions. What is the link between insulin resistance (IR), cellular aging and diseases? Why complications such as retinopathy may paradoxically precede the onset of type II diabetes. Why intensive insulin therapy may initially worsen retinopathy. How nutrient- and insulin-sensing mammalian target of rapamycin (mTOR) pathway can drive insulin resistance and diabetic complications. And how rapamycin, at rational doses and schedules, may prevent IR, retinopathy, nephropathy and beta-cell failure, without causing side effects.
Collapse
|
122
|
Abstract
Recent groundbreaking discoveries have revealed that IGF-1, Ras, MEK, AMPK, TSC1/2, FOXO, PI3K, mTOR, S6K, and NFκB are involved in the aging process. This is remarkable because the same signaling molecules, oncoproteins and tumor suppressors, are well-known targets for cancer therapy. Furthermore, anti-cancer drugs aimed at some of these targets have been already developed. This arsenal could be potentially employed for anti-aging interventions (given that similar signaling molecules are involved in both cancer and aging). In cancer, intrinsic and acquired resistance, tumor heterogeneity, adaptation, and genetic instability of cancer cells all hinder cancer-directed therapy. But for anti-aging applications, these hurdles are irrelevant. For example, since anti-aging interventions should be aimed at normal postmitotic cells, no selection for resistance is expected. At low doses, certain agents may decelerate aging and age-related diseases. Importantly, deceleration of aging can in turn postpone cancer, which is an age-related disease.
Collapse
|
123
|
Protumorigenic effects of mir-145 loss in malignant pleural mesothelioma. Oncogene 2013; 33:5319-31. [PMID: 24240684 DOI: 10.1038/onc.2013.476] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 12/11/2022]
Abstract
We identified a discrete number of microRNAs differentially expressed in benign or malignant mesothelial tissues. We focused on mir-145 whose levels were significantly downregulated in malignant mesothelial tissues and malignant pleural mesothelioma (MPM) cell lines as compared to benign tissues (pleura, peritoneum or cysts). We show that promoter hyper-methylation caused very low levels in MPM cell lines and specimens. Treatment of MPM cell lines with mir-145 agonists negatively modulated some protumorigenic properties of MPM cells, such as clonogenicity, cell migration and resistance to pemetrexed treatment. The main effector mechanism of the clonogenic death induced by mir-145 was that of accelerated senescence. We found that mir-145 targeted OCT4 via specific binding to its 3'-UTR. Increased intracellular levels of mir-145 decreased the levels of OCT4 and its target gene ZEB1, thereby counteracting the increase of OCT4 induced by pemetrexed treatment which is known to favor the development of chemoresistant cells. In line with this, reintroduction of OCT4 into mimic-145 treated cells counteracted the effects on clonogenicity and replicative senescence. This further supports the relevance of the mir-145-OCT4 interaction for the survival of MPM cells. The potential use of mir-145 expression levels to classify benign vs malignant mesothelial tissues and the differences between pemetrexed-induced senescence and that induced by the re-expression of mir-145 are discussed.
Collapse
|
124
|
Abstract
Cardiac senescence and age-related disease development have gained general attention and recognition in the past decades due to increased accessibility and quality of health care. The advancement in global civilization is complementary to concerns regarding population aging and development of chronic degenerative diseases. Cardiac degeneration has been rigorously studied. The molecular mechanisms of cardiac senescence are on multiple cellular levels and hold a multilayer complexity level, thereby hampering development of unambiguous treatment protocols. In particular, the synergistic exchange of the senescence phenotype through a senescence secretome between myocytes and stem cells appears complicated and is of great future therapeutic value. The current review article will highlight hallmarks of senescence, cardiac myocyte and stem cell senescence, and the mutual exchange of senescent secretome. Future cardiac cell therapy approaches require a comprehensive understanding of myocardial senescence to improve therapeutic efficiency as well as efficacy.
Collapse
|
125
|
Abstract
Human cells do not indefinitely proliferate. Upon external and/or intrinsic cues, cells might die or enter a stable cell cycle arrest called senescence. Several cellular mechanisms, such as telomere shortening and abnormal expression of mitogenic oncogenes, have been shown to cause senescence. Senescence is not restricted to normal cells; cancer cells have also been reported to senesce. Chemotherapeutical drugs have been shown to induce senescence in cancer cells. However, it remains controversial whether senescence prevents or promotes tumorigenesis. As it might eventually be patient-specific, a rapid and sensitive method to assess senescence in cancer cell will soon be required. To this end, the standard β-galactosidase assay, the currently used method, presents major drawbacks: it is time consuming and not sensitive. We propose here a flow cytometry-based assay to study senescence on live cells. This assay offers the advantage of being rapid, sensitive, and can be coupled to the immunolabeling of various cellular markers.
Collapse
Affiliation(s)
- Julie Cahu
- MILPAT (EA 4652), Université de Caen Basse-Normandie.
| | | |
Collapse
|
126
|
MEK drives cyclin D1 hyperelevation during geroconversion. Cell Death Differ 2013; 20:1241-9. [PMID: 23852369 DOI: 10.1038/cdd.2013.86] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 12/26/2022] Open
Abstract
When the cell cycle becomes arrested, MTOR (mechanistic Target of Rapamycin) converts reversible arrest into senescence (geroconversion). Hyperexpression of cyclin D1 is a universal marker of senescence along with hypertrophy, beta-Gal staining and loss of replicative/regenerative potential (RP), namely, the ability to restart proliferation when the cell cycle is released. Inhibition of MTOR decelerates geroconversion, although only partially decreases cyclin D1. Here we show that in p21- and p16-induced senescence, inhibitors of mitogen-activated/extracellular signal-regulated kinase (MEK) (U0126, PD184352 and siRNA) completely prevented cyclin D1 accumulation, making it undetectable. We also used MEL10 cells in which MEK inhibitors do not inhibit MTOR. In such cells, U0126 by itself induced senescence that was remarkably cyclin D1 negative. In contrast, inhibition of cyclin-dependent kinase (CDK) 4/6 by PD0332991 caused cyclin D1-positive senescence in MEL10 cells. Both types of senescence were suppressed by rapamycin, converting it into reversible arrest. We confirmed that the inhibitor of CDK4/6 caused cyclin D1 positive senescence in normal RPE cells, whereas U0126 prevented cyclin D1 expression. Elimination of cyclin D1 by siRNA did not prevent other markers of senescence that are consistent with the lack of its effect on MTOR. Our data confirmed that a mere inhibition of the cell cycle was sufficient to cause senescence, providing MTOR was active, and inhibition of MEK partially inhibited MTOR in a cell-type-dependent manner. Second, hallmarks of senescence may be dissociated, and hyperelevated cyclin D1, a marker of hyperactivation of senescent cells, did not necessarily determine other markers of senescence. Third, inhibition of MEK was sufficient to eliminate cyclin D1, regardless of MTOR.
Collapse
|
127
|
Role of inflammasomes and their regulators in prostate cancer initiation, progression and metastasis. Cell Mol Biol Lett 2013; 18:355-67. [PMID: 23793845 PMCID: PMC6275599 DOI: 10.2478/s11658-013-0095-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/13/2013] [Indexed: 11/22/2022] Open
Abstract
Prostate cancer is one of the main cancers that affect men, especially older men. Though there has been considerable progress in understanding the progression of prostate cancer, the drivers of its development need to be studied more comprehensively. The emergence of resistant forms has also increased the clinical challenges involved in the treatment of prostate cancer. Recent evidence has suggested that inflammation might play an important role at various stages of cancer development. This review focuses on inflammasome research that is relevant to prostate cancer and indicates future avenues of study into its effective prevention and treatment through inflammasome regulation. With regard to prostate cancer, such research is still in its early stages. Further study is certainly necessary to gain a broader understanding of prostate cancer development and to create successful therapy solutions.
Collapse
|