51
|
Hu L, Li H, Zi M, Li W, Liu J, Yang Y, Zhou D, Kong QP, Zhang Y, He Y. Why Senescent Cells Are Resistant to Apoptosis: An Insight for Senolytic Development. Front Cell Dev Biol 2022; 10:822816. [PMID: 35252191 PMCID: PMC8890612 DOI: 10.3389/fcell.2022.822816] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/26/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a process that leads to a state of irreversible cell growth arrest induced by a variety of intrinsic and extrinsic stresses. Senescent cells (SnCs) accumulate with age and have been implicated in various age-related diseases in part via expressing the senescence-associated secretory phenotype. Elimination of SnCs has the potential to delay aging, treat age-related diseases and extend healthspan. However, once cells becoming senescent, they are more resistant to apoptotic stimuli. Senolytics can selectively eliminate SnCs by targeting the SnC anti-apoptotic pathways (SCAPs). They have been developed as a novel pharmacological strategy to treat various age-related diseases. However, the heterogeneity of the SnCs indicates that SnCs depend on different proteins or pathways for their survival. Thus, a better understanding of the underlying mechanisms for apoptotic resistance of SnCs will provide new molecular targets for the development of cell-specific or broad-spectrum therapeutics to clear SnCs. In this review, we discussed the latest research progresses and challenge in senolytic development, described the significance of regulation of senescence and apoptosis in aging, and systematically summarized the SCAPs involved in the apoptotic resistance in SnCs.
Collapse
Affiliation(s)
- Li Hu
- Department of Geriatrics, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,College of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Huiqin Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Meiting Zi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wen Li
- Department of Endocrinology, The Third People's Hospital of Yunnan Province, Kunming, China
| | - Jing Liu
- Lab of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yang Yang
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yunxia Zhang
- Department of Geriatrics, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.,College of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Yonghan He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
52
|
The Correlation of HK2 Gene Expression with the Occurrence, Immune Cell Infiltration, and Prognosis of Renal Cell Carcinoma. DISEASE MARKERS 2022; 2022:1452861. [PMID: 35265223 PMCID: PMC8898847 DOI: 10.1155/2022/1452861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 11/29/2022]
Abstract
Objectives Hexokinase 2 (HK2) is one of the key factors involved in the development of several human cancers. However, its role in immune cell infiltration (ICI) and tumor development in renal cell carcinoma is not yet known. Thus, we aimed to explore its relationship with ICI, overall survival, and prognosis of renal cell carcinoma. Methods In this study, RNA-seq data from renal cancer and normal tissues were extracted from TCGA and the relationship between HK2 expression and pathological features of RCC patients was analyzed using the GEPIA and UALCAN databases. Subsequently, Western blot and qRT-PCR were performed to analyze the protein and mRNA expression of HK2 in renal cell carcinoma tissues and cell lines. Lastly, various bioinformatics tools were applied to determine the immune cell infiltration, survival, and developing prediction model. Results The analysis of RNA-seq data revealed a high expression of HK2 in renal cell carcinoma; furthermore, Western blot and qRT-PCR also showed high expression of HK2 in renal cancer tissues and cell lines. The high expression of HK2 showed a significant positive correlation with the advanced stage of the tumor, lymph node metastasis, and worst survival in renal carcinoma patients. The high expression of HK2 was further identified as an independent risk factor of RCC patients; it also showed a significant positive immune cell infiltration RCC tumor microenvironment including macrophages, B cells, neutrophils, dendritic cells, and CD8+ T cells. Conclusion the expression of HK2 is positively correlated with the immune cell infiltration and prognosis of renal cell carcinoma patients, thus playing an important role in renal cancer development.
Collapse
|
53
|
Gu Y, Xue F, Xiao H, Chen L, Zhang Y. Bamboo Leaf Flavonoids Suppress Oxidative Stress-Induced Senescence of HaCaT Cells and UVB-Induced Photoaging of Mice through p38 MAPK and Autophagy Signaling. Nutrients 2022; 14:nu14040793. [PMID: 35215447 PMCID: PMC8876272 DOI: 10.3390/nu14040793] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 01/27/2023] Open
Abstract
With the global escalation of the aging process, the research on aging mechanisms and anti-aging strategies has become a hot spot. As the most external organ of the human body, skin can be used as an ideal organ for the study of endogenous and exogenous aging. Bamboo leaf flavonoids (BLF) possess a variety of biological effects such as antioxidant, anti-bacterial, anti-inflammatory, lipid-lowering, anti-radiation, and anti-aging. However, it is still unclear whether they can delay skin aging. This study aimed to analyze the inhibitory effect of BLF on skin aging and explore their molecular mechanisms. We found that 10–40 μg/mL BLF significantly inhibited the senescence of HaCaT cells induced by AAPH, which might be related to their antioxidant and anti-inflammatory abilities. Further mechanism studies showed that mitogen-activated protein kinase (MAPK), especially the p38 MAPK pathway, was the key to BLF to alleviate the senescence of HaCaT cells. In addition, autophagy was also involved in the anti-senescence effect of BLF. The results were also verified in UVB-induced photoaging mice. Therefore, BLF can be used as a potential therapeutic agent to intervene skin aging in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | | | - Ying Zhang
- Correspondence: ; Tel.: +86-057-188-982-164
| |
Collapse
|
54
|
Yu D, Li X, Wang Z, Jiang S, Yan T, Fang K, Shi Y, Jiang Z, Zhang S. Role of AUF1 in modulating the proliferation, migration and senescence of skin cells. Exp Ther Med 2021; 23:45. [PMID: 34934424 PMCID: PMC8652399 DOI: 10.3892/etm.2021.10967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/11/2021] [Indexed: 11/05/2022] Open
Abstract
AU-rich element RNA-binding factor 1 (AUF1) is a classical RNA-binding protein. AUF1 influences the process of development, apoptosis and tumorigenesis by interacting with adenylate-uridylate rich element-bearing mRNAs. Human skin is the largest organ of the body and acts as a protective barrier against pathogens and injuries. The aim of the present study was to explore the function and potential molecular pathways of AUF1 in human skin cells. AUF1 was overexpressed in human keratinocyte HaCaT cells and human skin fibroblast WS1 cells using adenoviruses and silenced using lentiviruses. AUF1 overexpression facilitated cell proliferation, whereas AUF1 knockdown induced the opposite effect. AUF1 reduced apoptosis but did not affect cell cycle progression. Forced AUF1 expression promoted the migration of human skin cells, as demonstrated by a scratch wound healing assay. Cell senescence was alleviated in AUF1-overexpressing skin cells, while AUF1 knockdown increased cell senescence. WS1 cells with AUF1 overexpression and silencing were used for RNA-sequencing and Kyoto Encyclopedia of Genes and Genomes-based pathway analysis to identify AUF1-affected mRNAs. A total of 18 mRNAs (eight mRNAs with positive associations and 10 mRNAs with negative associations) revealed consistent associations with both AUF1 overexpression and silencing. Enriched pathways associated with AUF1 expression included 'MAPK', 'cell adhesion molecules', 'proteasome', 'cellular senescence' and 'TGF-β signaling', indicating a complex regulatory network. Overall, the results of the present study revealed that AUF1 is involved in the proliferation, migration and senescence of skin cells in vitro and may be a potential target for cosmetic and disease treatment of skin.
Collapse
Affiliation(s)
- Daojiang Yu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China
| | - Xiaoqian Li
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhenyu Wang
- School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Sheng Jiang
- Department of Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China
| | - Tao Yan
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Kai Fang
- Department of Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China
| | - Yuhong Shi
- Department of Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China
| | - Zhiqiang Jiang
- Department of Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China
| | - Shuyu Zhang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Surgery, Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China.,Department of Oncology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213100, P.R. China
| |
Collapse
|
55
|
Boyajian JL, Ghebretatios M, Schaly S, Islam P, Prakash S. Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence. Nutrients 2021; 13:nu13124550. [PMID: 34960102 PMCID: PMC8705837 DOI: 10.3390/nu13124550] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
The role of the microbiome in human aging is important: the microbiome directly impacts aging through the gastrointestinal system. However, the microbial impact on skin has yet to be fully understood. For example, cellular senescence is an intrinsic aging process that has been recently associated with microbial imbalance. With age, cells become senescent in response to stress wherein they undergo irreversible growth arrest while maintaining high metabolic activity. An accumulation of senescent cells has been linked to various aging and chronic pathologies due to an overexpression of the senescence-associated secretory phenotype (SASP) comprised of proinflammatory cytokines, chemokines, growth factors, proteases, lipids and extracellular matrix components. In particular, dermatological disorders may be promoted by senescence as the skin is a common site of accumulation. The gut microbiota influences cellular senescence and skin disruption through the gut-skin axis and secretion of microbial metabolites. Metabolomics can be used to identify and quantify metabolites involved in senescence. Moreover, novel anti-senescent therapeutics are warranted given the poor safety profiles of current pharmaceutical drugs. Probiotics and prebiotics may be effective alternatives, considering the relationship between the microbiome and healthy aging. However, further research on gut composition under a senescent status is needed to develop immunomodulatory therapies.
Collapse
|
56
|
Delfarah A, Hartel NG, Zheng D, Yang J, Graham NA. Identification of a Proteomic Signature of Senescence in Primary Human Mammary Epithelial Cells. J Proteome Res 2021; 20:5169-5179. [PMID: 34637314 DOI: 10.1021/acs.jproteome.1c00659] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Senescence is a permanent cell cycle arrest that occurs in response to cellular stress and promotes age-related disease. Because senescence differs greatly depending on cell type and senescence inducer, continued progress in the characterization of senescent cells is needed. Here, we analyzed primary human mammary epithelial cells (HMECs), a model system for aging and cancer, using mass spectrometry-based proteomics. By integrating data from replicative senescence, immortalization by telomerase reactivation, and quiescence, we identified a robust proteomic signature of HMEC senescence consisting of 34 upregulated and 10 downregulated proteins. This approach identified known senescence biomarkers including β-galactosidase (GLB1) as well as novel senescence biomarkers including catechol O-methyltransferase (COMT), synaptic vesicle membrane protein VAT-1 homolog (VAT1), and plastin-1/3 (PLS1/PLS3). Gene ontology enrichment analysis demonstrated that senescent HMECs upregulated lysosomal proteins and downregulated RNA metabolic processes. In addition, a classification model based on our proteomic signature successfully discriminated proliferating and senescent HMECs at the transcriptional level. Finally, we found that the HMEC senescence signature was positively and negatively correlated with proteomic alterations in HMEC aging and breast cancer, respectively. Taken together, our results demonstrate the power of proteomics to identify cell type-specific signatures of senescence and advance the understanding of senescence in HMECs.
Collapse
Affiliation(s)
- Alireza Delfarah
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - Nicolas G Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - DongQing Zheng
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - Jesse Yang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089, United States.,Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
57
|
Abstract
Obesity is a major risk factor for the development of comorbidities such as type 2 diabetes, neurodegenerative disorders, osteoarthritis, cancer, cardiovascular and renal diseases. The onset of obesity is linked to an increase of senescent cells within adipose tissue and other organs. Cellular senescence is a stress response that has been shown to be causally linked to aging and development of various age-related diseases such as obesity. The senescence-associated-secretory phenotype of senescent cells creates a chronic inflammatory milieu that leads to local and systemic dysfunction. The elimination of senescent cells using pharmacological approaches (i.e., senolytics) has been shown to delay, prevent, or alleviate obesity-related organ dysfunction.
Collapse
Affiliation(s)
- Selim Chaib
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
58
|
Posch C. Ageing research: rethinking primary prevention of skin cancer. J Eur Acad Dermatol Venereol 2021; 35:2216-2218. [PMID: 34647667 DOI: 10.1111/jdv.17660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022]
Affiliation(s)
- C Posch
- Department of Dermatology and Allergy, School of Medicine, German Cancer Consortium (DKTK), Technical University of Munich, Munich, Germany.,Faculty of Medicine, Sigmund Freud University Vienna, Vienna, Austria
| |
Collapse
|
59
|
Wiley CD, Campisi J. The metabolic roots of senescence: mechanisms and opportunities for intervention. Nat Metab 2021; 3:1290-1301. [PMID: 34663974 PMCID: PMC8889622 DOI: 10.1038/s42255-021-00483-8] [Citation(s) in RCA: 235] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022]
Abstract
Cellular senescence entails a permanent proliferative arrest, coupled to multiple phenotypic changes. Among these changes is the release of numerous biologically active molecules collectively known as the senescence-associated secretory phenotype, or SASP. A growing body of literature indicates that both senescence and the SASP are sensitive to cellular and organismal metabolic states, which in turn can drive phenotypes associated with metabolic dysfunction. Here, we review the current literature linking senescence and metabolism, with an eye toward findings at the cellular level, including both metabolic inducers of senescence and alterations in cellular metabolism associated with senescence. Additionally, we consider how interventions that target either metabolism or senescent cells might influence each other and mitigate some of the pro-aging effects of cellular senescence. We conclude that the most effective interventions will likely break a degenerative feedback cycle by which cellular senescence promotes metabolic diseases, which in turn promote senescence.
Collapse
Affiliation(s)
- Christopher D Wiley
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, CA, USA.
- Buck Institute for Research on Aging, Novato, CA, USA.
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
60
|
Saleh T, Carpenter VJ. Potential Use of Senolytics for Pharmacological Targeting of Precancerous Lesions. Mol Pharmacol 2021; 100:580-587. [PMID: 34544896 DOI: 10.1124/molpharm.121.000361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/10/2021] [Indexed: 01/10/2023] Open
Abstract
Senescence is a cell state that contributes to several homeostatic and pathologic processes. In addition to being induced in somatic cells in response to replicative exhaustion (replicative senescence) as part of organismal aging, senescence can also be triggered prematurely by oncogene hyperactivation or tumor suppressor dysfunction [oncogene-induced senescence (OIS)]. Consequently, senescent cells comprise a major component of precancerous lesions of skin, oral mucosa, nasopharynx, prostate, gut, and lung. Unfortunately, invasive (or minimally invasive) interventions are currently the only available approach employed to eradicate premalignant lesions that carry the potential for cancer progression. Senolytics are a newly emerging drug class capable of selectively eliminating senescent cells. Although senolytics have been successfully demonstrated to mitigate a myriad of aging-related pathologies and to cull senescent cancer cells, there is a paucity of evidence for the potential use of senolytics as a novel approach to eliminate oncogene-induced senescent cells. This Emerging Concepts commentary will 1) summarize evidence in established models of OIS including B-Raf-induced nevi, transgenic lung cancer, and pancreatic adenocarcinoma models, as well as evidence from clinical precancerous lesions; 2) suggest that OIS is targetable; and 3) propose the utilization of senolytic agents as a revolutionary means to interfere with the ability of senescent premalignant cells to progress to cancer in vitro and in vivo If proven to be effective, senolytics will represent an emerging tool to pharmacologically treat precancerous lesions. SIGNIFICANCE STATEMENT: The treatment of premalignant lesions is largely based on the utilization of invasive (or minimally invasive) measures. Oncogene-induced senescence (OIS) is one form of senescence that occurs in response to oncogene overexpression in somatic cells and is present in precancerous lesions. Although the contribution of OIS to disease progression is undetermined, recent evidence suggests that senescent cells are permissive for malignant transformation. Accordingly, the pharmacological targeting of oncogene-induced senescent cells could potentially provide a novel, less invasive, means for the treatment of premalignant disease.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan (T.S.); Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (V.J.C.)
| | - Valerie J Carpenter
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa, Jordan (T.S.); Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia (V.J.C.)
| |
Collapse
|
61
|
De Blander H, Morel AP, Senaratne AP, Ouzounova M, Puisieux A. Cellular Plasticity: A Route to Senescence Exit and Tumorigenesis. Cancers (Basel) 2021; 13:4561. [PMID: 34572787 PMCID: PMC8468602 DOI: 10.3390/cancers13184561] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 01/10/2023] Open
Abstract
Senescence is a dynamic, multistep program that results in permanent cell cycle arrest and is triggered by developmental or environmental, oncogenic or therapy-induced stress signals. Senescence is considered as a tumor suppressor mechanism that prevents the risk of neoplastic transformation by restricting the proliferation of damaged cells. Cells undergoing senescence sustain important morphological changes, chromatin remodeling and metabolic reprogramming, and secrete pro-inflammatory factors termed senescence-associated secretory phenotype (SASP). SASP activation is required for the clearance of senescent cells by innate immunity. Therefore, escape from senescence and the associated immune editing would be a prerequisite for tumor initiation and progression as well as therapeutic resistance. One of the possible mechanisms for overcoming senescence could be the acquisition of cellular plasticity resulting from the accumulation of genomic alterations and genetic and epigenetic reprogramming. The modified composition of the SASP produced by these reprogrammed cancer cells would create a permissive environment, allowing their immune evasion. Additionally, the SASP produced by cancer cells could enhance the cellular plasticity of neighboring cells, thus hindering their recognition by the immune system. Here, we propose a comprehensive review of the literature, highlighting the role of cellular plasticity in the pro-tumoral activity of senescence in normal cells and in the cancer context.
Collapse
Affiliation(s)
- Hadrien De Blander
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
| | - Anne-Pierre Morel
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
- Institut Curie “EMT and Cancer Cell Plasticity”, Consortium Centre Léon Bérard, 69008 Lyon, France
| | - Aruni P. Senaratne
- UMR3664—Nuclear Dynamics, Development, Biology, Cancer, Genetics and Epigenetics, Institut Curie, PSL Research University, 75005 Paris, France;
| | - Maria Ouzounova
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
- Institut Curie “EMT and Cancer Cell Plasticity”, Consortium Centre Léon Bérard, 69008 Lyon, France
- CNRS UMR3666, Inserm U1143, Cellular and Chemical Biology, Institut Curie, PSL Research University, 75005 Paris, France
| | - Alain Puisieux
- Equipe Labellisée Ligue Contre le Cancer “EMT and Cancer Cell Plasticity”, CNRS 5286, INSERM 1052, Centre Léon Bérard, Cancer Research Center of Lyon, Université Claude Bernard Lyon 1, 69008 Lyon, France; (A.-P.M.); (M.O.)
- LabEx DEVweCAN, Université de Lyon, 69008 Lyon, France
- Institut Curie “EMT and Cancer Cell Plasticity”, Consortium Centre Léon Bérard, 69008 Lyon, France
- CNRS UMR3666, Inserm U1143, Cellular and Chemical Biology, Institut Curie, PSL Research University, 75005 Paris, France
| |
Collapse
|
62
|
Karen-Ng LP, James EL, Stephen A, Bennett MH, Mycielska ME, Parkinson EK. The Extracellular Metabolome Stratifies Low and High Risk Potentially Premalignant Oral Keratinocytes and Identifies Citrate as a Potential Non-Invasive Marker of Tumour Progression. Cancers (Basel) 2021; 13:cancers13164212. [PMID: 34439366 PMCID: PMC8394991 DOI: 10.3390/cancers13164212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The early detection of oral cancer is a high priority, as improvements in this area could lead to greater cure rates and reduced disability due to extensive surgery. Oral cancer is very difficult to detect in over 70% of cases as it develops unseen until quite advanced, sometimes rapidly. Therefore, the development of markers in body fluids (liquid biopsies) indicative of cancerous changes have a high priority. We show here that small molecules called metabolites can distinguish between non-diseased oral cells and two types of cells found in oral cells on the road to cancer. Although our investigation is preliminary, some of the metabolites have already been detected in the saliva (split) of oral cancer patients, and could eventually help detect oral cancer development at an earlier stage. Abstract Premalignant oral lesions (PPOLs) which bypass senescence (IPPOL) have a much greater probability of progressing to malignancy, but pre-cancerous fields also contain mortal PPOL keratinocytes (MPPOL) that possess tumour-promoting properties. To identify metabolites that could potentially separate IPPOL, MPPOL and normal oral keratinocytes non-invasively in vivo, we conducted an unbiased screen of their conditioned medium. MPPOL keratinocytes showed elevated levels of branch-chain amino acid, lipid, prostaglandin, and glutathione metabolites, some of which could potentially be converted into volatile compounds by oral bacteria and detected in breath analysis. Extracellular metabolites were generally depleted in IPPOL, and only six were elevated, but some metabolites distinguishing IPPOL from MPPOL have been associated with progression to oral squamous cell carcinoma (OSCC) in vivo. One of the metabolites elevated in IPPOL relative to the other groups, citrate, was confirmed by targeted metabolomics and, interestingly, has been implicated in cancer growth and metastasis. Although our investigation is preliminary, some of the metabolites described here are detectable in the saliva of oral cancer patients, albeit at a more advanced stage, and could eventually help detect oral cancer development earlier.
Collapse
Affiliation(s)
- Lee Peng Karen-Ng
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
- Oral Cancer Research & Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Emma Louise James
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
| | - Abish Stephen
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
| | - Mark Henry Bennett
- Department of Life Science, South Kensington Campus, Imperial College London, London SW7 2AZ, UK;
| | - Maria Elzbieta Mycielska
- Department of Surgery, University Medical Center, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany;
| | - Eric Kenneth Parkinson
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK; (L.P.K.-N.); (E.L.J.); (A.S.)
- Correspondence: ; Tel.: +44-(0)207-882-7185 or +44-(0)78546536
| |
Collapse
|
63
|
Pulido T, Velarde MC, Alimirah F. The senescence-associated secretory phenotype: Fueling a wound that never heals. Mech Ageing Dev 2021; 199:111561. [PMID: 34411604 DOI: 10.1016/j.mad.2021.111561] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022]
Abstract
Wound healing is impaired with advanced age and certain chronic conditions, such as diabetes and obesity. Moreover, common cancer treatments, including chemotherapy and radiation, can cause unintended tissue damage and impair wound healing. Available wound care treatments are not always effective, as some wounds fail to heal or recur after treatment. Hence, a more thorough understanding of the pathophysiology of chronic, nonhealing wounds may offer new ideas for the development of effective wound care treatments. Cancers are sometimes referred to as wounds that never heal, sharing mechanisms similar to wound healing. We describe in this review how cellular senescence and the senescence-associated secretory phenotype (SASP) contribute to chronic wounds versus cancer.
Collapse
Affiliation(s)
- Tanya Pulido
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Michael C Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, 1101, Philippines.
| | | |
Collapse
|
64
|
Yang J, Liu M, Hong D, Zeng M, Zhang X. The Paradoxical Role of Cellular Senescence in Cancer. Front Cell Dev Biol 2021; 9:722205. [PMID: 34458273 PMCID: PMC8388842 DOI: 10.3389/fcell.2021.722205] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence occurs in proliferating cells as a consequence of various triggers including telomere shortening, DNA damage, and inappropriate expression of oncogenes. The senescent state is accompanied by failure to reenter the cell cycle under mitotic stimulation, resistance to cell death and enhanced secretory phenotype. A growing number of studies have convincingly demonstrated a paradoxical role for spontaneous senescence and therapy-induced senescence (TIS), that senescence may involve both cancer prevention and cancer aggressiveness. Cellular senescence was initially described as a physiological suppressor mechanism of tumor cells, because cancer development requires cell proliferation. However, there is growing evidence that senescent cells may contribute to oncogenesis, partly in a senescence-associated secretory phenotype (SASP)-dependent manner. On the one hand, SASP prevents cell division and promotes immune clearance of damaged cells, thereby avoiding tumor development. On the other hand, SASP contributes to tumor progression and relapse through creating an immunosuppressive environment. In this review, we performed a review to summarize both bright and dark sides of senescence in cancer, and the strategies to handle senescence in cancer therapy were also discussed.
Collapse
Affiliation(s)
- Jing Yang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dongchun Hong
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
65
|
Leon KE, Buj R, Lesko E, Dahl ES, Chen CW, Tangudu NK, Imamura-Kawasawa Y, Kossenkov AV, Hobbs RP, Aird KM. DOT1L modulates the senescence-associated secretory phenotype through epigenetic regulation of IL1A. J Cell Biol 2021; 220:e202008101. [PMID: 34037658 PMCID: PMC8160577 DOI: 10.1083/jcb.202008101] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 04/06/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Oncogene-induced senescence (OIS) is a stable cell cycle arrest that occurs in normal cells upon oncogene activation. Cells undergoing OIS express a wide variety of secreted factors that affect the senescent microenvironment termed the senescence-associated secretory phenotype (SASP), which is beneficial or detrimental in a context-dependent manner. OIS cells are also characterized by marked epigenetic changes. We globally assessed histone modifications of OIS cells and discovered an increase in the active histone marks H3K79me2/3. The H3K79 methyltransferase disruptor of telomeric silencing 1-like (DOT1L) was necessary and sufficient for increased H3K79me2/3 occupancy at the IL1A gene locus, but not other SASP genes, and was downstream of STING. Modulating DOT1L expression did not affect the cell cycle arrest. Together, our studies establish DOT1L as an epigenetic regulator of the SASP, whose expression is uncoupled from the senescence-associated cell cycle arrest, providing a potential strategy to inhibit the negative side effects of senescence while maintaining the beneficial inhibition of proliferation.
Collapse
Affiliation(s)
- Kelly E. Leon
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Biomedical Sciences Graduate Program, Penn State College of Medicine, Hershey, PA
| | - Raquel Buj
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Elizabeth Lesko
- Department of Dermatology, Penn State College of Medicine, Hershey, PA
| | - Erika S. Dahl
- Biomedical Sciences Graduate Program, Penn State College of Medicine, Hershey, PA
| | - Chi-Wei Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Naveen Kumar Tangudu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | - Ryan P. Hobbs
- Department of Dermatology, Penn State College of Medicine, Hershey, PA
| | - Katherine M. Aird
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
66
|
Persaud Y, Seddiq M, Wu YM, Robinson DR, Poulik J, Altinok D, Roarty JD, Mody R, Taub JW. Unusual clinical behavior of a very late retinoblastoma relapse in a patient with a germline RB mutation. Pediatr Blood Cancer 2021; 68:e29064. [PMID: 34003562 DOI: 10.1002/pbc.29064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 11/06/2022]
Affiliation(s)
- Yogindra Persaud
- Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Marjilla Seddiq
- Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Yi-Mi Wu
- Department of Pathology and Clinical Laboratories, University of Michigan, Ann Arbor, Michigan, USA
| | - Dan R Robinson
- Department of Pathology and Clinical Laboratories, University of Michigan, Ann Arbor, Michigan, USA
| | - Janet Poulik
- Division of Pathology, Children's Hospital of Michigan, Detroit, Michigan, USA.,Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Deniz Altinok
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Radiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - John D Roarty
- Division of Ophthalmology, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Rajen Mody
- Department of Pediatric Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey W Taub
- Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA.,Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Pediatrics, Central Michigan University, Mt. Pleasant, Michigan, USA
| |
Collapse
|
67
|
Tan Q, Liang N, Zhang X, Li J. Dynamic Aging: Channeled Through Microenvironment. Front Physiol 2021; 12:702276. [PMID: 34366891 PMCID: PMC8334186 DOI: 10.3389/fphys.2021.702276] [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] [Received: 04/29/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
Aging process is a complicated process that involves deteriorated performance at multiple levels from cellular dysfunction to organ degeneration. For many years research has been focused on how aging changes things within cell. However, new findings suggest that microenvironments, circulating factors or inter-tissue communications could also play important roles in the dynamic progression of aging. These out-of-cell mechanisms pass on the signals from the damaged aging cells to other healthy cells or tissues to promote systematic aging phenotypes. This review discusses the mechanisms of how senescence and their secretome, NAD+ metabolism or circulating factors change microenvironments to regulate systematic aging, as well as the potential therapeutic strategies based on these findings for anti-aging interventions.
Collapse
Affiliation(s)
- Qing Tan
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Liang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqian Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
68
|
Prime SS, Cirillo N, Cheong SC, Prime MS, Parkinson EK. Targeting the genetic landscape of oral potentially malignant disorders has the potential as a preventative strategy in oral cancer. Cancer Lett 2021; 518:102-114. [PMID: 34139286 DOI: 10.1016/j.canlet.2021.05.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022]
Abstract
This study reviews the molecular landscape of oral potentially malignant disorders (OPMD). We examine the impact of tumour heterogeneity, the spectrum of driver mutations (TP53, CDKN2A, TERT, NOTCH1, AJUBA, PIK3CA, CASP8) and gene transcription on tumour progression. We comment on how some of these mutations impact cellular senescence, field cancerization and cancer stem cells. We propose that OPMD can be monitored more closely and more dynamically through the use of liquid biopsies using an appropriate biomarker of transformation. We describe new gene interactions through the use of a systems biology approach and we highlight some of the first studies to identify functional genes using CRISPR-Cas9 technology. We believe that this information has translational implications for the use of re-purposed existing drugs and/or new drug development. Further, we argue that the use of digital technology encompassing clinical and laboratory-based data will create relevant datasets for machine learning/artificial intelligence. We believe that therapeutic intervention at an early molecular premalignant stage should be an important preventative strategy to inhibit the development of oral squamous cell carcinoma and that this approach is applicable to other aerodigestive tract cancers.
Collapse
Affiliation(s)
- S S Prime
- Centre for Immunology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4NS, UK.
| | - N Cirillo
- Melbourne Dental School, University of Melbourne, 720 Swanson Street, Carlton, Melbourne, Victoria, 3053, Australia.
| | - S C Cheong
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 1 Jalan SS12/1A, Subang Jaya, Selangor, Malaysia.
| | - M S Prime
- Roche Diagnostics Information Solutions, Hoffman-La Roche Ltd., Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - E K Parkinson
- Centre for Immunology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4NS, UK.
| |
Collapse
|
69
|
Gonzalez-Meljem JM, Martinez-Barbera JP. Adamantinomatous craniopharyngioma as a model to understand paracrine and senescence-induced tumourigenesis. Cell Mol Life Sci 2021; 78:4521-4544. [PMID: 34019103 PMCID: PMC8195904 DOI: 10.1007/s00018-021-03798-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/03/2020] [Accepted: 01/15/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a process that can prevent tumour development in a cell autonomous manner by imposing a stable cell cycle arrest after oncogene activation. Paradoxically, senescence can also promote tumour growth cell non-autonomously by creating a permissive tumour microenvironment that fuels tumour initiation, progression to malignancy and metastasis. In a pituitary tumour known as adamantinomatous craniopharyngioma (ACP), cells that carry oncogenic β-catenin mutations and overactivate the WNT signalling pathway form cell clusters that become senescent and activate a senescence-associated secretory phenotype (SASP). Research in mouse models of ACP has provided insights into the function of the senescent cell clusters and revealed a critical role for SASP-mediated activities in paracrine tumour initiation. In this review, we first discuss this research on ACP and subsequently explore the theme of paracrine tumourigenesis in other tumour models available in the literature. Evidence is accumulating supporting the notion that paracrine signalling brought about by senescent cells may underlie tumourigenesis across different tumours and cancer models.
Collapse
Affiliation(s)
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Research and Teaching Programme, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
70
|
Senolytic targets and new strategies for clearing senescent cells. Mech Ageing Dev 2021; 195:111468. [PMID: 33741395 DOI: 10.1016/j.mad.2021.111468] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 01/10/2023]
Abstract
Senescent cells (SCs) accumulate with age and cause various age-related diseases. Clearance of SCs by transgenic or pharmaceutical strategies has been demonstrated to delay aging, treat age-related diseases and extend healthspan. SCs are resistant to various stressors because they are protected from apoptosis by SC anti-apoptotic pathways (SCAPs). Targeting the proteins in the SCAPs with small molecules can selectively kill SCs, the effector proteins are called senolytic targets and the small molecules are called senolytics. Until now, a series of senolytic targets, such as BCL-XL, heat shock protein 90 (HSP90), Na+/K+ ATPase, bromodomain containing 4 (BRD4), and oxidation resistance 1 (OXR1) have been identified. However, current senolytics targeting these proteins still have some limitations in killing SCs in terms of safety, specificity and broad-spectrum activity. To overcome the challenges, some new strategies, such as proteolysis-targeting chimera (PROTAC) technology, chimeric antigen receptor (CAR) T cells, and β-galactosidase-modified prodrugs, were developed to clear SCs and shown to have promising therapeutic potential. Here we review the significance of SCs in aging and age-related diseases, summarize the known senolytic targets and highlight the emerging new strategies for clearing SCs.
Collapse
|
71
|
Young ARJ, Cassidy LD, Narita M. Autophagy and senescence, converging roles in pathophysiology as seen through mouse models. Adv Cancer Res 2021; 150:113-145. [PMID: 33858595 DOI: 10.1016/bs.acr.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Both senescence and autophagy have been strongly linked to aging and also cancer development. Numerous molecular, cellular, and physiological changes are known to correlate with an increasing age, yet our understanding of what underlies these changes or how they combine to give rise to the various pathologies associated with aging is still unclear. Levels of autophagy activity are known to decrease with advancing age, in a variety of organisms including mammals. Whereas senescent cells are known to accumulate in our bodies with age. Herein we review evidence from some elegant genetic mouse models linking senescence and also autophagy to aging and cancer. It is especially interesting to note the convergence in the pathological phenotypes of these two processes, senescence and autophagy, in these mouse models.
Collapse
Affiliation(s)
- Andrew R J Young
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom.
| | - Liam D Cassidy
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Masashi Narita
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, United Kingdom.
| |
Collapse
|
72
|
Yousefzadeh M, Henpita C, Vyas R, Soto-Palma C, Robbins P, Niedernhofer L. DNA damage-how and why we age? eLife 2021; 10:62852. [PMID: 33512317 PMCID: PMC7846274 DOI: 10.7554/elife.62852] [Citation(s) in RCA: 196] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/15/2021] [Indexed: 12/16/2022] Open
Abstract
Aging is a complex process that results in loss of the ability to reattain homeostasis following stress, leading, thereby, to increased risk of morbidity and mortality. Many factors contribute to aging, such as the time-dependent accumulation of macromolecular damage, including DNA damage. The integrity of the nuclear genome is essential for cellular, tissue, and organismal health. DNA damage is a constant threat because nucleic acids are chemically unstable under physiological conditions and vulnerable to attack by endogenous and environmental factors. To combat this, all organisms possess highly conserved mechanisms to detect and repair DNA damage. Persistent DNA damage (genotoxic stress) triggers signaling cascades that drive cells into apoptosis or senescence to avoid replicating a damaged genome. The drawback is that these cancer avoidance mechanisms promote aging. Here, we review evidence that DNA damage plays a causal role in aging. We also provide evidence that genotoxic stress is linked to other cellular processes implicated as drivers of aging, including mitochondrial and metabolic dysfunction, altered proteostasis and inflammation. These links between damage to the genetic code and other pillars of aging support the notion that DNA damage could be the root of aging.
Collapse
Affiliation(s)
- Matt Yousefzadeh
- Institute on the Biology of Aging and Metabolism Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, United States
| | - Chathurika Henpita
- Institute on the Biology of Aging and Metabolism Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, United States
| | - Rajesh Vyas
- Institute on the Biology of Aging and Metabolism Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, United States
| | - Carolina Soto-Palma
- Institute on the Biology of Aging and Metabolism Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, United States
| | - Paul Robbins
- Institute on the Biology of Aging and Metabolism Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, United States
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, United States
| |
Collapse
|
73
|
Mongiardi MP, Pellegrini M, Pallini R, Levi A, Falchetti ML. Cancer Response to Therapy-Induced Senescence: A Matter of Dose and Timing. Cancers (Basel) 2021; 13:484. [PMID: 33513872 PMCID: PMC7865402 DOI: 10.3390/cancers13030484] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Accepted: 01/23/2021] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence participates to fundamental processes like tissue remodeling in embryo development, wound healing and inhibition of preneoplastic cell growth. Most senescent cells display common hallmarks, among which the most characteristic is a permanent (or long lasting) arrest of cell division. However, upon senescence, different cell types acquire distinct phenotypes, which also depend on the specific inducing stimuli. Senescent cells are metabolically active and secrete a collection of growth factors, cytokines, proteases, and matrix-remodeling proteins collectively defined as senescence-associated secretory phenotype, SASP. Through SASP, senescent cells modify their microenvironment and engage in a dynamic dialog with neighbor cells. Senescence of neoplastic cells, at least temporarily, reduces tumor expansion, but SASP of senescent cancer cells as well as SASP of senescent stromal cells in the tumor microenvironment may promote the growth of more aggressive cancer subclones. Here, we will review recent data on the mechanisms and the consequences of cancer-therapy induced senescence, enlightening the potentiality and the risk of senescence inducing treatments.
Collapse
Affiliation(s)
- Maria Patrizia Mongiardi
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| | - Manuela Pellegrini
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| | - Roberto Pallini
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy;
| | - Andrea Levi
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| | - Maria Laura Falchetti
- CNR-Institute of Biochemistry and Cell Biology, Campus Adriano Buzzati Traverso, Via Ercole Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy; (M.P.M.); (M.P.); (A.L.)
| |
Collapse
|
74
|
Zhang M, Wang Y, Jiang L, Song X, Zheng A, Gao H, Wei M, Zhao L. LncRNA CBR3-AS1 regulates of breast cancer drug sensitivity as a competing endogenous RNA through the JNK1/MEK4-mediated MAPK signal pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:41. [PMID: 33494806 PMCID: PMC7830819 DOI: 10.1186/s13046-021-01844-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022]
Abstract
Background Adriamycin (ADR) resistance is one of the main obstacles to improving the clinical prognosis of breast cancer patients. Long noncoding RNAs (lncRNAs) can regulate cell behavior, but the role of these RNAs in the anti-ADR activity of breast cancer remains unclear. Here, we aim to investigate the imbalance of a particular long noncoding RNA, lncRNA CBR3 antisense RNA 1 (CBR3-AS1), and its role in ADR resistance. Methods Microarray analysis of ADR-resistant breast cancer cells was performed to identify CBR3-AS1. CCK-8 and colony formation assays were used to detect the sensitivity of breast cancer cells to ADR. Dual-luciferase reporter, RNA pulldown, IHC and western blot analyses were used to verify the relationship between the expression of CBR3-AS1, miRNA and target genes. For in vivo experiments, the effect of CBR3-AS1 on breast cancer resistance was observed in a xenograft tumor model. The role of CBR3-AS1 in influencing ADR sensitivity was verified by clinical breast cancer specimens from the TCGA, CCLE, and GDSC databases. Results We found that CBR3-AS1 expression was significantly increased in breast cancer tissues and was closely correlated with poor prognosis. CBR3-AS1 overexpression promoted ADR resistance in breast cancer cells in vitro and in vivo. Mechanistically, we identified that CBR3-AS1 functioned as a competitive endogenous RNA by sponging miR-25-3p. MEK4 and JNK1 of the MAPK pathway were determined to be direct downstream proteins of the CBR3-AS1/miR-25-3p axis in breast cancer cells. Conclusions In summary, our findings demonstrate that CBR3-AS1 plays a critical role in the chemotherapy resistance of breast cancer by mediating the miR-25-3p and MEK4/JNK1 regulatory axes. The potential of CBR3-AS1 as a targetable oncogene and therapeutic biomarker of breast cancer was identified. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01844-7.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Ang Zheng
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Hua Gao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China.
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|